You are on page 1of 12

Neuropsychol Rev (2007) 17:61–72

DOI 10.1007/s11065-006-9017-3

ORIGINAL PAPER

Pharmacologic Treatment of Attention-Deficit/Hyperactivity


Disorder: Efficacy, Safety and Mechanisms of Action
Steven R. Pliszka

Received: 30 November 2006 / Accepted: 30 November 2006 / Published online: 23 January 2007

C Springer Science+Business Media, LLC 2007

Abstract Studies examining the efficacy, safety and mech- Introduction


anisms of action of agents for the treatment of attention-
deficit/hyperactivity disorder (ADHD) are reviewed, with an The pharmacological treatment of attention-deficit/hyper-
emphasis on newer agents such as the long acting stimulants activity disorder (ADHD) is one of the most thoroughly
and atomoxetine. Recent studies of medications are charac- researched topics in the mental health arena. The use of
terized by large, rigorously diagnosed samples of children, amphetamine in children with disruptive behavior disorders
adolescents and adults with ADHD, use of standardized rat- was first described by Bradley (1937) seven decades ago,
ing scales and extensive safety data. These studies confirm thus the treatment of ADHD (albeit referred to by many dif-
a robust treatment effect for the Food and Drug Adminis- ferent names) predates the use of antibiotics. The negative
tration approved agents ranging from 0.7 to 1.5. The most impact of symptoms of ADHD on many different aspects of
common short term side effects to the most commonly used patient and family life is well established (Barkley, 2006);
agents include insomnia, loss of appetite, and headaches. fortunately, a wide array of pharmacological agents is now
Despite public controversy and labeling changes to warn of available for the treatment of ADHD. Advances in functional
extremely rare cardiovascular and psychiatric side effects, magnetic resonance imaging (fMRI), electroencephalogram
the evidence does not support the hypothesis that medication (EEG) techniques, event related potentials (ERP), and ra-
for ADHD increases risk for sudden death, mania or psy- dionucleotide imaging have begun to examine the mecha-
chosis. A wide variety of neuroimaging techniques including nisms of action of these agents, particularly the stimulants.
electrocephalogram (EEG) power, event related potentials This article will examine the efficacy and safety of phar-
(ERP), functional magnetic resonance imaging (fMRI), and macological agents for ADHD, with an emphasis on the
positron emission tomography (PET) are beginning to exam- more recently developed medications. Recent neuroimaging
ine the mechanisms of action of medications for ADHD, and studies examining the action of methylphenidate in normal
implicating the catecholamines and prefrontal and anterior volunteers and patients with ADHD will then be reviewed.
cingulate cortices as prime sites of actions for these agents.
Stimulants
Keywords ADHD . Psychopharmacology . Magnetic
resonance imaging . PET Stimulants have consistently shown robust behavioral ef-
ficacy in hundreds of randomized controlled trials (RCT)
conducted since the 1960’s. By 1993, Swanson’s “Review
of reviews” reported over 3,000 citations and 250 reviews
of stimulant treatment (Swanson et al., 1993). Robust short-
S. R. Pliszka () term stimulant-related improvements in ADHD symptoms
Division of Child and Adolescent Psychiatry, were found in 161 studies encompassing 5 preschool, 140
Department of Psychiatry,
7703 Floyd Curl Drive MC 7792,
school age, 7 adolescent and 9 adult RCTs (Spencer et al.,
San Antonio, TX 78229-3900, USA 1996). Improvement was noted for 65–75% of the 5,899
e-mail: pliszka@uthscsa.edu patients assigned to stimulant treatment versus only 4–30%

Springer
62 Neuropsychol Rev (2007) 17:61–72

of those assigned to placebo for methylphenidate (MPH) also reviewed post-marketing safety data for stimulants for
(n = 133 trials), dextroamphetamine (DEX) (n = 22 tri- reports of mania/psychotic symptoms, aggression, and suici-
als), and pemoline (n = 6 trials). This body of data has dality (Gelperin, 2006). Such reports have many limitations,
continued to grow since then with the introduction of the as information about dosage, comorbid diagnoses, and con-
short acting mixed salts of amphetamine (MSA) (Pliszka, comitant medications is often not available. Nonetheless, for
Browne, Olvera, & Wynne, 2000). MPH has both a dextro each of the stimulants, there occurred very rare events of
(d) and levo (l) isomer; the d-MPH isomer alone shows effi- psychotic symptoms, specifically involving visual and tac-
cacy equal to that of the d, 1-MPH form, with some evidence tile hallucinations of insects. Symptoms of aggression and
that the d-isomer administered alone has a slightly longer suicidality (but no completed suicides) were also reported.
duration of action than d,l.-MPH (Weiss, Wasdell, & Patin, The FDA ordered changes to stimulant medication labeling
2004). Short acting stimulants rarely have a duration of ac- to more prominently discuss these risks. While such labeling
tion longer than six hours, requiring multiple doses per day. changes encourage families and clinicians to more closely
In recent years, long acting forms have been developed for monitor patients for these rare events, the new label does not
d,l MPH (Concerta, Daytrana Transdermal System, Metadate require a change in the clinical use of stimulant medications.
CD, Ritalin LA), MSA (Adderall XR) and d-MPH (Focalin Whether stimulants affect the growth of children with
XR). Studies documenting the efficacy of these agents rela- ADHD has been controversial for many years. Recently, two
tive to placebo are listed in Table 1. All of these studies are major reviews (Poulton, 2005; Faraone, Biederman, Morley,
characterized by large samples, rigorous diagnostic criteria & Spencer, 2006) examined all of the available data and con-
for ADHD, use of standardized rating scales for assessing cluded that stimulant treatment may be associated with a re-
symptoms of ADHD, and double blind, controlled designs. duction in expected height gain, at least in the first 1–3 years
Results showed long acting agents to have a response rate of treatment. The Multi-modality Treatment of ADHD study
similar to that shown by short acting stimulants in the earlier showed reduced growth in ADHD patients after 2 years of
studies. stimulant treatment compared with those patients who re-
ceived no medication (MTA Cooperative Group, 2004) and
Side effects of stimulants these differences persisted at 36 months (MTA Coopera-
tive Group, 2006). Interestingly, the children with ADHD in
As noted in Table 1, the most common stimulant side ef- the MTA study were taller and heavier than average at the
fects are headache, insomnia and loss of appetite. Despite start of the study; those treated continuously with medication
many years of use, there continues to be some controversy tended to approach the mean of the population for height and
regarding stimulant safety. Many of the short term studies in weight by year three of follow up. Charach et al. (2006) found
Table 1 were followed by longer term open label studies higher doses of stimulant correlated with reduced gains in
lasting 1–2 years (Wilens et al., 2005; McGough et al., height and weight. The effect did not become significant until
2005). No serious adverse events were reported and lab- the dose in MPH equivalents was >2.5 mg/kg/day for four
oratory and electrocardiogram (ECG) findings were unre- years. In a review by Poulton (2005) concern was raised that
markable (Findling et al., 2005). In 2005, Health Canada children with ADHD treated with stimulants might show a
briefly suspended the sale of Adderall XR due to concerns reduced height gain of about 1 cm/year and more long term
regarding a small number of cases of sudden death in both studies were recommended. However, two recent 1–3 years
children and adolescents. A review by the U.S. Food and follow up studies did not show an impact on growth (Pliszka,
Drug Administration (FDA) Villalaba (2006) estimated the Matthews, Braslow, & Watson, 2006c; Spencer et al., 2006a).
rate of sudden death in stimulant-treated ADHD children for Children with ADHD treated with stimulants should have
the exposure period January 1, 1992 to December 31, 2004 height and weight measured at least semi-annually. Children
to be 0.2/100,000 patient years for MPH, and 0.3/100,000 whose height or weight percentile drops across two major
patient-years for amphetamine (the differences between the lines (5th, 10th, 25th, 50th, 75th, 90th, and 95th) should be
agents not clinically meaningful). Thus the sudden death considered for treatment with a non stimulant. Drug holidays
rate for children on ADHD medications does not exceed appear to eliminate any growth effects of stimulants (Pliszka
the base rate of sudden death in the general population. No et al., 2006c) and should be considered if the child’s ADHD
evidence currently indicates a need for routine cardiac eval- symptoms do not cause severe impairment on weekends and
uation (i.e., ECG) before starting stimulant treatment in oth- during the summer.
erwise healthy individuals. However, these findings suggest
that ADHD medications should be used cautiously in chil- Atomoxetine
dren and adolescents with pre-existing heart disease. These
cases should be referred for a pediatric cardiology consul- Atomoxetine is a noradrenergic reuptake inhibitor that is
tation for possible EKG and/or echocardiogram. The FDA superior to placebo in the treatment of ADHD in children,

