ArticlePDF Available

A randomized, double-blind, placebo-controlled, parallel-group study to evaluate the efficacy and safety of osmotic-controlled release oral delivery system methylphenidate HCl in adults with attention-deficit/hyperactivity disorder in Japan

Authors:

Abstract and Figures

Objectives. To evaluate the safety and efficacy of osmotic-controlled release oral delivery system (OROS) methylphenidate (MPH) HCl in adults with attention-deficit/hyperactivity disorder (ADHD). Methods. In this study, 284 adults with ADHD were randomized to OROS MPH or placebo. During the 4-week titration period, patients were titrated from a starting dose of 18 mg once daily to an individually-optimized dose of up to 72 mg once daily in weekly 18-mg increments. Patients continued on their individualized dose during the 4-week efficacy assessment period. The primary efficacy endpoint was change in DSM-IV Total ADHD Symptoms subscale score of Conners' Adult ADHD Rating Scale-Observer: Screening Version (CAARS-O:SV) from baseline to endpoint. Results. The mean change in DSM-IV Total ADHD Symptoms subscale score of CAARS-O:SV was significantly larger with OROS MPH compared with placebo (P < 0.0001, ANCOVA). Similar results were observed for the majority of secondary endpoints, including CAARS-O:SV total score and other subscale scores. Although treatment-emergent adverse events were reported more frequently in the OROS MPH group (81.8%) versus the placebo group (53.9%), OROS-MPH showed a well-tolerated safety profile overall. Conclusions. OROS MPH in a dose range of 18-72 mg once daily was effective and well-tolerated in adult patients with ADHD.
Content may be subject to copyright.
ORIGINAL ARTICLE
A randomized, double-blind, placebo-controlled, parallel-group
study to evaluate the effi cacy and safety of osmotic-controlled
release oral delivery system methylphenidate HCl in adults
with attention-defi cit/hyperactivity disorder in Japan
NAGAHIDE TAKAHASHI
1 , TADAISHI KOH
2 , YUSHIN TOMINAGA
3 , YUKI SAITO
4 ,
YUJI KASHIMOTO
5 & TAKA MATSUMURA
6
1
Clinical Responsible Physician Department, Clinical Science Division, Janssen Pharmaceutical K.K, Tokyo, Japan,
2
Clinical Science Initiative Department, Clinical Science Division, Phar maceutical K.K, Tokyo, Japan,
3
Project Lead
Department, Project Development Division, Pharmaceutical K.K, Tokyo, Japan,
4
Biostatistics Department, Quantitavie
Science Division, Pharmaceutical K.K, Tokyo, Japan,
5
Tr ial Management Department, Japan Clinical Operations
Division, Pharmaceutical K.K, Tokyo, Japan, and
6
Clinical Science Division, Pharmaceutical K.K, Tokyo, Japan
Abstract
Objectives. To evaluate the safety and effi cacy of osmotic-controlled release oral delivery system (OROS) methylphenidate
(MPH) HCl in adults with attention-defi cit/hyperactivity disorder (ADHD). Methods. In this study, 284 adults with ADHD
were randomized to OROS MPH or placebo. During the 4-week titration period, patients were titrated from a starting
dose of 18 mg once daily to an individually-optimized dose of up to 72 mg once daily in weekly 18-mg increments. Patients
continued on their individualized dose during the 4-week effi cacy assessment period. The primary effi cacy endpoint was
change in DSM-IV Total ADHD Symptoms subscale score of Conners Adult ADHD Rating Scale-Observer: Screening
Version (CAARS-O:SV) from baseline to endpoint. Results. The mean change in DSM-IV Total ADHD Symptoms subscale
score of CAARS-O:SV was signifi cantly larger with OROS MPH compared with placebo ( P 0.0001, ANCOVA). Similar
results were observed for the majority of secondary endpoints, including CAARS-O:SV total score and other subscale
scores. Although treatment-emergent adverse events were reported more frequently in the OROS MPH group (81.8%)
versus the placebo group (53.9%), OROS-MPH showed a well-tolerated safety profi le overall. Conclusions. OROS MPH
in a dose range of 18 72 mg once daily was effective and well-tolerated in adult patients with ADHD.
Key words: attention defi cit disorder with hyperactivity , methylphenidate , randomized controlled trial , adult , Japan
Introduction
Attention-defi cit/hyperactivity disorder (ADHD) is
a psychiatric disorder characterized by hyperactive,
impulsive and inattentive behaviour . Although the
onset of ADHD symptoms typically occurs before
the age of 7 according to the Diagnostic And Statis-
tical Manual of Mental Disorders 4th Edition
(DSM-IV) criteria, it is now considered to be a
chronic condition that continues into adulthood in
up to 80% of patients (Takahashi et al. 2011a).
Adults with ADHD experience impairment of exec-
utive functioning, which interferes with their ability
to manage their daily lives, including meeting dead-
lines, staying focused on their work and organizing
their schedule (Baron et al. 2011). The consequences
may include educational underachievement, loss
of employment and diffi culty maintaining interper-
sonal relationships (Biederman et al. 2007; Baron
et al. 2011).
Stimulants, including methylphenidate (MPH)
HCl, are currently the preferred medication for
the fi rst-line treatment of adults with ADHD in
many countries around the world (Baron et al. 2011;
Wilens et al. 2011). Extended-release stimulants are
recommended over their immediate-release coun-
terparts in adult patients because the convenience
of once-daily dosing encourages treatment compli-
ance and the potential for abuse is decreased with
Correspondence: Nagahide Takahashi, MD, PhD, Clinical Responsible Physician Department, Clinical Science Division, Janssen
Pharmaceutical K.K, 5-2 Nishi-kana 3-chome, Chiyoda-ku, Tokyo 101-0065, Japan. Tel: 81 3 4411 5509. Fax: 81 3 4411 5086.
E-mail: ntakaha4@its.jnj.com
(Recei ved 10 May 2013; accep ted 18 November 2013)
The World Journal of Biological Psychiatry, 2014; Early Online: 1–11
ISSN 1562 -2975 print /ISSN 1814-1412 online © 2014 Infor ma Healt hcare
DOI : 10.3109/ 15622 975.2 013.8 68925
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
2 N. Takahashi et al.
were also required to have a DSM-IV Total ADHD
Symptoms subscale score of 24 at baseline on the
investigator-rated Conners Adult ADHD Rating
Scale-Observer: Screening Version (CAARS-O:SV)
(Conners et al. 1999).
Patients were excluded from the study if they were
a non-responder to MPH and/or had a history of
hypersensitivity or intolerance to MPH or had been
treated with MPH or any other medications for
ADHD within 4 weeks before the screening visit.
Investigators determined non-responders to MPH
using all available information, including medical
chart records and information from parents or guard-
ians of patients seeking enrolment in this trial. Other
exclusion criteria included diagnosis of bipolar I dis-
order, schizophrenia, schizoaffective disorder, severe
obsessive compulsive disorder, pervasive developmen-
tal disorder (e.g., autistic disorder or Asperger s
disorder) or suicidality. Patients with confi rmed cancer
or other serious illnesses (e.g., hepatic or renal insuf-
ciency or signifi cant cardiac, gastrointestinal, psychi-
atric, or metabolic disturbances) were also excluded.
Study design
This was a randomized, double-blind, placebo-
controlled, parallel-group phase 3 clinical trial con-
ducted at 39 sites in Japan between 22 February
2011 and 19 April 2012 and consisting of three parts:
a screening period, a double-blind treatment period
and a post-study period. A schematic diagram of the
study design is shown in Figure 1. Initially, patients
underwent a screening period, which consisted of a
single visit 1 2 weeks before study agent administra-
tion. During this visit, patients gave their informed
consent and were screened for eligibility. The
screening period also allowed for washout of prohib-
ited medications in order to appropriately assess the
safety and effi cacy of OROS MPH. Up to 4 weeks
before study drug administration were allowed for
washout of medi cations used for the treatment of
ADHD (e.g., MPH and atomoxetine HCl).
The second part of the study was an 8-week dou-
ble-blind phase, which consisted of a 4-week titration
period followed by a 4-week effi cacy assessment
period. Eligible patients were randomized 1:1 to
receive once-daily OROS MPH or placebo in the
morning using a computer-generated randomization
schedule. During the titration period, patients
received a starting dose of OROS MPH 18 mg/day
or placebo for the fi rst week. Each of the following
weeks, the dose was incremented by 18 mg up to a
maximum dose of 72 mg/day until an individually
optimized dose was achieved. Titration continued
until one of the following occurred: either the patient s
DSM-IV Total ADHD Symptoms score of the
extended-release formulations (Biederman et al.
2007; Buitelaar et al. 2009; Baron et al. 2011).
Although the non-stimulant atomoxetine was rec-
ently approved for the treatment of adults with
ADHD in Japan, there is currently no stimulant
approved for the treatment of this patient popula-
tion (Takahashi et al. 2011a; Pharmaceuticals and
Medical Devices Agency 2012). Because of the
growing consensus supporting the use of stimulants
to treat adults with ADHD, the lack of an approved
stimulant for adult ADHD in Japan represents an
unmet medical need.
Osmotic-controlled release oral delivery system
(OROS) MPH HCl extended-release tablets (Con-
certa 2007) are a long-acting preparation of MPH
designed to take effect within 1.5 h and maintain
effectiveness for 12 h after administration (Johnson
& Johnson Pharmaceutical Research & Development
2011). OROS MPH has been shown to be a safe and
effective treatment for ADHD in children and adults
in studies conducted in many countries and areas
around the world (Pelham et al. 2001; Medori et al.
