You are on page 1of 21

PFI_12mmX178mm.

pdf + eps format

Journal of Child Psychology and Psychiatry **:* (2020), pp **–** doi:10.1111/jcpp.13305

PRACTITIONER REVIEW

Practitioner Review: Pharmacological treatment


of attention-deficit/hyperactivity disorder symptoms
in children and youth with autism spectrum disorder:
a systematic review and meta-analysis
Rebecca Rodrigues,1* Meng-Chuan Lai,2,3,4,5,6* Adam Beswick,1 Daniel A. Gorman,3,4
7 2,3,4
Evdokia Anagnostou, Peter Szatmari, Kelly K. Anderson,1,8* and
2,3,4
Stephanie H. Ameis *
1
Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University,
London, ON, Canada; 2The Margaret and Wallace McCain Centre for Child, Youth and Family Mental Health,
Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada;
3
Centre for Brain and Mental Health, Department of Psychiatry, The Hospital for Sick Children, Toronto, ON,
Canada; 4Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; 5Autism
Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK; 6Department of Psychiatry,
National Taiwan University Hospital and College of Medicine, Taipei, Taiwan; 7Holland Bloorview Kids Rehabilitation
Hospital, Bloorview Research Institute, Department of Pediatrics, University of Toronto, Toronto, ON, Canada;
8
Department of Psychiatry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada

Background: Clinically significant attention-deficit/hyperactivity disorder (ADHD) symptoms are common and
impairing in children and youth with autism spectrum disorder(ASD). The aim of this systematic review and meta-
analysis was to (a) evaluate the efficacy and safety of pharmacotherapy for the treatment of ADHD symptoms in ASD
and (b) distil findings for clinical translation. Methods: We searched electronic databases and clinical trial registries
(1992 onwards). We selected randomized controlled trials conducted in participants <25 years of age, diagnosed with
ASD that evaluated ADHD outcomes (hyperactivity/impulsivity and inattention) following treatment with stimulants
(methylphenidate or amphetamines), atomoxetine, alpha-2 adrenergic receptor agonists, antipsychotics, tricyclic
antidepressants, bupropion, modafinil, venlafaxine, or a combination, in comparison with placebo, any of the listed
medications, or behavioral therapies. Data were pooled using a random-effects model. Results: Twenty-five studies
(4 methylphenidate, 4 atomoxetine, 1 guanfacine, 14 antipsychotic, 1 venlafaxine, and 1 tianeptine) were included.
Methylphenidate reduced hyperactivity (parent-rated: standardized mean difference [SMD] = .63, 95%
CI = .95, .30; teacher-rated: SMD = .81, 95%CI = 1.43, .19) and inattention (parent-rated: SMD = .36,
95%CI = .64, .07; teacher-rated: SMD = .30, 95%CI = .49, .11). Atomoxetine reduced inattention (parent-
rated: SMD = .54, 95%CI = .98, .09; teacher/investigator-rated: SMD = 0.38, 95%CI = 0.75, 0.01) and
parent-rated hyperactivity (parent-rated: SMD = .49, 95%CI = .76, .23; teacher-rated: SMD = .43, 95%
CI = .92, .06). Indirect evidence for significant reductions in hyperactivity with second-generation antipsychotics
was also found. Quality of evidence for all interventions was low/very low. Methylphenidate was associated with a
nonsignificant elevated risk of dropout due to adverse events. Conclusions: Direct pooled evidence supports the
efficacy and tolerability of methylphenidate or atomoxetine for treatment of ADHD symptoms in children and youth
with ASD. The current review highlights the efficacy of standard ADHD pharmacotherapy for treatment of ADHD
symptoms in children and youth with ASD. Consideration of the benefits weighed against the limitations of safety/
efficacy data and lack of data evaluating long-term continuation is undertaken to help guide clinical decision-making
regarding treatment of co-occurring ADHD symptoms in children and youth with ASD. Keywords: Attention-deficit/
hyperactivity disorder; ADHD; hyperactivity; impulsivity; inattention; autism spectrum disorder; pervasive
developmental disorder; pharmacotherapy; stimulants; atomoxetine; alpha-2 agonists; antipsychotics; tricyclic
antidepressants; venlafaxine; bupropion; modafinil..

Szatmari, 2015; Gillberg, 2010; Lai, Lombardo, &


Introduction
Baron-Cohen, 2014; Soke, Maenner, Christensen,
Autism spectrum disorder (ASD) is an early-onset
Kurzius-Spencer, & Schieve, 2018). The latest sys-
neurodevelopmental disorder characterized by a
tematic review and meta-analysis indicated that in
high co-occurrence (>70%) of other neurodevelop-
population-based cohorts, 22% (95% confidence
mental and psychiatric challenges (Ameis &
interval, 95%CI 17%–26%) of individuals with ASD
meet the clinical criteria for attention-deficit/hyper-
*Authors contributed equally to this work.
activity disorder (ADHD), and the rate is even higher
Conflict of interest statement: See Acknowledgements for full in samples recruited from clinical settings (34%,
disclosures. 95%CI 29%–39%; Lai et al., 2019). Correspondingly,