Springer
Table 1 Summary of recent studies of the use of stimulants in the treatment of ADHD

Generic name Brand N, age group Design Efficacy data Safety Author

MSA Adderall XR 509 children, aged 6–12 4 week, DBPC parallel AM and PM teacher and behavior Decreased appetite-22% (Biederman, Lopez,
years groups rating scales improved relative to Headache-14% Boellner, & Chandler, 2002)
placebo Insomina-16%
Abdominal pain-14%
Moodiness-9%
OROS-MPH Concerta 282 children aged 6–12 MPH responders Response rates: OROS MPH: 47% Decreased appetite, headache and (Wolraich et al., 2001)
years randomized to placebo MPH-47% insomnia
Neuropsychol Rev (2007) 17:61–72

MPH or OPOS MPH Placebo-17%


MPH Metadate 321 children aged 6–16 3 week DBPC parallel Response rates: MPH-64% Headache-15% (Greenhill, Findling, &
CD years groups Placebo-27% Decreased appetite-10% Swanson, 2002)
Abdominal pain-10%
Insomnia-7%
MPH Ritalin LA 134 children aged 6–12 2 week DBPC parallel Teacher ratings significantly Decreased appetite and insomnia Package Insert
groups improved over placebo
d-MPH Focalin XR 97 children aged 6–17 7 week, DBPC parallel Response rates: d-MPH-67% (Greenhill et al., 2006b)
years groups Placebo-13%
OROS-MPH Concerta 141 adults 6 weeks, DBPC parallel 66% response to MPH, 39% to Small but clinically insignificant (Biederman et al., 2006)
groups placebo effects on blood pressure
MSA Adderall XR 278 adolescents aged 4 weeks, DBPC parallel Response rates: Placebo- 27% Decreased appetite- 36% (Spencer et al., 2006b)
10–17 years groups 10 mg-52% Headache-16%
20 mg 66% Weight loss-9%
30 mg 71% Insomnia-12;
40 mg 64%
MSA Adderall XR 223 adults 4 week, DBPC parallel Significant improvement of ADHD Insomnia-33% (Biederman et al., 2005;
group, placebo vs 4 symptoms relative to baseline Decreased appetite-32% Weisler et al., 2006)
doses of MSA Headache-30%
Nervousness-26%
ORPS MPH Concerta 220 adolescents Open titration to Response rates: OROS MPH-52% Headache-7% (Wilens et al., 2006)
efficacious dose of Placebo-31% Decreased appetite-2%
OROS MPH, then 37% of subjects required 72 mg of Insomnia-4% (Only MPH
randomized for two OROS MPH a day responders in study)
weeks to OROS MPH or
placebo
MPH Daytrana 270 children aged 6–12 5-week DBPC parallel Response rate: MPH MTS-72% Decreased appetite-25% (Findling & Lopez, 2005)
transdermal groups, 4 dose levels of Placebo-24% Insomnia 10%
system MTS Nausea-10%
(MTS) Decreased weight-10%
Tic-5%
Moodiness-5%

Springer
63
64 Neuropsychol Rev (2007) 17:61–72

adolescents, and adults (Michelson et al., 2003; Michelson ommended stages of pharmacologic treatment for ADHD.
et al., 2001; Michelson et al., 2002; Swensen, Michelsen, The algorithm recommends stimulants as the first stage of
Buesching, & Faries, 2001). Given its pharmacokinetic half- ADHD treatment due to their large effect size and superiority
life of 5 hours, it is generally dosed twice a day. Atomoxetine to non-stimulants in comparative trials (Biederman, Wigal,
may also reduce tics (Allen et al., 2005) and be effective in Spencer, McGough, & Mays, 2006; Wigal et al., 2005). It is
children with ADHD who have comorbid anxiety (Sumner also recommended that if one stimulant class (MPH or am-
et al., 2005). The FDA has issued warnings regarding rare phetamine) fails, then the alternative class should be tried.
side effects of hepatotoxicity and suicidal ideation (Food and Atomoxetine may also be combined in low doses with a
Drug Administration, 2005). stimulant. Clinical experience suggests this is helpful for
symptoms late in the day or at bedtime when administering a
Modafinil stimulant may induce insomnia. Buproprion or a tricyclic an-
tidepressant should be tried if the FDA-approved agents are
Modafinil showed efficacy in the treatment of ADHD in unsatisfactory, and alpha-agonists should be used for ADHD
two double-blind, placebo-controlled trials (Greenhill et al., alone only as medication of last resort.
2006a; Swanson et al., 2006) but the FDA declined to ap-
prove it for clinical use. There was one case of suspected Treatment of comorbidity
Stevens Johnson syndrome among the hundreds of children
who participated in the clinical trials, so the sponsor declined The Texas CMAP algorithm discusses the pharmacologic
to pursue further development of modafinil as a treatment for treatment of ADHD with major depressive disorder (MDD),
ADHD. anxiety disorder, tics disorder, and severe aggression (Pliszka
et al., 2006a). Treatment of comorbid MDD is complicated
Other agents by the association of antidepressant treatment with increased
suicidal ideation (4% on antidepressant vs. 2% on placebo)
A number of agents have historically been used off-label for in children and adolescents with depression (Hammad,
the treatment of ADHD. Controlled studies have shown that Laughren, & Racoosin, 2006). CMAP recommends that the
tricyclic antidepressants are superior to placebo in ADHD patient with both ADHD and MDD be assessed for the sever-
treatment, although the effect size is less than that of stimu- ity of the two disorders, and that the most severe disorder be
lants (Daly & Wilens, 1998). Bupropion is an antidepressant treated first. Thus, a child whose ADHD is causing greater
with effects primarily on the norepinephrine and dopamine impairment should be treated via the ADHD CMAP algo-
systems that also shows modest efficacy in the treatment of rithm. If the ADHD and MDD symptoms simultaneously
ADHD (Conners et al., 1996). These antidepressant med- resolve with ADHD treatment, no further pharmacologic
ications are used much less frequently today because of intervention is needed. If the MDD symptoms persist
the availability of atomoxetine and multiple stimulant for- despite resolution of the ADHD symptoms, then treatment
mulations. Nonetheless, they remain reasonable second-line of depression should be added; either an antidepressant or
agents. The alpha-agonist clonidine has long been used for a psychosocial intervention. In contrast, if the MDD symp-
treating ADHD. Connor, Fletcher, & Swanson (1999) per- toms are more severe (pervasive depression, weight loss,
formed a meta-analysis of 11 studies which suggested cloni- loss of functioning) than the ADHD symptoms, treatment
dine possessed efficacy in the treatment of ADHD, although for MDD should be initiated (usually a serotonin reuptake
no definitive studies have been performed. The closely re- inhibitor). If the ADHD symptoms persist after remission
lated alpha-2A agonist guanfacine was superior to placebo of the depression, than an ADHD treatment should be
in reducing ADHD symptoms in children with comorbid added.
ADHD and tics (Scahill et al., 2001), although it did not Since atomoxetine has been shown to be effective in the
improve executive functioning in healthy adult volunteers treatment of both anxiety and ADHD (Sumner et al., 2005), it
(Muller et al., 2005). The most common current use for the may be considered for first line use for children with ADHD
alpha-agonists is in combination with stimulants to optimally and comorbid anxiety. Serotonin reuptake inhibitors are com-
treat ADHD symptoms and tics, in those with this comor- monly added to stimulants for this subgroup of patients, al-
bidity (Tourette’s Syndrome Study Group, 2002). though data showing the efficacy of this approach are lacking
(Abikoff et al., 2005).
Medication algorithm for ADHD Children with tic disorders can be treated safely with
stimulants without increasing tics (Gadow, Sverd, Sprafkin,
The Texas Children’s Medication Algorithm Project Nolan, & Grossman, 1999) and the CMAP algorithm sug-
(CMAP) has recently updated its guidelines for ADHD gests beginning at Stage 1 with stimulant medications for
treatment (Pliszka et al., 2006a). Fig. 1 lays out the rec- this subgroup. If stimulants worsen tics, then the clinician