2008). In October 2007, OROS MPH became the
rst drug to receive approval in Japan for the
treatment of ADHD; however, its use is currently
restricted to adolescents and children under 18 years
of age and continuous treatment in adults who began
taking OROS MPH before the age of 18 (Takeda
2009). This is a concern because many patients with
ADHD are not diagnosed until adulthood, often
presenting for treatment due to the presence of
comorbid psychiatric conditions, such as anxiety or
depression (Baron et al. 2011). Therefore, there is an
urgent need to evaluate stimulant medications, such
as OROS MPH, in adult patients with ADHD in
Japan so that more treatment options may become
available for this patient population.
This study evaluated the safety and effi cacy of
OROS MPH titrated to a daily dose of 18 to 72 mg
in adults with ADHD.
Methods
Patients
Eligible patients were men and women between 18
and 64 years of age, who met the DSM-IV Text Revi-
sion (DSM-IV-TR) criteria for ADHD (American
Psychiatric Association 2000) both at present and in
childhood (onset of symptoms before the age of
7 years according to DSM-IV-TR criteria) based on
Conners Adult ADHD Diagnostic Interview for
DSM-IV (CAADID) Japanese version at screening.
The CAADID assesses patients based on the 18
symptoms criteria for ADHD contained in the
DSM-IV. To be enrolled in this study, patients
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
OROS MPH in adults with ADHD in Japan 3
Effi cacy evaluation
The primary and secondary analysis populations
included the full analysis set, defi ned as patients who
received at least one dose of study drug and had base-
line and at least one post-dose effi cacy assessment.
The Japanese version of the CAARS-O:SV and
the Conners Adult ADHD Rating Scale-Self:
Screening Version (CAARS-S:SV) were used in this
study (Takahashi et al. 2011b). The CAARS-O:
SV consists of 30 investigator-rated items to measure
symptoms of ADHD in adult patients. Items are
rated on a scale of 0 (not at all) to 3 (very much/very
frequently). The CAARS-O:SV has four subscales:
the DSM-IV Total ADHD Symptoms subscale (18
items), the DSM-IV Inattentive Symptoms subscale
(nine items), the DSM-IV Hyperactive-Impulsive
Symptoms subscale (nine items) and the ADHD
Index (12 items). The CAARS-O:SV also provides a
total score. The CAARS-S:SV has the same struc-
ture as the observer version, but it is based on patient
self-report. For both the CAARS-O:SV and the
CAARS-S:SV, a lower score indicates improvement.
For standardization of the results, all investigators
who administered the CAARS and CAADID were
trained and certifi ed in these assessments before the
study. In addition, the same investigator adminis-
tered the assessments to a specifi c patient at all visits
whenever possible.
Effi cacy was also assessed using the Clinical Global
Impression Severity (CGI-S) and CGI-C rating
scales. The CGI-S was used to rate the severity of
CAARS-O:SV decreased by 30% from baseline and
the patient received a Clinical Global Impression -
Change (CGI-C) rating of 1 (very much improved)
or 2 (much improved), or the maximum dose of 72
mg/day was reached. Once an individually optimized
dose had been achieved, patients remained on that
dose until the end of the titration period. If necessary
for safety reasons, the dose could be decreased by
18 mg once during the titration period. Once the
patient s dose was decreased, it could not be subse-
quently increased. During the effi cacy assessment
period, patients continued on their individualized
dose of OROS MPH or matching placebo for the
remainder of the double-blind treatment period.
The fi nal part of the study, the post-study phase,
consisted of a single follow-up visit 1 week after the
patient s fi nal dose of study agent. During the post-
study visit, additional safety data were collected, and
patients were assessed for possible symptoms of
withdrawal.
Ethical considerations
The study protocol and amendments were reviewed
by an institutional review board, and the study was
conducted in accordance with the ethical principles
of the Declaration of Helsinki, Good Clinical Prac-
tice (GCP), and applicable regulatory requirements.
The study is registered at ClinicalTrials.gov
(NCT01323192). All patients provided their written
consent to participate in the study.
Double-blind phase (8 weeks)
Efficacy assessment
period (4 weeks)
Titration period (4 weeks)
Screening
phase
Post-study
phase
OROS MPH treatment
Placebo treatment
Washout,
eligibility
For all
patients
who
received
the study
treatment
Patients
completed
the study
Baseline entry,
randomization
Informed
consent
49-week open
label long-term
extension study
Visit
Week
Day
1
-2/-1
-14/-7
8
8
56
9
9
+7
7
6
42
6
4
28
5
3
21
2
-
0
4
2
14
3
1
7
18 mg
36 mg
18 mg
54 mg
36 mg
18 mg
72 mg
54 mg
36 mg
18 mg
Individualized
dose
Figure 1. Study design. OROS, osmotic-controlled release oral delivery system; MPH, methylphenidate.
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
4 N. Takahashi et al.
completed suicide; if the same patient experienced
multiple events, all events were used for analysis. The
C-SSRS was administered at baseline, Week 2, Week
4, Week 6, the end of the double-blind phase (Week
8) and the post-study visit.
Sample size determination
The calculation of sample size was based on results
from a completed international Phase 3 study
(Medori et al. 2008). The mean (SD) change from
baseline to endpoint in the DSM-IV Total ADHD
Symptoms subscale (18 items) of the investigator-
rated CAARS-O:SV in the placebo, OROS MPH 18
mg/day, OROS MPH 36 mg/day and OROS MPH
72 mg/day groups was 7.6 (9.93), 10.6 (10.34),
11.5 (9.97) and 13.7 (11.11), respectively (Medori
et al. 2008). The mean difference between the pla-
cebo group and each active dose group (18, 36 and
72 mg) was 3.0, 3.9 and 6.1, respectively (Medori
et al. 2008). Based on these results, assuming a mean
difference between the OROS MPH group and the
placebo group of 4.0 and a standard deviation of
each treatment group of 10.0, the required number
of patients for analysis in this study was 133 per
treatment group to achieve a power of 90%. Assum-
ing that approximately 5% of enrolled patients would
be excluded from analyses, a total of 280 patients
(140 patients per treatment group) were planned to
be enrolled to ensure at least 133 evaluable patients
per treatment group.
Randomization and blinding
Patients were randomly assigned to receive OROS
MPH or placebo based on a computer-generated
randomization schedule prepared before the study
by the sponsor or under the supervision of the spon-
sor. The randomization was balanced by using ran-
domly permuted blocks of treatment and was
stratifi ed by study centre. Study drug code numbers
were preprinted on the study drug labels and were
assigned to patients as they qualifi ed for the study
and were randomly assigned to treatment.
To maintain the study blind, the study drug con-
tainer had a multipart label containing the protocol
number, medication kit number, code number and
other information on each part. It was impossible to
identify the study drug by using the label informa-
tion. The study drugs were identical in appearance
and were packaged in identical containers. Data that
may potentially unblind the treatment assignment
(e.g., study agent serum concentrations) were to be
handled with special care to ensure that the integrity
of the blind was maintained and the potential for
the patient s illness on a seven-point scale ranging
from 1 (not ill) to 7 (extremely ill). The CGI-C was
used to rate the patient s total improvement due to
treatment with the study agent on a seven-point scale
ranging from 1 (very much improved) to 7 (very
much worse). A lower score on the CGI-S or CGI-C
rating scales indicates improvement. The Quality
of Life Enjoyment and Satisfaction Questionnaire
Short Form (Q-LES-Q-SF) was used to assess the
degree of enjoyment and satisfaction experienced
by patients in various areas of daily functioning,
including physical health, feelings, work, household
duties, leisure time activities and social relations.
The Q-LES-Q-SF is a self-administered question-
naire that consists of 16 items rated on a scale from
1 (very poor) to 5 (very good). A higher total score
on the Q-LES-Q-SF indicates improvement.
The primary effi cacy endpoint was the change
in DSM-IV Total ADHD Symptoms subscale scores
from baseline to the end of the double-blind treat-
ment period. Secondary effi cacy endpoints included
the change in CAARS-O:SV total score and subscale
scores (other than the DSM-IV Total ADHD Symp-
toms subscale) and the change in CAARS-S:SV total
score and subscale scores. CGI-S score, CGI-C
score and Q-LES-Q-SF total score were also included
as secondary effi cacy endpoints.
Safety evaluation
Treatment-emergent adverse events (TEAEs) were
recorded for the safety analyses, which also included
clinical laboratory test results, vital sign measure-
ments, electrocardiogram (ECG) parameters and
body weight. The safety analysis population was
defi ned as all patients who received at least one dose
of study drug.
To assess the potential for abuse of study drug, a
six-item questionnaire was administered at the post-
study visit (Week 9). Patients were asked to choose
the most appropriate answer to questions asking
about their experiences in obtaining, taking and run-
ning out/not taking study drug, as well as the con-
sistency between prescribed and actual dose of drug
taken.
As suicide risk has not yet been evaluated in adults
with ADHD receiving OROS MPH, patients in this
study were also administered the Columbia-Suicide
Severity Rating Scale (C-SSRS) to evaluate suicidal
tendency. The C-SSRS contains two categories
suicidal ideation and suicidal behaviour. Suicidal
ideation responses are scaled from 1 to 5, with a
higher number indicating a high degree of severity,
while suicidal behaviour was classifi ed as actual
attempt, interrupted attempt, aborted attempt, pre-
paratory acts or behaviour, suicidal behaviour, or
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
OROS MPH in adults with ADHD in Japan 5
CGI-C scores. For missing data in the effi cacy anal-
yses, the last observation carried forward (LOCF)
method was used. All statistical tests were performed
at the 5% level of signifi cance (two-sided). For cal-
culation of confi dence intervals (CIs), two-tailed
95% CIs were used. No adjustments were made for
multiple comparisons.