Springer
Neuropsychol Rev (2007) 17:61–72 65

Diagnostic Assessment and


Stage 0 Family Consultation Regarding
Treatment Alternatives
Any stage(s) can be skipped Non-Medication
depending on the clinical picture. Treatment Alternatives

Stage 1 Methylphenidate or Amphetamine

Response

Partial Stage 1A
Response (Optional)
(if MSA or AMP formulation Response
Continuation
DEX used not used in Stage 1
in Stage 1)
Partial Response
or Nonresponse Partial Response
or Nonresponse
Stage 2 Stimulant not used in Stage 1

Response

Partial Stage 2A
Response (Optional)
(if MSA or Response
AMP formulation
Continuation
DEX used not used in Stage 2
in Stage 2)
Partial Response
or Nonresponse Partial Response
or Nonresponse
Stage 3 Atomoxetine

Response

Stage 3A
Partial (Optional)
Response Response
Combine stimulant
Continuation
to stimulant and atomoxetine
or atomoxetine
Partial Response
or Nonresponse Partial Response
or Nonresponse
Stage 4 Bupropion or TCA

Response
Continuation
Partial Response
or Nonresponse

Stage 5 Agent not used in Stage 4

Response
Continuation
Partial Response
or Nonresponse

Stage 6 Alpha Agonist


AMP = Amphetamine
DEX = Dextroamphetamine
Clinical MSA = Mixed salts amphetamine Maintenance
Consultation TCA = Tricyclic antidepressant
Fig. 1 Algorithm for the psychopharmacological treatment of ADHD

should utilize a non-stimulant medication such as atomox- to reduce both classes of symptoms (Tourette’s Syndrome
etine (Allen et al., 2005). Some children with ADHD and Study Group, 2002).
comorbid tics respond only to stimulants in terms of their Severe aggressive behavior occurs in substantial num-
ADHD, yet the stimulant worsens their tics. In such cases, bers of children with ADHD, particularly in those with co-
alpha-agonist medication should be added to the stimulant morbid oppositional defiant, conduct disorder, or bipolar

Springer
66 Neuropsychol Rev (2007) 17:61–72

disorder (Jensen et al., 2005). Stimulant or atomoxetine acutely increases the amount of dopamine in the synap-
treatment of children with ADHD often reduces comorbid tic cleft. Next, Volkow et al. (2004) examined the effect
oppositional or aggressive behavior (Connor, Glatt, Lopez, of MPH on dopamine release in two different situations:
Jackson, & Melloni, 2002; Newcorn, Spencer, Biederman, while subjects passively viewed pictures and again while
Milton, & Michelson, 2005). Thus, aggressive and conduct performing a difficult mathematics task. Relative to placebo,
disorders symptoms will resolve in the comorbid child when MPH produced much greater reduction in raclopride bind-
the ADHD is robustly treated (Klein et al., 1997). If treat- ing (dopamine release was increased) during the mathemat-
ment of ADHD is not sufficient to eliminate the aggressive ics tasks than in the passive condition. Thus it appears that
behavior, then a behavioral treatment should be added to the the dopamine release is associated with tasks requiring ex-
ADHD treatment. It is prudent to address any psychosocial ecutive functions or response to a stimuli signaling reward.
or parent management problems contributing to the aggres- MPH can block reuptake of dopamine only when it is being
sion (Pappadopulos et al., 2003; Schur et al., 2003). If the released in significant amounts. This may explain why the ef-
aggressive behavior is not resolved with such an approach, fects of MPH (and other stimulants) are most pronounced in
and the aggressive behavior is dangerous to the patient or the classroom or other structured settings where cognitively
others, than a second generation antipsychotic should be stressful activities occur. Recently, Rosa-Neto et al. (2005)
added to the stimulant (Pliszka et al., 2006a). The efficacy of performed a PET raclopride study with nine adolescents with
second generation antipsychotic monotherapy in treating ag- ADHD; the subjects were scanned both on placebo and again
gression is increasingly well established by controlled trials on a therapeutic dose of MPH. MPH induced a decrease in
(Snyder et al., 2002). A recent study by Aman et al. (2004) raclopride binding relative to placebo. The magnitude of the
randomized a group of aggressive children with ADHD to decrease in raclopride binding correlated well with MPH-
risperidone or placebo; one-half of these children were also induced improvements on cognitive testing.
already being treated with a stimulant for their ADHD and Another approach that is becoming more fruitful in as-
remained on it during the double-blind trial testing risperi- sessing stimulant action in the brain is the direct study of the
done or placebo. No significant increases in side effects were binding of ligands to the dopamine transporter (Spencer et al.,
seen in the combined risperidone-stimulant group, although 2005). Several ligands, including 123I-Altropane, 123I-IPT
both groups had similar reductions in aggression. Second and 99Tc-TRODAT-1, and 123I-citalopram are available
generation antipsychotics should be used with caution, how- which bind to the dopamine transporter and give a measure
ever, as children exposed to them chronically are at long of its binding potential. Typically, Single Positron Emission
term risk of weight gain, diabetes, and metabolic syndrome Tomography (SPECT) is used to assess the amount of lig-
(Correll & Carlson, 2006). and binding to the receptor, although Altropane can be use
with PET (Spencer et al., 2005). The PET ligand PE2I can
Neurobiological mechanisms of action of ADHD treatments also be used to image the dopamine transporter (Jucaite,
Fernell, Halldin, Forssberg, & Farde, 2005). In seven studies
Radio-labeled ligands can be used to study cerebral blood reviewed by Spencer et al. (2005), five showed the dopamine
flow, or they can be designed to bind to areas of interest transporter binding to be increased in subjects with ADHD
in the brain, particularly the dopamine transporter (Spencer relative to controls, while two studies showed no difference
et al., 2005) or dopamine receptors (Volkow, Wang, Fowler, between the ADHD and control groups. Some early stud-
& Ding, 2005). Methylphenidate itself can be labeled with ies have begun to examine the effect of MPH on dopamine
carbon-11 to determine its distribution in the brain (Volkow transporter binding.
et al., 2005). A typical therapeutic dose of 0.5 mg/kg of Krause et al. (2003) obtained SPECT using TRPDAT-1 in
MPH will lead to the blockade of about 60% of the dopamine a small number of adults with ADHD both at baseline and
transporters in the striatum (Volkow et al., 1998). Another again after a dose of MPH. They found the dopamine trans-
approach to examine the effects of MPH in the brain is to porter binding potential to be reduced with treatment, but
image subjects with positron emission tomography (PET) the SPECT was done 90 minutes after the MPH dose when
using 11C-raclopride, which binds to dopamine-2 receptors. the medication is expected to be binding to the transporter.
Volkow et al. (2001) performed PET using this ligand in Thus it cannot be concluded that MPH down regulated the
healthy men on both placebo and 60 mg of MPH. On MPH, transporter (Spencer et al., 2005). Vles et al. (2003) also
more dopamine was released, leading to a decline in raclo- found that dopamine transporter binding potential was re-
pride binding relative to placebo. MPH (and amphetamine) duced after three months of MPH treatment in six boys with
also have affinity for the norepinephrine transporter as well, ADHD, but the timing of SPECT in relation to the last MPH
but since the raclopride does not bind to it, studies of this dose was not reported. When MPH treatment was withdrawn
sort cannot elucidate the effect of MPH on norepinephrine. from five children with ADHD, dopamine transporter bind-
Nonetheless, Volkow et al. (2001) established that MPH ing potential immediately increased to pretreatment values,