Results
Of the 306 patients who provided informed consent,
284 patients were randomized to treatment and
were included in the safety analysis population, and
283 patients were included in the full analysis set
(Figure 2). One patient in the placebo group was
excluded from the full analysis set because no post-
dose effi cacy data were available for this patient. The
demographic and baseline characteristics were well-
balanced between treatment groups (OROS MPH
group, n 143; placebo group, n 141; Table I). Of
the randomized patients, 134 (93.7%) patients in
the OROS MPH group and 135 (95.7%) patients
in the placebo group completed the double-blind
treatment period. Within 3 months prior to screen-
ing, 14.7% of patients in the OROS MPH group
and 12.1% of patients in the placebo group received
one or more psychotropic agents for ADHD.
The mean duration of exposure to study drug was
54.0 days in the OROS MPH group and 55.1 days
bias was minimized. This could include making
special provisions, such as segregating the data in
question from view by the investigators, clinical
team, or others as appropriate until the time of data-
base lock and unblinding.
Statistical analyses
An analysis of covariance (ANCOVA) model using
treatment and sex as factors and baseline score as a
covariate was used to assess the change from base-
line in CAARS-O:SV, CAARS-S:SV and Q-LES-
Q-SF scores. For the change from baseline in
DSM-IV Total ADHD Symptoms subscale score,
subgroup analyses were performed by the same
ANCOVA model used for the primary analysis, and
using the following factors: age at ADHD diagnosis,
type of ADHD at entry, current psychiatric comor-
bidity, history of or current psychiatric comorbidity,
age at informed consent, sex, body mass index
(BMI) at baseline, exposure to MPH during the
3-month period before screening, fi nal dose of study
drug and baseline DSM-IV Total ADHD Symptoms
subscale score of the CAARS-O:SV. CGI-S score
was analysed using an ANCOVA model on the ranks
of change from baseline with treatment as a factor
and baseline score as a covariate. An analysis of vari-
ance (ANOVA) model on the ranks of actual values
with treatment as a factor was used to evaluate
Screened patients
N = 306
Randomized patients
N = 284
Placebo
n = 141
Completed
n = 134
Completed
n = 135
Discontinued n = 9
AE, n = 6
Withdrawal
of consent, n = 3
Discontinued n = 6
AE, n = 1
Noncompliance with
study drug, n = 3
Nonmedical
reasons, n = 1
Withdrawal
of consent, n = 1
OROS MPH
n = 143
Figure 2. Disposition of study patients. OROS, osmotic-controlled release oral delivery system; MPH, methylphenidate; AE, adverse
event.
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
6 N. Takahashi et al.
effi cacy endpoint, none of the examined factors
appeared to affect the treatment effect of OROS
MPH relative to placebo (data not shown).
The mean changes from baseline to the end of the
double-blind period were signifi cantly larger in
the OROS MPH group than in the placebo group
for the CAARS-O:SV total score ( P 0.0002) and
subscale scores (Inattentive Symptoms, P 0.0001;
Hyperactive-impulsive Symptoms, P 0.0053; ADHD
Index, P 0.0030; Table II). The mean changes
from baseline were also signifi cantly larger in the
OROS MPH group than in the placebo group for
the CAARS-S:SV total score ( P 0.0003) and sub-
scale scores (Total ADHD Symptoms, P 0.0003;
Inattentive Symptoms, P 0.0001; Hyperactive-
impulsive Symptoms, P 0.0471; ADHD Index,
P 0.0006; Table II).
The improvement in CGI-S score and CGI-C
score was signifi cantly greater in the OROS MPH
group than in the placebo group ( P 0.0001; Table
II). The frequency distribution of CGI-S and
CGI-C scores over time are shown in Figure 4. For
both CGI-S and CGI-C scores, a greater percent-
age of patients in the OROS MPH group experi-
enced improvement compared with the placebo
group. The percentage of patients assessed as mod-
erately ill or worse on the CGI-S scale decreased
from 97.2% at baseline to 43.4% at endpoint in
the OROS MPH group and from 92.9 to 62.1%
in the placebo group. At endpoint, the percentage
in the placebo group. The majority of patients
( 93%) in both groups received the study drug for
at least 50 days. The mean dose and mean fi nal dose
of study drug was 44.2 and 51.5 mg, respectively, in
the OROS MPH group and 50.7 and 61.1 mg,
respectively, in the placebo group. At Week 8 (the
end of the double-blind period), 42.5% of patients
in the OROS MPH group and 72.1% of patients in
the placebo group received the maximum dose of
72 mg/day. The remaining patients in the OROS
MPH group received 18 mg/day (11.9%), 36 mg/day
(26.9%), or 54 mg/day (18.7%) at Week 8.
The baseline mean (SD) DSM-IV Total ADHD
Symptoms subscale score of the CAARS-O:SV was
similar between groups: 31.8 (5.96) for the OROS
MPH group versus 31.5 (6.44) for the placebo
group. The least-squares mean change in the DSM-IV
Total ADHD Symptoms subscale score of the
CAARS-O:SV from baseline to the end of the
double-blind period was 12.5 (95% CI, 14.0 to
11.0) in the OROS MPH group and
7.9 (95% CI,
9.5 to 6.4) in the placebo group (Table II). The
decrease (improvement) was signifi cantly larger in
the OROS MPH group ( P 0.0001). A statistically
signifi cant difference between the OROS MPH and
placebo groups with regard to the mean change from
baseline in the Total ADHD Symptoms score of the
CAARS-O:SV was initially observed at Week 2;
this difference was maintained in subsequent weeks
(Figure 3). In the subgroup analyses of the primary
Table I. Demographic and baseline characteristics (safety analysis population).
Characteristic
OROS MPH
( n 143)
Placebo
( n 141)
Age, years
Mean (SD) 33.4 (8.85) 34.1 (9.05)
Range 18 58 19 59
Gender, n (%)
Male 71 (49.7) 68 (48.2)
Female 72 (50.3) 73 (51.8)
Age at initial ADHD diagnosis, years
Mean (SD) 31.4 (9.81) 32.4 (9.98)
Range 5 58 1 58
Type of ADHD at study entry, n (%)
Predominantly inattentive 80 (55.9) 79 (56.0)
Predominantly hyperactive-impulsive 1 (0.7) 1 (0.7)
Combined type 62 (43.4) 61 (43.3)
Type of ADHD in childhood, n (%)
Predominantly inattentive 77 (53.8) 78 (55.3)
Predominantly hyperactive-impulsive 1 (0.7) 0 (0)
Combined type 65 (45.5) 63 (44.7)
Baseline DSM-IV Total ADHD
Symptoms score of CAARS-O:SV
Mean (SD) 31.8 (5.96) 31.5 (6.42)
OROS, osmotic-controlled release oral delivery system; MPH, methylphenidate; SD, standard deviation;
ADHD, attention-defi cit/hyperactivity disorder; DSM-IV, Diagnostic And Statistical Manual of Mental
Disorders 4th Edition; CAARS-O:SV, Conners Adult ADHD Rating Scale-Observer: Screening
Version.
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
OROS MPH in adults with ADHD in Japan 7
a 5% higher incidence in the OROS MPH group
than in the placebo group.
There were two serious TEAEs (psychotic disor-
der and pneumothorax spontaneous tension, n 1
for each) reported in the OROS MPH group. The
relationship between the study drug and psychotic
disorder was considered probable, while pneumotho-
rax spontaneous tension was considered by the inves-
tigator to be unrelated to the study drug. There were
no serious TEAEs in the placebo group. No deaths
were reported in this study. Discontinuation of study
drug due to one or more TEAEs was reported in 6
(4.2%) patients in the OROS MPH group and one
(0.7%) patient in the placebo group.
During the post-study period, 13 (9.1%) patients
in the OROS MPH group and fi ve (3.5%) patients
in the placebo group experienced at least one AE.
The only AE that was reported by more than one
patient in the OROS MPH group during the post-
study period was nasopharyngitis ( n 3).
of patients assessed as much improved or very
much improved on the CGI-C scale was 48.3% in
the OROS MPH group and 27.9% in the placebo
group. Based on Q-LES-Q-SF total scores, there
was no signifi cant difference in daily functioning
between the OROS MPH group and the placebo
group (Table II).
The overall incidence of TEAEs was higher in the
OROS MPH group (117/143; 81.8%) than in the
placebo group (76/141; 53.9%). TEAEs occurring
in 5% of patients in either treatment group during
the double-blind period are shown in Figure 5. The
most commonly reported TEAEs in the OROS MPH
group ( 5% of patients) were decreased appetite
(39.9%), palpitations (18.2%), nasopharyngitis
(16.8%), nausea (14.7%), thirst (14.0%), insomnia
(10.5%), headache (8.4%), weight decrease (7.0%)
and tachycardia (5.6%). Among the commonly
reported TEAEs, decreased appetite, palpitations,
nausea, thirst, weight decrease and tachycardia had
Table II. Change from baseline to end of double-blind period (LOCF) for effi cacy analyses.
Effi cacy measure OROS MPH ( n 143) Placebo ( n 140) P value
CAARS-O:SV
DSM-IV Total ADHD Symptoms
LS means (95% CI) change 12.5 ( 14.0 to 11.0) 7.9 ( 9.5 to 6.4) 0.0001
Total score
Mean (SD) change 19.5 (15.42) 12.5 (15.87) 0.0002
Inattentive Symptoms
Mean (SD) change 8.0 (6.26) 5.0 (5.93) 0.0001
Hyperactive-impulsive Symptoms
Mean (SD) change 4.5 (4.51) 2.9 (4.84) 0.0053
ADHD Index
Mean (SD) change 7.0 (6.99) 4.6 (6.95) 0.0030
CAARS-S:SV
DSM-IV Total ADHD Symptoms
Mean (SD) change 10.7 (9.85)
a 6.6 (10.06) 0.0003
Total score
Mean (SD) change 18.0 (16.45)
a 10.9 (16.55) 0.0003
Inattentive Symptoms
Mean (SD) change 6.5 (6.35)
a 3.4 (5.99) 0.0001
Hyperactive-impulsive Symptoms
Mean (SD) change 4.2 (4.73)
a 3.2 (5.02) 0.0471
ADHD Index
Mean (SD) change 7.3 (7.24)
a 4.3 (6.69) 0.0006
CGI-S
Median (range) change 1.0 ( 5 to 1) 1.0 ( 4 to 2) 0.0001
CGI-C
Median (range) change 3 (1 to 6) 3 (1 to 6) 0.0001
Q-LES-Q-SF
Mean (SD) change 2.4 (14.00)
b 0.8 (12.69) 0.4041
LOCF, last observation carried forward; OROS, osmotic-controlled release oral delivery system; MPH,
methylphenidate; CAARS-O:SV, Conners Adult ADHD Rating Scale-Observer: Screening Version;
DSM-IV, Diagnostic And Statistical Manual of Mental Disorders 4th Edition; ADHD, attention-defi cit/
hyperactivity disorder; LS, least squares; CI, confi dence interval; SD, standard deviation; CAARS-S:SV,
Conners Adult ADHD Rating Scale-Self: Screening Version; CGI-S, Clinical Global Impression Severity;
CGI-C, Clinical Global Impression Change; Q-LES-Q-SF, Quality of Life Enjoyment and Satisfaction
Questionnaire Short Form.
a n 141.
b n 142.
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
8 N. Takahashi et al.
in Japan. OROS MPH was effective and well-
tolerated in adults, consistent with previous studies
in other adult patient populations (Biederman et al.
2007; Medori et al. 2008). Based on analysis of the
primary endpoint, OROS MPH was statistically
superior to placebo. The superiority of OROS MPH
in reducing ADHD symptoms was initially observed
at Week 2 and continued throughout the remainder
of the double-blind period. Subgroup analyses of
the primary endpoint confi rmed that no specifi c fac-
tor, such as age at ADHD diagnosis, type of ADHD
at entry, or baseline DSM-IV Total ADHD Symp-
toms subscale score of the CAARS-O:SV, affected
responsiveness to OROS MPH in this patient popu-
lation. The results of the majority of the secondary
endpoints consistently demonstrated the superiority
of OROS MPH over placebo except Q-LES-Q-SF.
Because ADHD was originally considered to only
affect children and adolescents, there have been
many more studies of this condition in these patient
populations than in adults (Asherson et al. 2007).
Therefore, the validity of the diagnosis of ADHD
in adults is still questioned by some clinicians and
researchers (Medori et al. 2008). More recently,
awareness of adult ADHD has increased due
to mounting evidence supporting the persistence
of symptoms into adulthood (Kooij et al. 2005;
Buitelaar et al. 2009). In addition, treatment with
No AEs related to drug abuse or misuse were
reported during the double-blind or post-study period.
Additionally, a minority of patients indicated that they
wanted to take more than the prescribed dose ( n 9
in the OROS MPH group and n 8 in the placebo
group) or to continue taking study drug for any rea-
son other than drug effi cacy ( n 5 in the OROS
MPH group and none in the placebo group).
In general, no clinically signifi cant trend in vital
signs, body weight, clinical laboratory test values, or
ECG parameters was observed in either group.
However, 10 patients in the OROS MPH group
experienced weight decrease compared with no
patients in the placebo group.
At any time during double-blind phase, nine
(6.3%) patients in the OROS MPH and nine (6.4%)
patients in the placebo group answered yes to at
least one of fi ve questions asking about suicidal ide-
ation. One patient in the placebo group answered
yes to two of six questions asking about suicidal
behaviour, while no patient in the OROS MPH
group answered yes to these questions.
Discussion
This study was designed to evaluate the effi cacy and
safety of OROS MPH in adult patients with ADHD
–20
–15
–10
–5
0
5
Endpoint864321Baseline
Change from baseline (mean ± SE)
Number of patients
OROS MPH
Placebo
P value
143
140
142
140
0.0796
142
140
0.0131
143
140
0.0014
143
140
0.0007
143
140
0.0001
143
140
<0.0001
143
140
<0.0001
PlaceboOROS MPH
Week
Figure 3. Mean change from baseline in DSM-IV Total ADHD Symptoms score of CAARS-O:SV over time. DSM-IV, Diagnostic And
Statistical Manual of Mental Disorders 4th Edition; ADHD, attention-defi cit/hyperactivity disorder; CAARS-O:SV, Conners Adult
ADHD Rating Scale-Observer: Screening Version; OROS, osmotic-controlled release oral delivery system; MPH, methylphenidate;
SE, standard error.
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
OROS MPH in adults with ADHD in Japan 9
EndpointWeek 8Week 4BaselineEndpointWeek 8Week 4Baseline
Patients (%)
Not ill/borderline ill
PlaceboOROS MPH
0
20
40
60
80
100
Mildly ill Moderately ill
Markedly ill Severely ill/extremely ill
EndpointWeek 8Week 4EndpointWeek 8Week 4
Patients (%)
Very much improved
PlaceboOROS MPH
0
20
40
60
80
100
Much improved Minimally improved
No change Minimally worse/much worse/very much worse
(A)
(B)
Figure 4. Frequency distribution of (A) CGI-S and (B) CGI-C scores over time. CGI-S, Clinical Global Impression Severity; CGI-C,
Clinical Global Impression Change; OROS, osmotic-controlled release oral delivery system; MPH, methylphenidate.
medication has been shown to reduce ADHD symp-
toms in adults with similar effi cacy to that seen
in children (Asherson et al. 2007; Medori et al.
2008). In this study, adult patients were screened
for inclusion using the Japanese version of the
CAADID to confi rm the diagnosis of ADHD. The
psychometric properties of the original English
version of this structured interview have been eval-
uated and have been found to be adequate for the
diagnosis of ADHD in adult patients (Epstein and
Kollins 2006). Additionally, the Japanese version
has been validated from the English version (Takahashi
et al. 2011b). A confi rmed diagnosis of ADHD
based on the CAADID is widely used as a criterion
for inclusion in clinical trials in adult patient popu-
lations (Medori et al. 2008; Buitelaar et al. 2011;
Takahashi et al. 2011a).
While treatment with OROS MPH resulted in
statistically signifi cant improvements in ADHD
symptoms relative to placebo, a smaller reduction in
ADHD symptoms was observed in the placebo
group. The placebo effect that was observed in this
study is consistent with previous studies that evalu-
ated OROS MPH in other adult patient populations
(Biederman et al. 2007; Medori et al. 2008; Buitelaar
et al. 2009, 2011). One possible explanation for the
placebo effect observed in this study is that there
may have been an apparent benefi t for patients to
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
10 N. Takahashi et al.
reported in 18.2% of patients in this study. The low
risk of suicide of OROS MPH was also confi rmed.
The potential for abuse continues to be a major
concern for regulatory authorities in Japan regarding
the use of stimulants for the treatment of adult
patients with ADHD (Takahashi et al. 2011a). In
comparison with immediate-release formulations of
MPH, extended-release OROS MPH has been
shown to have a lower risk of misuse (Biederman
et al. 2007; Buitelaar et al. 2009). The results of this
study support the low abuse potential of OROS
MPH. During the double-blind and post-study peri-
ods, there were no AEs related to drug abuse or mis-
use reported. Data from a questionnaire support this
notion. In addition, only one AE (nasopharyngitis)
was reported by more than one patient in the OROS
MPH group during the post-study period. This sug-
gests that after discontinuation of treatment with
OROS MPH, there were no dominant AEs that
would be indicative of a withdrawal phenomenon.
This will be further confi rmed in a 49-week open
label long-term extension study in which patients
who completed this study were eligible to enroll.
A limitation of this study was the short treatment
period (8 weeks); additional studies will be needed
to evaluate the long-term effi cacy and safety of
OROS MPH in adults. A longer study period could
assess whether the short-term effects observed in
this study are maintained over the course of long-
term treatment. Additionally, it is possible that
a signifi cant improvement in Q-LES-Q-SF score
in the OROS MPH group compared with the pla-
cebo group may have emerged if the study had been
enrol in the study, including those who were ran-
domized to the placebo group. Although initiation of
new psychosocial treatment for patients was restricted
during the observation period, all patients in the
study could obtain medical advice and education
about their illness from the investigators.
There are several possible reasons for the low
dropout rate that was observed in this study. First,
MPH had been approved for the treatment of chil-
dren with ADHD in Japan, but there was no medica-
tion approved for adults with ADHD at the time of
the study. Because of this situation, it could be con-
sidered that the motivation for patients to continue
participation in this study was very high. Second,
there was a dose titration period in this trial, which
may have decreased the rate of dropout due to lack
of effi cacy or AEs. Third, only patients who com-
pleted this initial study were eligible for enrolment
in the open label long-term extension study.