Springer
Neuropsychol Rev (2007) 17:61–72 67

suggesting that MPH does not induce any long term down treatment of ADHD can increase these ratios, “normaliz-
regulation of the transporter (Feron et al., 2005). ing” the EEG (Clarke, Barry, Bond, McCarthy, & Selikowitz,
Cerebral blow flood can be assessed by either SPECT 2002). This would also suggest that stimulants might act by
(2001) or PET (Matochik et al., 1993; Matochik et al., increasing cortical arousal, consistent with the blood flow
1994) but this technique has not been as fruitful as might be studies of Kim et al. (2001) noted above. Clarke et al. (2002)
expected, either as a diagnostic tool or to assess stimulant compared 20 children with ADHD who responded well to
effects in the brain. In the largest study to date, Kim et al. MPH to 20 children who had responded well to dextroam-
(2001) assessed 32 boys with ADHD using 99 Tc HMPAO phetamine using EEG power. Theta/beta ratios in the frontal,
SPECT at baseline and after a trial of MPH. The subjects central, and posterior leads were higher for the MPH respon-
were rigorously screened to insure the absence of psychiatric ders than the dextroamphetamine responders, MPH respon-
comorbidity or learning disabilities. Significant cerebral ders had greater theta/alpha ratios in the frontal leads. Loo
blow flow increases after MPH were found in bilateral et al. (2004), found similar results, adding that MPH non
prefrontal cortex, caudate, and thalamus of subjects who responders tended to show decreased beta while on medi-
were MPH responders. Unfortunately, studies performed cation. Increased beta also correlated with improvements in
since then have not had the power to allow firm conclusions ADHD symptoms on parent behavior rating scales. It is im-
to be made about the effects of MPH on cerebral blood portant to note, however, that these results represent group
flow. Ten adult subjects had cerebral blood flow assessed by averages. At the level of the individual patient, EEG does
PET before and after a three week MPH trial. MPH induced not predict stimulant response above the rate predicted by
increases in blood flow in cerebellar vermis, but decreases the clinical diagnostic information (Loo and Barkley, 2005).
in precentral gyri, left caudate nucleus, and right claustrum Nonetheless, while not yet appropriate for standard clinical
(Schweitzer et al., 2003). In contrast, Szobot et al. (2003) practice, examination of EEG power may help elucidate pat-
found decreased left parietal blood flow on MPH relative to terns of medication response that are not yet distinguishable
placebo in a relatively large (n = 36) study of children and on clinical measures. Loo et al. (2003) compared the MPH
adolescents with ADHD; the same group also found that response of children with ADHD who were homozygous for
MPH withdrawal led to increases in blood flow as assessed the 10-repeat allele of the dopamine transporter with those
by SPECT in the motor, premotor, and anterior cingulate who were heterozygous for the 9-repeat allele. The 10-R
cortex (Langleben et al., 2002). Reviewing these data, homozygotes showed increased beta power and increased
Castellanos (2002) questioned the utility of SPECT/PET beta/theta ratios on MPH relative to placebo, while children
cerebral blood flow studies, suggesting that fMRI and with ADHD who were 9-repeat allele carriers showed the
assessment of catecholamine transporter would be more opposite pattern. In the future, use of EEG in conjunction
likely to lead to further understanding of the mechanisms with genetic and functional MRI may allow a more compre-
of pharmacological treatments for ADHD. In view of hensive picture of the neurophysiology of stimulant effects.
this, the American Psychiatric Association recommended In ERP, the subject performs a repetitive cognitive task and
against the use of SPECT for the diagnosis or monitoring EEG is obtained during each trial. The EEG is then averaged
of treatment for ADHD or other psychiatric disorders over many trials, canceling out random brain activity and
(http://www.psych.org/psych pract/clin issues/populations/ producing a waveform which represents the brain’s response
children/SPECT.pdf). to each class of stimuli in the task. In studies of ADHD,
EEG and ERP measures have also been used to study oddball auditory ERP tasks and inhibitory tasks such as the
the response of children with ADHD to medication. In continuous performance test (CPT) have been most utilized
this line of work, EEG activity is examined within specific (Barry, Johnstone, & Clarke, 2003). In the auditory oddball
frequency bands. The complex EEG waveform is decom- task, the subject must detect rare tones among a long string
posed via Fournier analysis into component frequency bands: of common tones. Healthy controls produce a larger P300
delta (<4 Hz), theta (4–7 Hz), alpha (8–12 Hz) and beta wave to the oddball tones than to the common tones, but
(>13 Hz). The beta band is associated with mental alert- this difference is markedly reduced in children with ADHD
ness; most studies have shown that children with ADHD (Barry et al., 2003). The meaning of this difference in the
show decreased beta and increased amounts of the other fre- P300 is debated. The P300, which in these tasks is most
quencies, particularly in the frontal area (Barry, Clarke, & prominent over the parietal areas, possibly reflects activity
Johnstone, 2003; Loo & Barkley, 2005). While there is a related to evaluation of the stimuli but may also represent
subgroup of children who show an excess of beta (Clarke, the amount of mental capacity that is invested in the task
Barry, McCarthy, & Selikowitz, 2001), they do not appear (Kok, 1997). In most treatment studies of oddball tasks using
to be clinically distinguishable from those with decreased ERP, MPH enhances the P300 response to the rare stimuli
beta. Children with ADHD also show decreased theta/beta (Klorman, 1991). Thus it is tempting to conclude that MPH
and theta/alpha ratios relative to controls; stimulant increases the allocation of mental resources to a task, but