The results of this study support the safety of
OROS MPH for use in adult patients. Among the
commonly reported TEAEs, several were observed
more frequently with OROS MPH than placebo,
including decreased appetite, palpitations and weight
decrease. These were expected, mild to moderate
in severity, and did not generally lead to treatment
discontinuation. Additionally, with the exception of
palpitations, these AEs were similar to those observed
in studies that have been conducted in other adult
patient populations (Medori et al. 2008). In a study
conducted in Europe, the incidence of palpitations
was 3.9% among patients treated with OROS MPH
(Medori et al. 2008). In contrast, palpitations were
0 5 10 15 20 25 30 35 40 45 50
Weight decrease
Thirst
Nausea
Tachycardia
Palpitations
Headache
Insomnia
Decreased appetite
Nasopharyngitis 16.8%
39.9%
10.5%
8.4%
18.2%
0%
5.6%
14.7%
14.0%
7.0%
13.5%
7.1%
9.9%
6.4%
1.4%
2.8%
4.3%
0%
PlaceboOROS MPH
Patients (%)
Figure 5. TEAEs occurring in 5% of patients in either treatment group during the double-blind period. TEAE, treatment-emergent
adverse event; OROS, osmotic-controlled release oral delivery system; MPH, methylphenidate.
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
OROS MPH in adults with ADHD in Japan 11
Asherson P , Chen W , Craddock B , Taylor E . 2007 . Adult atten-
tion-defi cit hyperactivity disorder: recognition and treatment in
general adult psychiatry . Br J Psychiatry 190 : 4 5 .
Baron DA , Pato MT , Cyr RL . 2011 . Treatment of adults with
attention-defi cit/hyperactivity disorder . J Am Osteopath Assoc
111 : 610 614 .
Biederman J , Mick EO , Surman C , Doyle R , Hammerness P ,
Michel E , et al . 2007 . Comparative acute effi cacy and tolerability
of OROS and immediate release formulations of methylphenidate
in the treatment of adults with attention-defi cit/hyperactivity dis-
order . BMC Psychiatr y 7 : 49 .
Buitelaar JK , Ramos-Quiroga JA , Casas M , Kooij JJ , Niemela A ,
Konofal E , et al . 2009 . Safety and tolerability of fl exible dosages
of prolonged-release OROS methylphenidate in adults with
attention-defi cit/hyperactivity disorder . Neuropsychiatr Dis
Treat 5 : 457 466 . Epub@2009 Sep 15:457 466.
Buitelaar JK , Trott GE , Hofecker M , Waechter S , Berwaerts J ,
Dejonkheere J , et al . 2011 . Long-term effi cacy and safety
outcomes with OROS-MPH in adults with ADHD . Int J
Neuropsychopharmacol 15 : 1 13 .
Concer ta [package insert] . Tokyo: Janssen Pharmaceutical KK ;
2007 .
Conners CK , Erhardt D , Sparrow E . 1999 . Conners Adult ADHD
Rating Scales (CAARS): Technical Manual . NY: Multi-Health
Systems Inc .
Epstein JN , Kollins SH . 2006 . Psychometric properties of an adult
ADHD diagnostic interview . J Atten Disord 9 : 504 514 .
Johnson & Johnson Pharmaceutical Research & Development .
2011 . Investigator s Brochure, CONCERTA
®
(methylpheni-
date HCl) . Edition 16.
Kooij JJ , Buitelaar JK , van den Oord EJ , Furer JW , Rijnders CA ,
Hodiamont PP . 2005 . Internal and external validity of
attention-defi cit hyperactivity disorder in a population-based
sample of adults . Psychol Med 35 : 817 827 .
Medori R , Ramos-Quiroga JA , Casas M , Kooij JJ , Niemela A ,
Trott GE , et al . 2008 . A randomized, placebo-controlled trial
of three fi xed dosages of prolonged-release OROS methylphe-
nidate in adults with attention-defi cit/hyperactivity disorder .
Biol Psychiatry 63 : 981 989 .
Pelham WE , Gnagy
EM , Burrows-Maclean L , Williams A ,
Fabiano GA , Morrisey SM , et al . 2001 . Once-a-day Concerta
methylphenidate versus three-times-daily methylphenidate in
laboratory and natural settings . Pediatrics 107 : E105 .
Pharmaceuticals and Medical Devices Agency . 2012 . [in Japanese].
Available at: http://www.info.pmda.go.jp/shinyaku/P201200114/
53047100_22100AMX00644_A100_1.pdf.
Takahashi M , Takita Y , Goto T , Ichikawa H , Saito K ,
Matsumoto H , et al . 2011a . An open-label, dose-titration
tolerability study of atomoxetine hydrochloride in Japanese
adults with attention-defi cit/hyperactivity disorder . Psychiatry
Clin Neurosci 65 : 55 63 .
Takahashi M , Takita Y , Ichikawa
H , Enomoto T , Okada T ,
Saito K , et al . 2011b . Reliability and Validity of the Japanese
Version of CAARS-Screening Version (CAARS-SV), Adult
ADHD Symptom Rating Scale [in Japanese] . Seishin Igaku
53 : 23 34 .
Takeda T . 2009 . Psychopharmacology for attention-defi cit/hyper-
activity disorder in Japan . Curr Attention Dis Rep 1 : 21 28 .
Wilens TE , Morrison NR , Prince J . 2011 . An update on the
pharmacotherapy of attention-defi cit/hyperactivity disorder in
adults . Expert Rev Neurother 11 : 1443 1465 .
conducted over a longer period of time. The contin-
ued safety and effi cacy of OROS MPH in adults will
be assessed in the long-term extension study. Third,
ADHD is more likely to be complicated with other
psychiatric illness, such as anxiety disorders. We
excluded these patients in this study, and the effi cacy
of OROS MPH for these patients should be tested
in the future. It could be considered that excluding
non-responders to MPH from this study may have
had a positive bias on patient selection. However,
no patient was excluded from the study due to
this exclusion criterion, so it is unlikely that this
exclusion had an effect on the overall results. Finally,
this study was funded and executed by Janssen
Pharmaceuticals, which could be perceived as a
potential confl ict of interest. It should be noted
that the study was performed by strictly following
GCP guidelines.
In conclusion, the results of this study demon-
strated that OROS MPH in a dose range of 18 72
mg/day was effective in the treatment of ADHD
in adult patients. Compared with placebo, patients
who received OROS MPH experienced signifi cant
improvements in ADHD symptoms based on mul-
tiple measures of assessment. OROS MPH was
also well-tolerated in adult patients in Japan, with an
AE profi le that was mostly similar to other clinical
studies in distinct patient populations.
Acknowledgements
We would like to acknowledge all patients, physicians
and research coordinators who participated in this
clinical trial.
Statement of interest
NT, TK, YT, YS, YK and TM are employees of
Janssen Pharmaceutical KK, Japan.
This study was supported by Janssen Pharmaceu-
tical KK, Japan. Editorial support was provided by
Allison Michaelis, PhD, of Medergy and was funded
by Janssen Global Services, LLC.
References
American Psychiatric Association . 2000 . Attention-defi cit and dis-
ruptive behavior disorders . In: Diagnostic and statistical man-
ual of mental disorders. 4th ed. text revision (DSM-IV-TR) .
Washington, DC: American Psychiatric Association . p 85 93 .
Supplementary material available online
Supplementary Table I to be found online at http://
informahealthcare.com/doi/abs/10.3109/15622975.
2013.868925.
World J Biol Psychiatry Downloaded from informahealthcare.com by 201.210.105.125 on 05/20/14
For personal use only.
... The data extracted from analyzed studies are presented in detail in Supplementary materials in Table S1 [28][29][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46]. ...
... However, none of these situations led to permanent discontinuation of treatment and exclusion from the analysis by the investigators [37,46]. A psychotic disorder was reported once in the group taking MPH, which was assessed as a likely outcome of MPH use [42]. Psychotic and manic symptoms with methylphenidate have been reported in the medical literature. ...
... The researchers considered this case as possibly treatmentrelated [32]. One adverse reaction reported by more than one person from the MPH population in the post-study period was nasopharyngitis [42]. There was one death of a 51-year-old man reported 21 days after the end of the study (21 days after receiving the last dose of study drug) due to rupture of aortic dissection. ...
Article
Full-text available
Attention deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders. It was once thought to be a disorder affecting only children, but in those undiagnosed in childhood, symptoms do not disappear with age. There is now a growing recognition of the late diagnosis and treatment of adults with ADHD. The first-line drug in pharmacotherapy is methylphenidate, and information about its adverse effects, when used by adults, has not been as extensively described as in children. The aim of this article was to review the literature describing the risks of methylphenidate therapy for adults with ADHD. A total of 19 articles—15 clinical trials and 4 case reports presenting rare side effects resulting from methylphenidate therapy, such as reversible ischemic stroke, myocardial infarction, and psychotic episodes, were analyzed. The analysis from clinical trials included 3458 adult patients with ADHD and described the most common side effects, psychiatric adverse events, effects of methylphenidate treatment on sleep, laboratory results, body mass, and cardiovascular symptoms. Methylphenidate treatment is well tolerated, with side effects described, according to severity, as mild to moderate. We conclude that pharmacotherapy is not risk-free and methylphenidate, due to its side effects, may not be the first drug of choice for every patient.
... Se identificó un ensayo clínico de ocho semanas de duración llevado a cabo en población adulta japonesa con metilfenidato de liberación osmótica con el objetivo de evaluar su seguridad y eficacia en adultos con TDAH 114 . Los participantes tenían entre 18 y 64 años de edad y un diagnóstico de TDAH antes de los 7 años de edad y en edad adulta. ...
... El MPH de liberación osmótica (OROS) fue eficaz y bien tolerado en adultos sin que ningún factor como la edad de diagnóstico o el grado de TDAH interaccionen con la eficacia 114 . ...
... Five RCTs [26,[47][48][49]75] were judged to be at low risk of bias across the six domains assessed (Fig 2). Less than half (46%) of the included RCTs were at low ROB for blinding, which is an important consideration for subjective outcomes. ...
... Compared with placebo, there was no significant difference in the risk of a serious adverse event with use of an ADHD pharmacotherapy (RR 1.46, 95% CI 0.52 to 4.09; I 2 = 0%) (Appendix M in S2 File). A total of 16 RCTs with any treatment duration reported the occurrence of serious adverse events [32,34,35,44,47,49,50,52,56,63,68,72,73,75,78,82], while an additional 17 RCTs reported that no serious adverse events had occurred during the study [21, 26, 31, 33, 40, 41, 48, 53, 76-78, 80, 81, 88, 93, 101, 103]. Compared with placebo, there was no significant difference in the risk of a serious adverse event with use of an ADHD pharmacotherapy (RR 1.21, 95% CI 0.67 to 2.18; I 2 = 0%). ...
Article
Full-text available
Background Attention deficit hyperactivity disorder (ADHD) affects approximately 3% of adults globally. Many pharmacologic treatments options exist, yet the comparative benefits and harms of individual treatments are largely unknown. We performed a systematic review and network meta-analysis to assess the relative effects of individual pharmacologic treatments for adults with ADHD. Methods We searched English-language published and grey literature sources for randomized clinical trials (RCTs) involving pharmacologic treatment of ADHD in adults (December 2018). The primary outcome was clinical response; secondary outcomes were quality of life, executive function, driving behaviour, withdrawals due to adverse events, treatment discontinuation, serious adverse events, hospitalization, cardiovascular adverse events, and emergency department visits. Data were pooled via pair-wise meta-analyses and Bayesian network meta-analyses. Risk of bias was assessed by use of Cochrane’s Risk of Bias tool, and the certainty of the evidence was assessed by use of the GRADE framework. Results Eighty-one unique trials that reported at least one outcome of interest were included, most of which were at high or unclear risk of at least one important source of bias. Notably, only 5 RCTs were deemed at overall low risk of bias. Included pharmacotherapies were methylphenidate, atomoxetine, dexamfetamine, lisdexamfetamine, guanfacine, bupropion, mixed amphetamine salts, and modafinil. As a class, ADHD pharmacotherapy improved patient- and clinician-reported clinical response compared with placebo (range: 4 to 15 RCTs per outcome); however, these findings were not conserved when the analyses were restricted to studies at low risk of bias, and the certainty of the finding is very low. There were few differences among individual medications, although atomoxetine was associated with improved patient-reported clinical response and quality of life compared with placebo. There was no significant difference in the risk of serious adverse events or treatment discontinuation between ADHD pharmacotherapies and placebo; however, the proportion of participants who withdrew due to adverse events was significantly higher among participants who received any ADHD pharmacotherapy. Few RCTs reported on the occurrence of adverse events over a long treatment duration. Conclusions Overall, despite a class effect of improving clinical response relative to placebo, there were few differences among the individual ADHD pharmacotherapies, and most studies were at risk of at least one important source of bias. Furthermore, the certainty of the evidence was very low to low for all outcomes, and there was limited reporting of long-term adverse events. As such, the choice between ADHD pharmacotherapies may depend on individual patient considerations, and future studies should assess the long-term effects of individual pharmacotherapies on patient-important outcomes, including quality of life, in robust blinded RCTs. Registration PROSPERO no. CRD 42015026049
... It also discusses the limitations and future directions of these potential biomarkers for application in clinical practice. (Takahashi et al., 2014), many studies focusing on this neurotransmitter system have been conducted to search for potential biomarkers of ADHD. Several studies have investigated dopamine (DA) and its metabolites using plasma, urine, and cerebrospinal fluid (CSF) from ADHD. ...
Article
Attention-deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders, characterized by a persistent pattern of inattention, hyperactivity, and impulsivity. Since the diagnosis of ADHD is defined by operational diagnostic criteria consisting of several clinical symptoms, a number of heterogeneous mechanisms have been considered to be implicated in its pathophysiology. Although no clinically reliable biomarkers are available for the diagnosis of ADHD, several plausible candidate biomarkers have been proposed based on recent advances in biochemistry and molecular biology. This review article summarizes potential peripheral biomarkers associated with ADHD, mainly from recently published case-control studies. These include 1) biochemical markers: neurotransmitters and their receptors, neurotrophic factors, serum electrolytes, and inflammation markers; 2) genetic and epigenetic markers: microRNA, mRNA expression, and peripheral DNA methylation; 3) physiological markers: eye movement and electroencephalography. It also discusses the limitations and future directions of these potential biomarkers for application in clinical practice.
Article
Importance Placebo is the only substance systematically evaluated across common psychiatric diagnoses, but comprehensive cross-diagnostic comparisons are lacking. Objective To compare changes in placebo groups in recent high-quality randomized clinical trials (RCTs) across a broad spectrum of psychiatric disorders in adult patients. Data Sources MEDLINE and the Cochrane Database of Systematic Reviews were systematically searched in March 2022 for the latest systematic reviews meeting predetermined high-quality criteria for 9 major psychiatric diagnoses. Study Selection Using these reviews, the top 10 highest-quality (ie, lowest risk of bias, according to the Cochrane Risk of Bias tool) and most recent placebo-controlled RCTs per diagnosis (totaling 90 RCTs) were selected, adhering to predetermined inclusion and exclusion criteria. Data Extraction and Synthesis Following the Cochrane Handbook, 2 authors independently carried out the study search, selection, and data extraction. Cross-diagnosis comparisons were based on standardized pre-post effect sizes (mean change divided by its SD) for each placebo group. This study is reported following the Meta-analysis of Observational Studies in Epidemiology (MOOSE) reporting guideline. Main Outcome and Measure The primary outcome, pooled pre-post placebo effect sizes ( d av ) with 95% CIs per diagnosis, was determined using random-effects meta-analyses. A Q test assessed statistical significance of differences across diagnoses. Heterogeneity and small-study effects were evaluated as appropriate. Results A total of 90 RCTs with 9985 placebo-treated participants were included. Symptom severity improved with placebo in all diagnoses. Pooled pre-post placebo effect sizes differed across diagnoses ( Q = 88.5; df = 8; P < .001), with major depressive disorder ( d av = 1.40; 95% CI, 1.24-1.56) and generalized anxiety disorder ( d av = 1.23; 95% CI, 1.06-1.41) exhibiting the largest d av . Panic disorder, attention-deficit/hyperactivity disorder, posttraumatic stress disorder, social phobia, and mania showed d av between 0.68 and 0.92, followed by OCD ( d av = 0.65; 95% CI, 0.51-0.78) and schizophrenia ( d av = 0.59; 95% CI, 0.41-0.76). Conclusion and Relevance This systematic review and meta-analysis found that symptom improvement with placebo treatment was substantial in all conditions but varied across the 9 included diagnoses. These findings may help in assessing the necessity and ethical justification of placebo controls, in evaluating treatment effects in uncontrolled studies, and in guiding patients in treatment decisions. These findings likely encompass the true placebo effect, natural disease course, and nonspecific effects.
Chapter
This chapter reviews the effectiveness of the psychostimulants amphetamine, methylphenidate, and modafinil in the treatment of mental disorders. It will provide an overview of the existing randomized, controlled studies that have been carried out with these substances. Although possible side effects must always be considered in relation to efficacy of psychopharmacological treatment of mental disorders, the main focus of this overview is on the evidence of effectiveness of psychostimulants. Wherever possible, the results of meta-analytical investigations of the existing studies are also presented.
Article
ADHD in adulthood is often overlooked, which negatively affects the individual’s well-being. First-line pharmacological interventions are effective in many ADHD patients, relieving symptoms rapidly. However, there seems to be a proportion of individuals who discontinue, or fail to respond to these treatments. For these individuals, alternative treatment options should be explored. A retrospective survey study reported that using classic psychedelics in low, repeated doses, so called microdosing (MD), was rated as being more effective than conventional treatments for ADHD. The current prospective study aimed to measure the effect of MD on ADHD symptoms, well-being and time perception. Adults with ADHD who had the intention to start MD on their own initiative to self-treat their symptoms were measured before MD and two- and four weeks later. It was expected that ADHD symptoms would decrease, well-being would increase, and performance on a time perception task would improve after MD. It was investigated if conventional medication use alongside MD and comorbidities alongside ADHD influenced the effect of MD. Sample sizes included N=233, N=66, and N=47, respectively. The results showed decreases and increases in ADHD symptoms and well-being, respectively. No improved performance on a time perception task was found. Conventional medication use and having comorbidities did not change the effect of MD on ADHD symptomatology and well-being after four weeks of MD. Placebo-controlled experimental studies are needed to explore further whether there is a beneficial effect of MD for ADHD, beyond the placebo-effect.