Springer
68 Neuropsychol Rev (2007) 17:61–72

further studies have not borne this out. Jonkman et al. (2000) Functional MRI (fMRI), which is non invasive and
obtained ERP on healthy controls and children with ADHD involves no nuclear radiation, is an expanding research tool.
while they performed a decision task, which was presented Using primarily measures of inhibitory control, comparisons
in both easy and hard conditions. Irrelevant probes appeared of ADHD children to controls during fMRI have shown
at various times in the task. Consistent with other studies, differences in fronto-striatal activity (Bush, Valera, &
children with ADHD had a reduced P300, particularly in the Seidman, 2005), particularly in the right prefrontal cortex
hard condition. In the easy condition, the P300 to the probe (Rubia, Smith, Brammer, Toone, & Taylor, 2005) and
was higher in both groups relative to the hard condition. anterior cingulate (Bush et al., 1999; Pliszka et al., 2006b).
This is because in the hard condition, attentional capacity Studies of stimulant effects on the brain using fMRI have
must be reallocated from the probes to the main task. Since been limited to date, but are increasing. Initially, Vaidya
the groups did not differ on the P300 to the probe, this et al. (1998) showed that MPH, relative to placebo, increased
indicated that the groups were not different in total attentional frontal activation in both children with ADHD and controls.
capacity. Since the P300 was smaller to the difficult task Striatal activation was increased on MPH relative to placebo
stimuli in the ADHD children, it was concluded that they in children with ADHD but controls showed the opposite
did not allocate those attentional resources as efficiently as pattern, even though both groups improved in cognitive task
controls. ADHD subjects then performed the task on placebo performance. In a small heterogeneous group of adolescents
and again on MPH. Relative to placebo, MPH increased P300 with ADHD and reading disorders, MPH increased activity
to task stimuli in both the easy and hard conditions, and did in the left ventral basal ganglia relative to placebo, but had
not affect P300 to the probe stimuli. The lack of an effect on no effect on task performance (Shafritz, Marchione, Gore,
the probe means that MPH did not increase overall attentional Shaywitz, & Shaywitz, 2004). Adults with ADHD showed
capacity, but the equal effect of MPH on P300 in both the increased activation of the anterior cingulate on MPH
easy and hard tasks also means that the stimulant was not relative to placebo while performing a difficult interference
excreting its effect by changing the allocation of resources. task (Bush, 2005). Larger scales treatment studies with
Thus the effect of MPH on P300 in this type of task is unclear. children, adolescents, and adults remain to be done.
Since ADHD (particularly the combined type) has been
conceptualized as a disorder of inhibitory control (Barkley, Future directions
1997), more clear results have emerged from studies us-
ing tasks assessing this cognitive domain. On the CPT, the Today there is a wide array of effective agents for the treat-
child must respond to target stimuli and avoid responding ment of ADHD. Stimulants are the most effective agents
to other stimuli, in the Go/No Go task the child responds available, but atomoxetine is a valuable agent, particularly
to a Go stimuli the majority of the time but must refrain for those who do not respond to stimulants or who have co-
from responding when the No Go stimuli are presented. morbid anxiety or depressive disorders. Older agents such
No-Go or stop signals on an inhibitory task are associated as bupropion and tricyclic antidepressants now have a lesser
with a right lateralized N200 which may signal the trig- role, although alpha agonists have a valuable role in treat-
gering of the prefrontal inhibitory processes (Kok, 1986); ing comorbid tics. Neuroimaging studies will soon begin
this N200 has been shown to be reduced in children with to unlock the mechanisms of actions of these agents and
ADHD (Pliszka, Liotti, & Woldorff, 2000). When on MPH, genetic studies may identify subtypes that are clinically in-
the N200 to No Go stimuli of children with ADHD no longer distinguishable but respond preferentially to certain agents.
showed any differences from controls (Broyd et al., 2005). In general, understanding of the pathophysiology of ADHD
No Go stimuli of inhibitory tasks also elicit a frontocentral will hopefully lead to the development of new and more
P300 (to be distinguished from the parietal P300 discussed effective agents.
above) which is thought to be generated by the anterior cin-
gulate (Schmajuk, Liotti, Busse, & Woldorff, 2006). Seven-
teen boys with ADHD performed the CPT while ERP was
References
obtained at baseline and then one week later after a 10 mg
dose of MPH (Seifert, Scheuerpflug, Zillessen, Fallgatter, & Abikoff, H., McGough, J., Vitiello, B., McCracken, J., Davies, M.,
Warnke, 2003). A group of healthy controls was also stud- Walkup, J., et al. (2005). Sequential pharmacotherapy for children
ied. P300 to the No Go stimuli was greater than that to with comorbid attention-deficit/hyperactivity and anxiety disor-
the Go stimuli, the No Go P300 of children with ADHD ders. Journal of the American Academy of Child and Adolescent
Psychiatry, 44, 418–427.
was significantly increased after treatment with MPH. Thus Allen, A. J., Kurlan, R. M., Gilbert, D. L., Coffey, B. J., Linder, S. L.,
there is some evidence that MPH improves the functioning Lewis, D. W., et al. (2005). Atomoxetine treatment in children and
of prefrontal and anterior cingulate mechanisms involved in adolescents with ADHD and comorbid tic disorders. Neurology,
inhibition. 65, 1941–1949.