Article
Background: Attention deficit hyperactivity disorder (ADHD) is a psychiatric diagnosis increasingly used in adults. The recommended first-line pharmacological treatment is central nervous system (CNS) stimulants, such as methylphenidate, but uncertainty remains about its benefits and harms. Objectives: To assess the beneficial and harmful effects of extended-release formulations of methylphenidate in adults diagnosed with ADHD. Search methods: We searched CENTRAL, MEDLINE, Embase, nine other databases and four clinical trial registries up to February 2021. We searched 12 drug regulatory databases for clinical trial data up to 13 May 2020. In addition, we cross-referenced all available trial identifiers, handsearched reference lists, searched pharmaceutical company databases, and contacted trial authors. Selection criteria: Randomised, double-blind, parallel-group trials comparing extended-release methylphenidate formulations at any dose versus placebo and other ADHD medications in adults diagnosed with ADHD. Data collection and analysis: Two review authors independently extracted data. We assessed dichotomous outcomes as risk ratios (RRs), and rating scales and continuous outcomes as mean differences (MDs) or standardised mean differences (SMDs). We used the Cochrane risk of bias tool to assess risks of bias, and GRADE to assess the certainty of the evidence. We meta-analysed the data using a random-effects model. We assessed three design characteristics that may impair the trial results' 'generalisability'; exclusion of participants with psychiatric comorbidity; responder selection based on previous experience with CNS stimulants; and risk of withdrawal effects. Our prespecified primary outcomes were functional outcomes, self-rated ADHD symptoms, and serious adverse events. Our secondary outcomes included quality of life, ADHD symptoms rated by investigators and by peers such as family members, cardiovascular variables, severe psychiatric adverse events, and other adverse events. Main results: We included 24 trials (5066 participants), of which 21 reported outcome data for this review. We also identified one ongoing study. We included documents from six drug regulatory agencies covering eight trials. Twenty-one trials had an outpatient setting and three were conducted in prisons. They were primarily conducted in North America and Europe. The median participant age was 36 years. Twelve trials (76% of participants) were industry-sponsored, four (14% of participants) were publicly funded with industry involvement, seven (10% of participants) were publicly funded, and one had unclear funding. The median trial duration was eight weeks. One trial was rated at overall unclear risk of bias and 20 trials were rated at overall high risk of bias, primarily due to unclear blinding of participants and investigators, attrition bias, and selective outcome reporting. All trials were impaired in at least one of the three design characteristics related to 'generalisability'; for example, they excluded participants with psychiatric comorbidity such as depression or anxiety, or included participants only with a previous positive response to methylphenidate, or similar drugs. This may limit the trials' usefulness for clinical practice, as they may overestimate the benefits and underestimate the harms. Extended-release methylphenidate versus placebo (up to 26 weeks) For the primary outcomes, we found very low-certainty evidence that methylphenidate had no effect on 'days missed at work' at 13-week follow-up (mean difference (MD) -0.15 days, 95% confidence interval (CI) -2.11 to 1.81; 1 trial, 409 participants) or serious adverse events (risk ratio (RR) 1.43, CI 95% CI 0.85 to 2.43; 14 trials, 4078 participants), whereas methylphenidate improved self-rated ADHD symptoms (small-to-moderate effect; SMD -0.37, 95% CI -0.43 to -0.30; 16 trials, 3799 participants). For secondary outcomes, we found very low-certainty evidence that methylphenidate improved self-rated quality of life (small effect; SMD -0.15, 95% CI -0.25 to -0.05; 6 trials, 1888 participants), investigator-rated ADHD symptoms (small-to-moderate effect; SMD -0.42, 95% CI -0.49 to -0.36; 18 trials, 4183 participants), ADHD symptoms rated by peers such as family members (small-to-moderate effect; SMD -0.31, 95% CI -0.48 to -0.14; 3 trials, 1005 participants), and increased the risk of experiencing any adverse event (RR 1.27, 95% CI 1.19 to 1.37; 14 trials, 4214 participants). We rated the certainty of the evidence as 'very low' for all outcomes, primarily due to high risk of bias and 'indirectness of the evidence'. One trial (419 participants) had follow-up at 52 weeks and two trials (314 participants) included active comparators, hence long-term and comparative evidence is limited. Authors' conclusions: We found very low-certainty evidence that extended-release methylphenidate compared to placebo improved ADHD symptoms (small-to-moderate effects) measured on rating scales reported by participants, investigators, and peers such as family members. Methylphenidate had no effect on 'days missed at work' or serious adverse events, the effect on quality of life was small, and it increased the risk of several adverse effects. We rated the certainty of the evidence as 'very low' for all outcomes, due to high risk of bias, short trial durations, and limitations to the generalisability of the results. The benefits and harms of extended-release methylphenidate therefore remain uncertain.
Article
EEG studies have shown that adult ADHD patients have less stable brain arousal regulation than age and gender matched controls. Psychostimulants have brain arousal stabilising properties evident in EEG patterns. The aim of this study was to investigate whether the stability of brain arousal regulation has prognostic value in predicting response to methylphenidate therapy in adult ADHD patients. In an open-label, single-arm, multi-centre, confirmatory trial, 121 adult ADHD patients were recruited and 112 qualified for the full analysis set. All participants received an initial dose of 20 mg extended release methylphenidate at baseline. After a titration phase of up to 4 weeks, patients remained on a weight-based target dose of extended release methylphenidate for 4 weeks. Using the Vigilance Algorithm Leipzig (VIGALL 2.1), we assessed brain arousal regulation before the treatment with methylphenidate, based on a 15-min EEG at quiet rest recorded at baseline. Using automatic stage-classification of 1 s segments, we computed the mean EEG-vigilance (indexing arousal level) and an arousal stability score (indexing arousal regulation). The primary endpoint was the association between successful therapy, defined by a 30% reduction in CAARS, and stable/unstable brain arousal. 52 patients (46%) showed an unstable brain arousal regulation of which 23% had therapy success. In the stable group, 35% had therapy success, implying an absolute difference of 12 percentage points (95% CI −5 to 29, p = 0.17) in the direction opposite to the hypothesized one. There were no new findings regarding the tolerability and safety of extended release methylphenidate therapy.
Article
Full-text available
Objective: Methylphenidate (MPH), the most commonly prescribed drug for attention-deficit/hyperactivity disorder (ADHD), has a short half-life, which necessitates multiple daily doses. The need for multiple doses produces problems with medication administration during school and after-school hours, and therefore with compliance. Previous long-acting stimulants and preparations have shown effects equivalent to twice-daily dosing of MPH. This study tests the efficacy and duration of action, in natural and laboratory settings, of an extended-release MPH preparation designed to last 12 hours and therefore be equivalent to 3-times-daily dosing. Methods: Sixty-eight children with ADHD, 6 to 12 years old, participated in a within-subject, double-blind comparison of placebo, immediate-release (IR) MPH 3 times a day (tid), and Concerta, a once-daily MPH formulation. Three dosing levels of medication were used: 5 mg IR MPH tid/18 mg Concerta once a day (qd); 10 mg IR MPH tid/36 mg Concerta qd; and 15 mg IR MPH tid/54 mg Concerta qd. All children were currently medicated with MPH at enrollment, and each child's dose level was based on that child's MPH dosing before the study. The doses of Concerta were selected to be comparable to the daily doses of MPH that each child received. To achieve the ascending rate of MPH delivery determined by initial investigations to provide the necessary continuous coverage, Concerta doses were 20% higher on a daily basis than a comparable tid regimen of IR MPH. Children received each medication condition for 7 days. The investigation was conducted in the context of a background clinical behavioral intervention in both the natural environment and the laboratory setting. Parents received behavioral parent training and teachers were taught to establish a school-home daily report card (DRC). A DRC is a list of individual target behaviors that represent a child's most salient areas of impairment. Teachers set daily goals for each child's impairment targets, and parents provided rewards at home for goal attainment. Each weekday, teachers completed the DRC, and it was used as a dependent measure of individualized medication response. Teachers and parents also completed weekly standardized ratings of behavior and treatment effectiveness. To evaluate the time course of medication effects, children spent 12 hours in a laboratory setting on Saturdays and medication effects were measured using procedures and methods adapted from our summer treatment program. Measures of classroom behavior and academic productivity/accuracy were taken in a laboratory classroom setting during which children completed independent math and reading worksheets. Measures of social behavior were taken in structured, small-group board game settings and unstructured recess settings. Measures included behavior frequency counts, academic problems completed and accuracy, independent observations, teacher and counselor ratings, and individualized behavioral target goals. Reports of adverse events, sleep quality, and appetite were collected. Results: On virtually all measures in all settings, both drug conditions were significantly different from placebo, and the 2 drugs were not different from each other. In children's regular school settings, both medications improved behavior as measured by teacher ratings and individualized target behaviors (the DRC); these effects were seen into the evening as measured by parent ratings. In the laboratory setting, effects of Concerta were equivalent to tid MPH and lasted at least through 12 hours after dosing. Concerta was significantly superior to tid MPH on 2 parent rating scores, and when asked, more parents preferred Concerta than preferred tid IR MPH or placebo. Side effects on children's sleep and appetite were similar for the 2 preparations. In the lab setting, both medications improved productivity and accuracy on arithmetic seatwork assignments, disruptive and on-task behavior, and classroom rule following. Both medications improved children's rule following and negative behavior in small group board games, as well as in unstructured recess settings. Individual target behaviors also showed significant improvement with medication across domains in the laboratory setting. Children's behavior across settings deteriorated across the laboratory day, and the primary effect of medication was to prevent this deterioration as the day wore on. Results support the use of background behavioral treatment in clinical trials of stimulant medication, and illustrate the utility of a measure of individualized daily target goals (ie, the DRC) as an objective measure of medication response in both the laboratory and natural school settings. Conclusion: This investigation clearly supports the efficacy of the Concerta long-acting formulation of MPH for parents who desire to have medication benefits for their child throughout the day and early evening. (ABSTRACT TRUNCATED)
Article
Full-text available
Methylphenidate (MPH) is widely prescribed for adults with attention deficit hyperactivity disorder (ADHD), but data on long-term treatment and maintenance of effect are lacking. Osmotic release oral system-methylphenidate (OROS–MPH) was evaluated in a 52-wk open-label study in subjects who had previously completed a short-term placebo-controlled trial and short-term open-label extension. Efficacy was assessed using the investigator- and subject-rated Conners’ Adult ADHD Rating Scales (CAARS:O-SV and CAARS:S-S), and the Clinical Global Impression – Severity (CGI-S), Sheehan Disability Scale (SDS) and Quality of Life Enjoyment and Satisfaction Questionnaire (Q-LES-Q). Subjects completing ≥52 wk of treatment were eligible for a 4-wk randomized, placebo-controlled withdrawal phase in which loss of treatment effect was assessed using CAARS:O-SV and CGI-S. In the open-label phase (n=156), mean CAARS:O-SV score decreased from baseline by 1.9±7.8 (p<0.01), and small, statistically significant improvements from baseline were observed for CAARS:S-S, CGI-S and SDS. In the double-blind phase (OROS-MPH, n=23; placebo, n=22), CAARS:O-SV increased from double-blind baseline in the OROS-MPH and placebo arms (4.0±7.6 vs. 6.5±7.8, not statistically significant). Long-term OROS-MPH treatment was well tolerated, and there was no evidence of withdrawal or rebound after discontinuation. In conclusion, the short-term benefits of OROS-MPH continue during long-term open-label treatment. Maintenance of efficacy in a placebo-controlled withdrawal design remains to be confirmed in larger patient populations.