Springer
Neuropsychol Rev (2007) 17:61–72 69

Aman, M. G., Binder, C., & Turgay, A. (2004). Risperidone effects Clarke, A. R., Barry, R. J., McCarthy, R., & Selikowitz, M. (2001).
in the presence/absence of psychostimulant medicine in children Electroencephalogram differences in two subtypes of attention-
with ADHD, other disruptive behavior disorders, and subaverage deficit/hyperactivity disorder. Psychophysiology, 38, 212–221.
IQ. Journal of Child and Adolescent Psychopharmacology, 14, Clarke, A. R., Barry, R. J., McCarthy, R., & Selikowitz, M.
243–254. (2002). EEG differences between good and poor responders to
Barkley, R. A. (1997). ADHD and the nature of self control. New York: methylphenidate and dexamphetamine in children with attention-
Guildford Press. deficit/hyperactivity disorder. Clinical Neurophysiology, 113,
Barkley, R. A. (2006). Comorbid disorders, social and family adjust- 194–205.
ment, and subtyping. In R. A. Barkley (Ed.), Attention Deficit Conners, C. K., Casat, C. D., Gualtieri, C. T., Weller, E., Reader,
Hyperactivity Disorder (pp. 184–218). New York: Guilford Press. M., Reiss, A., et al. (1996). Bupropion hydrochloride in atten-
Barry, R. J., Clarke, A. R., & Johnstone, S. J. (2003). A review of elec- tion deficit disorder with hyperactivity. Journal of the American
trophysiology in attention-deficit/hyperactivity disorder: I. Qual- Academy of Child and Adolescent Psychiatry, 35, 1314–1321.
itative and quantitative electroencephalography. Clinical Neuro- Connor, D. F., Fletcher, K. E., & Swanson, J. M. (1999). A meta-analysis
physiology, 114, 171–183. of clonidine for symptoms of attention-deficit hyperactivity dis-
Barry, R. J., Johnstone, S. J., & Clarke, A. R. (2003). A review of elec- order. Journal of the American Academy of Child and Adolescent
trophysiology in attention-deficit/hyperactivity disorder: II. Event- Psychiatry, 38, 1551–1559.
related potentials. Clinical Neurophysiology, 114, 184–198. Connor, D. F., Glatt, S. J., Lopez, I. D., Jackson, D., & Melloni,
Biederman, J., Lopez, F. A., Boellner, S. W., & Chandler, M. C. R. H., Jr. (2002). Psychopharmacology and aggression. I: A meta-
(2002). A randomized, double-blind, placebo-controlled, parallel- analysis of stimulant effects on overt/covert aggression-related
group study of SLI381 (Adderall XR) in children with attention- behaviors in ADHD. Journal of the American Academy of Child
deficit/hyperactivity disorder. Pediatrics, 110, 258–266. and Adolescent Psychiatry, 41, 253–261.
Biederman, J., Mick, E., Surman, C., Doyle, R., Hammerness, P., Correll, C. U. & Carlson, H. E. (2006). Endocrine and metabolic ad-
Harpold, T., et al. (2006). A randomized, placebo-controlled verse effects of psychotropic medications in children and adoles-
trial of OROS methylphenidate in adults with attention- cents. Journal of the American Academy of Child and Adolescent
deficit/hyperactivity disorder. Biological Psychiatry, 59, 829–835. Psychiatry, 45, 771–791.
Biederman, J., Spencer, T. J., Wilens, T. E., Weisler, R. H., Read, S. C., Daly, J. M., & Wilens, T. (1998). The use of tricyclics antidepressants
& Tulloch, S. J. (2005). Long-term safety and effectiveness of in children and adolescents. Pediatric Clinics of North America,
mixed amphetamine salts extended release in adults with ADHD. 45, 1123–1135.
CNS Spectrum, 10, 16–25. Faraone, S. V., Biederman, J., Morley, C. P., & Spencer, T. J. (2006).
Biederman, J., Wigal, S. B., Spencer, T. J., McGough, J. J., & Mays, The effect of stimulants on height and weight: a review of the
D. A. (2006). A post hoc subgroup analysis of an 18-day ran- literature (manuscript in preparation).
domized controlled trial comparing the tolerability and efficacy Feron, F. J., Hendriksen, J. G., van Kroonenburgh, M. J.,
of mixed amphetamine salts extended release and atomoxetine Blom-Coenjaerts, C., Kessels, A. G., Jolles, J., et al. (2005).
in school-age girls with attention-deficit/hyperactivity disorder. Dopamine transporter in attention-deficit hyperactivity disorder
Clinical Therapeutics, 28, 280–293. normalizes after cessation of methylphenidate. Pediatric Neurol-
Bradley, C. (1937). The behavior of children receiving benzedrine. ogy, 33, 179–183.
American Journal of Psychiatry, 94, 577–585. Findling, R. L., & Lopez, F. A. (2005). Efficacy of transdermal
Broyd, S. J., Johnstone, S. J., Barry, R. J., Clarke, A. R., McCarthy, methylphenidate with reference to Concerta in ADHD. Presented
R., Selikowitz, M., et al. (2005). The effect of methylphenidate at the 25th Annual Meeting of the American Academy of Child
on response inhibition and the event-related potential of children and Adolescent Psychiatry, Toronto, CA, (Oct 18–23).
with attention deficit/hyperactivity disorder. International Journal Findling, R. L., Biederman, J., Wilens, T. E., Spencer, T. J., McGough, J.
of Psychophysiology, 58, 47–58. J., Lopez, F. A., et al. (2005). Short- and long-term cardiovascular
Bush, G. (2005). Funcitional MRI: Recent advances in studying ADHD effects of mixed amphetamine salts extended release in children.
pathophysiology and treatment. Presented at the 52nd Annual The Journal of Pediatrics, 147, 348–354.
Meeting of the American Academy of Child and Adolescent Psy- Food and Drug Administration (2005). FDA Alert [09/05]: Sui-
chiatry. Toronto, CA (Oct. 18–23). cidal thinking in children and adolescents. Available at.
Bush, G., Frazier, J. A., Rauch, S. L., Seidman, L. J., Whalen, P. J., Food and Drug Administration Website [On-line]. Available:
Jenike, M. A., et al. (1999). Anterior cingulate cortex dysfunction http://www.fda.gov/cder/drug/infopage/atomoxetine/default.htm
in attention-deficit/hyperactivity disorder revealed by fMRI and Gadow, K. D., Sverd, J., Sprafkin, J., Nolan, E. E., & Grossman, S.
the Counting Stroop. Biological Psychiatry, 45, 1542–1552. (1999). Long-term methylphenidate therapy in children with co-
Bush, G., Valera, E. M., & Seidman, L. J. (2005). Functional neu- morbid attention-deficit hyperactivity disorder and chronic multi-
roimaging of attention-deficit/hyperactivity disorder: A review ple tic disorder [see comments]. Archives of General Psychiatry,
and suggested future directions. Biological Psychiatry, 57, 1273– 56, 330–336.
1284. Gelperin, K. (2006). Psychiatric adverse events associated with drug
Castellanos, F. X. (2002). Proceed, with caution: SPECT cerebral blood treatment of ADHD: Review of post marketing safety data. Avail-
flow studies of children and adolescents with attention deficit able at. Food and Drug Administration Website [On-line]. Avail-
hyperactivity disorder. Journal of Nuclear Medicine, 43, 1630– able: http://www.fda.gov/ohrms/dockets/ac/06/briefing/2006-
1633. 4210B-Index.htm
Charach, A., Figueroa, M., Chen, S., Ickowicz, A., & Schachar, R. Greenhill, L. L., Biederman, J., Boellner, S. W., Rugino, T. A.,
(2006). Stimulant treatment over 5 years: Effects on growth. Jour- Sangal, R. B., Earl, C. Q., et al. (2006a). A randomized, double-
nal of the American Academy of Child and Adolescent Psychiatry, blind, placebo-controlled study of modafinil film-coated tablets in
45, 415–421. children and adolescents with attention-deficit/hyperactivity dis-
Clarke, A. R., Barry, R. J., Bond, D., McCarthy, R., & Selikowitz, order. Journal of the American Academy of Child and Adolescent
M. (2002). Effects of stimulant medications on the EEG of chil- Psychiatry, 45, 503–511.
dren with attention-deficit/hyperactivity disorder. Psychopharma- Greenhill, L. L., Findling, R. L., & Swanson, J. M. (2002).
cology (Berl.), 164, 277–284. A double-blind, placebo-controlled study of modified-release

Springer
70 Neuropsychol Rev (2007) 17:61–72

methylphenidate in children with attention-deficit/hyperactivity Matochik, J. A., Nordahl, T. E., Gross, M., Semple, W. E., King, A. C.,
disorder. Pediatrics, 109, E39. Cohen, R. M., et al. (1993). Effects of acute stimulant medication
Greenhill, L. L., Muniz, R., Ball, R. R., Levine, A., Pestreich, on cerebral metabolism in adults with hyperactivity. Neuropsy-
L., & Jiang, H. (2006b). Efficacy and safety of dexmethylphenidate chopharmacology, 8, 377–386.
extended-release capsules in children with attention- McGough, J. J., Biederman, J., Wigal, S. B., Lopez, F. A., McCracken,
deficit/hyperactivity disorder. Journal of the American Academy J. T., Spencer, T., et al. (2005). Long-term tolerability and effec-
of Child and Adolescent Psychiatry, 45, 817–823. tiveness of once-daily mixed amphetamine salts (Adderall XR) in
Hammad, T. A., Laughren, T., & Racoosin, J. (2006). Suicidality in children with ADHD. Journal of the American Academy of Child
pediatric patients treated with antidepressant drugs. Archives of and Adolescent Psychiatry, 44, 530–538.
General Psychiatry, 63, 332–339. Michelson, D., Adler, L., Spencer, T., Reimherr, F. W., West, S. A.,
Jensen, P. S., Youngstrom, Y., Steiner, H., Findling, R. L., Meyer, R. E., Allen, A. J., et al. (2003). Atomoxetine in adults with ADHD:
Malone, R., et al. (in press) Consensus report: Impulsive aggres- Two randomized, placebo-controlled studies. Biological Psychia-
sion as a symptom across diagnostic categories in child psychi- try, 53, 112–120.
atry. Journal of the American Academy of Child and Adolescent Michelson, D., Allen, A. J., Busner, J., Casat, C., Dunn, D., Kratochvil,
Psychiatry. C., et al. (2002). Once-daily atomoxetine treatment for children
Jonkman, L. M., Kemner, C., Verbaten, M. N., van Engeland, H., and adolescents with attention deficit hyperactivity disorder: A
Camfferman, G., Buitelaar, J. K., et al. (2000). Attentional ca- randomized, placebo-controlled study. American Journal of Psy-
pacity, a probe ERP study: Differences between children with chiatry, 159, 1896–1901.
attention-deficit hyperactivity disorder and normal control chil- Michelson, D., Faries, D., Wernicke, J., Kelsey, D., Kendrick, K., Sallee,
dren and effects of methylphenidate. Psychophysiology, 37, 334– R., et al. (2001). Atomoxetine in the treatment of children and ado-
346. lescents with attention-deficit/hyperactivity disorder: a random-
Jucaite, A., Fernell, E., Halldin, C., Forssberg, H., & Farde, L. (2005). ized, placebo-controlled, dose-response study. Pediatrics, 108, 1–
Reduced midbrain dopamine transporter binding in male adoles- 9.
cents with attention-deficit/hyperactivity disorder: Association be- MTA Cooperative Group (2004). National institute of mental health
tween striatal dopamine markers and motor hyperactivity. Biolog- multimodal treatment study of ADHD follow-up: Changes in ef-
ical Psychiatry, 57, 229–238. fectiveness and growth after the end of treatment. Pediatrics, 113,
Kim, B. N., Lee, J. S., Cho, S. C., & Lee, D. S. (2001). Methylphenidate 762–769.
increased regional cerebral blood flow in subjects with attention MTA Cooperative Group (2006). Effects of stimulant medication on
deficit/hyperactivity disorder. Yonsei Medical Journal, 42, 19–29. growth rates across three years in the MTA follow up. (in submis-
Klein, R. G., Abikoff, H., Klass, E., Ganeles, D., Seese, L. M., & sion).
Pollack, S. (1997). Clinical efficacy of methylphenidate in conduct Muller, U., Clark, L., Lam, M. L., Moore, R. M., Murphy, C. L.,
disorder with and without attention deficit hyperactivity disorder. Richmond, N. K., et al. (2005). Lack of effects of guanfacine
Archives of General Psychiatry, 54, 1073–1080. on executive and memory functions in healthy male volunteers.
Klorman, R. (1991). Cognitive event-related potentials in attention Psychopharmacology (Berl.), 182, 205–213.
deficit disorder. Journal of Learning Disabilities, 24, 130–140. Newcorn, J. H., Spencer, T. J., Biederman, J., Milton, D. R., &
Kok, A. (1986). Effects of degradation of visual stimuli on components Michelson, D. (2005). Atomoxetine treatment in children and ado-
of the event-related potential (ERP) in go/nogo reaction tasks. lescents with attention-deficit/hyperactivity disorder and comorbid
Biological Psychology, 23, 21–38. oppositional defiant disorder. Journal of the American Academy
Kok, A. (1997). Event-related-potential (ERP) reflections of mental of Child and Adolescent Psychiatry, 44, 240–248.
resources: A review and synthesis. Biological Psychology, 45, Pappadopulos, E., Macintyre Ii, J. C., Crismon, M. L., Findling, R. L.,
19–56. Malone, R. P., Derivan, A., et al. (2003). Treatment recommenda-
Krause, K. H., Dresel, S. H., Krause, J., la Fougere, C., & tions for the use of antipsychotics for aggressive youth (TRAAY).
Ackenheil, M. (2003). The dopamine transporter and neuroimag- Part II. Journal of the American Academy of Child and Adolescent
ing in attention deficit hyperactivity disorder. Neuroscience and Psychiatry, 42, 145–161.
Biobehavioral Reviews, 27, 605–613. Pliszka, S. R., Browne, R., Olvera, R. L., & Wynne, S. K. (2000). A
Langleben, D. D., Acton, P. D., Austin, G., Elman, I., Krikorian, G., double-blind placebo controlled trial of Adderall and methylphen-
Monterosso, J. R., et al. (2002). Effects of methylphenidate discon- diate in the treatment of Attention deficit hyperactivity disorder.
tinuation on cerebral blood flow in prepubescent boys with atten- Journal of the American Academy of Child and Adolescent Psy-
tion deficit hyperactivity disorder. Journal of Nuclear Medicine, chiatry, 39, 619–626.
43, 1624–1629. Pliszka, S. R., Crismon, M. L., Hughes, C. W., Corners, C. K., Emslie,
Loo, S. K. & Barkley, R. A. (2005). Clinical utility of EEG in attention G. J., Jensen, P. S., et al. (2006a). The Texas Children’s Medication
deficit hyperactivity disorder. Applied Neuropsychology, 12, 64– Algorithm Project: Revision of the algorithm for pharmacotherapy
76. of attention-deficit/hyperactivity disorder. Journal of the American
Loo, S. K., Hopfer, C., Teale, P. D., & Reite, M. L. (2004). EEG cor- Academy of Child and Adolescent Psychiatry, 45, 642–657.
relates of methylphenidate response in ADHD: Association with Pliszka, S. R., Glahn, D. C., Semrud-Clikeman, M., Franklin, C., Perez,
cognitive and behavioral measures. Journal of Clinical Neuro- R., Xiong, J., et al. (2006b). Neuroimaging of inhibitory control
physiology, 21, 457–464. areas in children with attention defict hyperactivity disorder who
Loo, S. K., Specter, E., Smolen, A., Hopfer, C., Teale, P. D., & Reite, were treatment naive or in long term treatment. American Journal
M. L. (2003). Functional effects of the DAT1 polymorphism on of Psychiatry, 163, 1052–1060.
EEG measures in ADHD. Journal of the American Academy of Pliszka, S. R., Liotti, M., & Woldorff, M. G. (2000). Inhibitory control in
Child and Adolescent Psychiatry, 42, 986–993. children with attention deficit/hyperactivity disorder: Event related
Matochik, J. A., Liebenauer, L. L., King, A. C., Szymanski, H. V., potentials identify the processing component and timing of an
Cohen, R. M., & Zametkin, A. J. (1994). Cerebral glucose impaired right-frontal response-inhibition mechanism. Biological
metabolism in adults with attention deficit hyperactivity disorder Psychiatry, 48, 238–246.
after chronic stimulant treatment. American Journal of Psychiatry, Pliszka, S. R., Matthews, T. L., Braslow, K. J., & Watson, M. A.
151, 658–664. (2006c). Comparative effects of methylphenidate and mixed salts