Article
Full-text available
Attention-deficit/hyperactivity disorder (ADHD) is a clinically important neuropsychiatric developmental disorder that affects children, adolescents, and adults. The disorder is characterized by core symptoms of inattention, hyperactivity, distractibility, impulsivity, and impaired executive functioning. It is estimated that 2% to 5% of the adult population in the United States has ADHD. Adults with ADHD are at an increased risk for experiencing comorbid psychiatric disorders, including mood disorders, anxiety disorders, and substance use disorders. The authors provide a brief clinical overview of ADHD and the treatment of adults with this disorder.
Article
Full-text available
Adults with attention-deficit/hyperactivity disorder (ADHD) are more frequently presenting for diagnosis and treatment. Medication is considered to be appropriate among available treatments for ADHD; however, the evidence supporting the use of pharmacotherapeutics for adults with ADHD remains less established. In this article, the effectiveness and dosing parameters of the various agents investigated for adult ADHD are reviewed. In adults with ADHD, short-term improvements in symptomatology have been documented through the use of stimulants and antidepressants. Studies suggest that methylphenidate and amphetamine maintained an immediate onset of action, whereas the ADHD response to the nonstimulants appeared to be delayed. At a group level, there appears to be some, albeit not entirely consistent, dose-dependent responses to amphetamine and methylphenidate. Generally speaking, variability in diagnostic criteria, dosing parameters and response rates between the various studies was considerable, and most studies were of a relatively short duration. The aggregate literature shows that the stimulants and catecholaminergic nonstimulants investigated had a clinically significant beneficial effect on treating ADHD in adults.
Article
Full-text available
The osmotic release oral system (OROS) methylphenidate formulation is a prolonged-release medication for the treatment of attention-deficit/hyperactivity disorder (ADHD) in children, adolescents, and adults. We conducted a seven-week open-label extension of a double-blind study to assess the safety and tolerability of OROS methylphenidate in a flexible dose regimen (18-90 mg daily) for the treatment of adults diagnosed with ADHD (N =370). Medication was adjusted to optimize efficacy and tolerability for each patient. Adverse events, vital signs, and laboratory parameters were assessed. Most patients (337; 91%) completed the seven-week treatment and the final dispensed dose was 18 mg (8%), 36 mg (29%), 54 mg (34%), 72 mg (20%), or 90 mg (9%). Adverse events were reported in 253 (68%) patients and most were mild or moderate in severity; most frequently reported included headache (17%), decreased appetite (13%), and insomnia (11%). Adverse events were rarely serious (<1%; 2/370). Small mean increases in systolic and diastolic blood pressure (both 2.4 mmHg) and pulse (3.2 bpm) were observed. Body weight decreased slightly (-1.5 kg). The results provide additional support for the safety and tolerability of prolonged-release OROS methylphenidate in a flexible dose regimen (18-90 mg/day) for the treatment of adults with ADHD.
Article
Full-text available
Follow-up studies of childhood ADHD have shown persistence of the disorder into adulthood, but no epidemiological data are yet available. ADHD DSM-IV symptoms were obtained by self-report in an adult population-based sample of 1813 adults (aged 18-75 years), that was drawn from an automated general practitioner system used in Nijmegen, The Netherlands. The structure of ADHD symptoms was analysed by means of confirmatory factor analyses. Other data used in this report are the General Health Questionnaire (GHQ-28), information about the presence of three core symptoms of ADHD in childhood, and about current psychosocial impairment. The three-factor model that allowed for cross-loadings provided the best fit in the entire sample. This result was replicated across gender and age subsamples. Inattentive and hyperactivity symptom scores were significantly associated with measures of impairment, even after controlling for the GHQ-28. Subjects with four or more inattentive or hyperactive-impulsive symptoms were significantly more impaired than subjects with two, one and no symptoms. The prevalence of ADHD in adults was 1.0% (95% CI 0.6-1.6) and 2.5% (1.9-3.4) using a cutoff of six and four current symptoms respectively, and requiring the presence of all three core symptoms in childhood. These results support the internal and external validity of ADHD in adults between 18 and 75 years. ADHD is not merely a child psychiatric disorder that persists into young adulthood, but an important and unique manifestation of psychopathology across the lifespan.
Article
Objective: Assessing the degree of problems related to drug abuse is important in each treatment setting. The Drug Abuse Screening Test-20 (DAST-20) is a brief, simple 20-item instrument to measure the degree of problems related to drug use. The objective of the present study is to examine the reliability and validity of the Japanese version of the DAST-20. Methods: We translated the DAST-20 into Japanese using back translation. The anonymous self-administered questionnaire was completed by 310 drug users at the Drug Addiction Rehabilitation Centers (DARC group, n = 113) and at HIV/AIDS regional hospitals (HIV group, n = 197) in Japan. Results: The average DAST-20 score was 7.6 (DARC group = 14.7, HIV group = 2.8). Each item score was highly correlated with the total score (r = 0.45-0.88). A high internal consistency (Cronbach's α = 0.95) was observed (men = 0.95, women = 0.84). Overall test-retest reliability was 0.86 (men = 0.85, women = 0.90). The total DAST-20 score was strongly positively correlated with the Severity of Dependence Scale-J score (r = 0.85), but moderately positively correlated with the Alcohol Use Disorders Identification Test score (r = 0.41). In addition, confirmatory factor analysis indicated an acceptable fit to the data (goodness-of-fit index [GFI] = 0.893, adjusted goodness-of-fit index [AGFI] = 0.854, comparative fit index [CFI] = 0.948, root mean square residual [RMR] = 0.008, root mean square error of approximation [RMSEA] = 0.073). Conclusion: Our results clearly suggest that the Japanese version of the DAST-20 has sufficient internal consistency and acceptable levels of concurrent validity and construct validity.
Article
In Japan, treatment with medication for attention-deficit/hyperactivity disorder (ADHD) is less favorable than psychosocial treatment, as in Europe. Overall, Japanese guidelines offer few descriptions of stimulant-refractory or complicated ADHD treatments. However, carbamazepine has been preferably prescribed for ADHD with electroencephalogram abnormality or aggression. Only one stimulant (long-acting methylphenidate) is now available in Japan. Under the current circumstances, Japanese psychiatrists and pediatricians are trying to make the most of available psychosocial treatment and medication, including formulation of the guidelines. Epidemiologic studies on the prevalence, comorbidity, and course of ADHD should also be administered in Japan. This article describes the history of psychopharmacology for minimal brain dysfunction or ADHD and introduces recently established guidelines for ADHD treatment in Japan compared with those in other countries.
Article
The main purpose of this first atomoxetine study in Japanese adults with attention-deficit/hyperactivity disorder (ADHD) was to investigate the tolerability of an 8-week treatment regimen. This was an open-label, dose escalation study conducted in 45 Japanese patients aged at least 18 years with DSM-IV-defined ADHD. Patients received atomoxetine orally for 8 weeks. Atomoxetine administration was started at 40 mg/day (7 days), and subsequently increased to a maximum dose of 120 mg/day. Tolerability was assessed by discontinuation rate due to adverse events. Adverse events, laboratory tests, vital signs and electrocardiograms were collected. In addition, ADHD symptoms were assessed by using the Japanese version of the Conners' Adult ADHD Rating Scale-Investigator Rated: Screening Version (CAARS-Inv:SV) scores. Thirty-nine patients completed the study period. Atomoxetine was well tolerated with a 6.7% (3/45) discontinuation rate due to nausea, malaise and anorexia. The most commonly reported adverse events were nausea, nasopharyngitis and headache; there were no unexpected safety concerns. No deaths or serious adverse events were reported. Mean CAARS-Inv:SV-J total ADHD symptom scores decreased in a time-dependent manner; the mean change from baseline to endpoint was -15.0 (P<0.001). This study showed that atomoxetine was well tolerated in these patients and suggested that atomoxetine at a maximum dose of 120 mg/day would be safe in Japanese ADHD patients.