Springer
Neuropsychol Rev (2007) 17:61–72 71

amphetamine on height and weight in children with attention- patients with ADHD and comorbid anxiety. Presented at the annual
deficit/hyperactivity disorder (ADHD). Journal of the American meeting of the American Psychiatric Association, Altanta, GA
Academy of Child and Adolescent Psychiatry, 45, 520–526. (May).
Poulton, A. (2005). Growth on stimulant medication; clarifying the Swanson, J. M., Greenhill, L. L., Lopez, F. A., Sedillo, A., Earl, C. Q.,
confusion: A review. Archives of Diseases of Childhood, 90, 801– Jiang, J. G., et al. (2006). Modafinil film-coated tablets in chil-
806. dren and adolescents with attention-deficit/hyperactivity disorder:
Rosa-Neto, P., Lou, H. C., Cumming, P., Pryds, O., Karrebaek, H., Results of a randomized, double-blind, placebo-controlled, fixed-
Lunding, J., et al. (2005). Methylphenidate-evoked changes in dose study followed by abrupt discontinuation. Journal of Clinical
striatal dopamine correlate with inattention and impulsivity in ado- Psychiatry, 67, 137–147.
lescents with attention deficit hyperactivity disorder. Neuroimage, Swanson, J. M., McBurnett, K., Wigal, T., Pfiffner, L. J., Lerner,
25, 868–876. M. A., Williams, L., et al. (1993). Effect of stimulant medica-
Rubia, K., Smith, A. B., Brammer, M. J., Toone, B., & Taylor, E. (2005). tion on children with attention deficit disorder: A “review of re-
Abnormal brain activation during inhibition and error detection in views.”Exceptional Children, 60, 154–162.
medication-naive adolescents with ADHD. American Journal of Swensen, A., Michelsen, D., Buesching, D., & Faries, D. E. (2001).
Psychiatry, 162, 1067–1075. Effects of atomoxetine on social and family functioning of ADHD
Scahill, L., Chappell, P. B., Kim, Y. S., Schultz, R. T., Katsovich, L., children and adolescents. Presented at the 48th Annual Meeting
Shepherd, E., et al. (2001). A placebo-controlled study of guan- of the American Academy of Child and Adolescent Psychiatry,
facine in the treatment of children with tic disorders and attention Honolulu,HI, (Oct 23–28).
deficit hyperactivity disorder. American Journal of Psychiatry, Szobot, C. M., Ketzer, C., Cunha, R. D., Parente, M. A., Langleben, D.
158, 1067–1074. D., Acton, P. D., et al. (2003). The acute effect of methylphenidate
Schmajuk, M., Liotti, M., Busse, L., & Woldorff, M. G. (2006). Elec- on cerebral blood flow in boys with attention-deficit/hyperactivity
trophysiological activity underlying inhibitory control processes disorder. European Journal of Nuclear Medicine and Molecular
in normal adults. Neuropsychologia., 44, 384–395. Imaging, 30, 423–426.
Schur, S. B., Sikich, L., Findling, R. L., Malone, R. P., Crismon, M. Tourette’s Syndrome Study Group (2002). Treatment of ADHD in chil-
L., Derivan, A., et al. (2003). Treatment recommendations for dren with tics: A randomized controlled trial. Neurology, 58, 527–
the use of antipsychotics for aggressive youth (TRAAY). Part I: A 536.
review. Journal of the American Academy of Child and Adolescent Vaidya, C. J., Austin, G., Kirkorian, G., Ridlehuber, H. W., Desmond,
Psychiatry, 42, 132–144. J. E., Glover, G. H., et al. (1998). Selective effects of
Schweitzer, J. B., Lee, D. O., Hanford, R. B., Tagamets, M. A., Hoff- methylphenidate in attention deficit hyperactivity disorder: A
man, J. M., Grafton, S. T., et al. (2003). A positron emission functional magnetic resonance study. Proceedings of the National
tomography study of methylphenidate in adults with ADHD: Al- Academy of Science, 95, 14494–14499.
terations in resting blood flow and predicting treatment response. Villalaba, L. (2006). Follow up review of AERS search identifying
Neuropsychopharmacology, 28, 967–973. cases of sudden death occuring with drugs used for the treatment
Seifert, J., Scheuerpflug, P., Zillessen, K. E., Fallgatter, A., & Warnke, of attention deficit hyperactivity disorder (ADHD). Available
A. (2003). Electrophysiological investigation of the effectiveness at. Food and Drug Administration Website [On-line]. Available:
of methylphenidate in children with and without ADHD. Journal http://www.fda.gov/ohrms/dockets/ac/06/briefing/2006-4210B-
of Neural Transmission, 110, 821–829. Index.htm
Shafritz, K. M., Marchione, K. E., Gore, J. C., Shaywitz, S. E., & Vles, J. S., Feron, F. J., Hendriksen, J. G., Jolles, J., van Kroonenburgh,
Shaywitz, B. A. (2004). The effects of methylphenidate on neural M. J., & Weber, W. E. (2003). Methylphenidate down-regulates
systems of attention in attention deficit hyperactivity disorder. the dopamine receptor and transporter system in children with
American Journal of Psychiatry, 161, 1990–1997. attention deficit hyperkinetic disorder (ADHD). Neuropediatrics.,
Snyder, R., Turgay, A., Aman, M., Binder, C., Fisman, S., & Carroll, A. 34, 77–80.
(2002). Effects of risperidone on conduct and disruptive behavior Volkow, N. D., Wang, G., Fowler, J. S., Logan, J., Gerasimov, M.,
disorders in children with subaverage IQs. Journal of the American Maynard, L., et al. (2001). Therapeutic doses of oral
Academy of Child and Adolescent Psychiatry, 41, 1026–1036. methylphenidate significantly increase extracellular dopamine in
Spencer, T., Biederman, J., Wilens, T., Harding, M., O’Donnell, D., the human brain. Journal of Neuroscience, 21, RC121.
& Griffin, S. (1996). Pharmacotherapy of attention-deficit hyper- Volkow, N. D., Wang, G. J., Fowler, J. S., & Ding, Y. S. (2005). Imaging
activity disorder across the life cycle. Journal of the American the effects of methylphenidate on brain dopamine: New model on
Academy of Child and Adolescent Psychiatry, 35, 409–432. its therapeutic actions for attention-deficit/hyperactivity disorder.
Spencer, T. J., Biederman, J., Madras, B. K., Faraone, S. V., Dougherty, Biological Psychiatry, 57, 1410–1415.
D. D., Bonab, A. A., et al. (2005). In vivo neuroreceptor imaging in Volkow, N. D., Wang, G. J., Fowler, J. S., Gatley, S. J., Logan, J.,
attention-deficit/hyperactivity disorder: A focus on the dopamine Ding, Y. S., et al. (1998). Dopamine transporter occupancies in the
transporter. Biological Psychiatry, 57, 1293–1300. human brain induced by therapeutic doses of oral methylphenidate.
Spencer, T. J., Faraone, S. V., Biederman, J., Lerner, M., Cooper, K. M., American Journal of Psychiatry, 155, 1325–1331.
& Zimmerman, B. (2006a). Does prolonged therapy with a long- Volkow, N. D., Wang, G. J., Fowler, J. S., Telang, F., Maynard, L.,
acting stimulant suppress growth in children with ADHD? Journal Logan, J., et al. (2004). Evidence that methylphenidate enhances
of the American Academy of Child and Adolescent Psychiatry, 45, the saliency of a mathematical task by increasing dopamine in
527–537. the human brain. American Journal of Psychiatry, 161, 1173–
Spencer, T. J., Wilens, T. E., Biederman, J., Weisler, R. H., Read, 1180.
S. C., & Pratt, R. (2006b). Efficacy and safety of mixed am- Weisler, R. H., Biederman, J., Spencer, T. J., Wilens, T. E., Faraone,
phetamine salts extended release (Adderall XR) in the manage- S. V., Chrisman, A. K., et al. (2006). Mixed amphetamine salts
ment of attention-deficit/hyperactivity disorder in adolescent pa- extended-release in the treatment of adult ADHD: A randomized,
tients: A 4-week, randomized, double-blind, placebo-controlled, controlled trial. CNS Spectrum, 11, 625–639.
parallel-group study. Clinical Therapeutics, 28, 266–279. Weiss, M., Wasdell, M., & Patin, J. (2004). A post hoc analy-
Sumner, C. S., Donnelly, C., Lopez, F. A., Sutton, V., Bakken, R., sis of d-threo-methylphenidate hydrochloride (focalin) versus
Paczkowski, M., et al. (2005). Atomoxetine treatment for pediatric d,l-threo-methylphenidate hydrochloride (ritalin). Journal of the

Springer
72 Neuropsychol Rev (2007) 17:61–72

American Academy of Child and Adolescent Psychiatry, 43, 1415– label study. Journal of the American Academy of Child and Ado-
1421. lescent Psychiatry, 44, 1015–1023.
Wigal, S. B., McGough, J. J., McCracken, J. T., Biederman, J., Spencer, Wilens, T. E., McBurnett, K., Bukstein, O., McGough, J., Greenhill,
T. J., Posner, K. L., et al. (2005). A laboratory school compari- L., Lerner, M., et al. (2006). Multisite controlled study of OROS
son of mixed amphetamine salts extended release (Adderall XR) methylphenidate in the treatment of adolescents with attention-
and atomoxetine (Strattera) in school-aged children with attention deficit/hyperactivity disorder. Archives of Pediatrics and Adoles-
deficit/hyperactivity disorder. Journal of Attention Disorders, 9, cent Medicine, 160, 82–90.
275–289. Wolraich, M. L., Greenhill, L. L., Pelham, W., Swanson, J., Wilens,
Wilens, T., McBurnett, K., Stein, M., Lerner, M., Spencer, T., & T., Palumbo, D., et al. (2001). Randomized, controlled trial of
Wolraich, M. (2005). ADHD treatment with once daily OROS oros methylphenidate once a day in children with attention-
methylphendiate treatment: Final results from a long term open- deficit/hyperactivity disorder. Pediatrics, 108, 883–892.

Springer

You might also like