© 2020 Association for Child and Adolescent Mental Health


Published by John Wiley & Sons Ltd, 9600 Garsington Road, Oxford OX4 2DQ, UK and 350 Main St, Malden, MA 02148, USA
2 Rebecca Rodrigues et al.

in children and adolescents with ADHD, 21% (95%CI evaluation and the other medication choice, based on
18%–24%) reach the diagnostic threshold for ASD their review of studies published between 2000 and
(Hollingdale et al., 2019). Dimensional traits of ASD 2010 (Mahajan et al., 2012). Their recommendations
and ADHD also co-occur in the general population include initial optimization of medical, mental health,
(Polderman et al., 2014; Stergiakouli et al., 2017). and educational/behavioral interventions, followed
The co-occurrence of ASD and ADHD is also associ- by first-line medication treatment with methylpheni-
ated with greater impairment in adaptive functioning date or amphetamine salts, second-line treatment
and poorer health-related quality of life (Sikora, with atomoxetine or alpha-2 agonists (guanfacine or
Vora, Coury, & Rosenberg, 2012). clonidine), and reserved use of atypical antipsychotics
There may be a number of mechanisms resulting for children with severe impulsivity and associated
in these high rates of phenotypic co-occurrence of safety concerns (Mahajan et al., 2012).
ADHD in individuals with ASD. Both ASD and ADHD The current literature suggests that additional
are common (Christensen et al., 2018; Willcutt, resources to guide clinical care for co-occurring
2012) and highly heritable neurodevelopmental con- ASD and ADHD are needed for the following reasons.
ditions (Palladino, McNeill, Reif, & Kittel-Schneider, First, psychotropic prescription rates are increasing
2019; Sandin et al., 2017). Large-scale population in children diagnosed with ASD (Dalsgaard, Nielsen,
and registry-based studies consistently show famil- & Simonsen, 2013; Frazier et al., 2011; Kamimura-
ial coaggregation of ASD and ADHD clinical diag- Nishimura et al., 2017). ADHD is the most common
noses and dimensional traits (Ghirardi et al., 2018; co-occurring mental health diagnosis driving
Ghirardi et al., 2019; Miller et al., 2019), indicative of increased rates of psychotropic drug use (Jobski,
shared underlying etiological (e.g., genetic) factors. H€ofer, Hoffmann, & Bachmann, 2017; Kamimura-
Overlapping biological pathways cutting across ASD Nishimura et al., 2017) as well as long-term psy-
and ADHD may involve overlapping rare mutations chotropic use (Houghton, Liu, & Bolognani, 2018).
in the form of copy number variants (Martin et al., Rates of polypharmacy in children with ASD are high
2014), with effects on specific genetic pathways that (Jobski et al., 2017). Finally, a variety of medication
regulate neural migration and synaptic development classes are being prescribed, without established
(Lionel et al., 2014). Nondistinct social cognitive evidence to support their use (Frazier et al., 2011;
(Bora and Pantelis 2016) and executive functioning Houghton et al., 2018; Jobski et al., 2017).
characteristics (Craig et al., 2016) across samples Since the publication of the aforementioned
with ASD or ADHD may also be linked to similar reviews on the efficacy of pharmacological interven-
large-scale neural network properties found across tions for ADHD symptoms in children with ASD, new
children with either diagnosis (Ameis et al., 2016; empirical studies are available that examine efficacy
Aoki et al., 2017; Di Martino et al., 2013). Conver- of additional medications (Loebel et al., 2016;
gent findings are thus suggestive of shared neurode- McDougle, 2017; Scahill et al., 2015) enabling
velopmental pathways that may confer risk for either meta-analysis and presentation of clinical effects
(or both) disorder(s) (Miller et al., 2019). across different medication classes to inform clinical
To date, one national guideline (National Institute recommendations. As prior meta-analyses on this
for Health & Care Excellence, 2013), five systematic topic provide quantitative analysis of the evidence for
reviews (Cortese, Castelnau, Morcillo, Roux, & Bon- a single medication class (Reichow et al., 2013, Patra
net-Brilhault, 2012; Ghanizadeh, 2013; Mahajan et al., 2019, Sturman et al., 2017) and prior clinical
et al., 2012; Patra, Nebhinani, Viswanathan, & guidance is based on a systematic review of roughly
Kirubakaran, 2019; Reichow, Volkmar, & Bloch, half of currently available RCTs, an updated system-
2013), and three Cochrane reviews (James, Mont- atic review and meta-analysis across different med-
gomery, & Williams, 2011; Millward, Ferriter, Calver, ication classes and clinical interpretation would
& Connell-Jones, 2008; Sturman, Deckx, & van provide valuable practical guidance for front-line
Driel, 2017) have evaluated the available literature clinicians.
on the efficacy of pharmacological or other biomed- The present systematic review and meta-analysis
ical interventions for ADHD symptoms in children aims to synthesize and re-evaluate randomized con-
with ASD. These reviews largely include empirical trolled trials (RCTs) for this indication, including all
studies published up to 2013 and highlight (a) relevant studies. We aim to update and expand on
evidence that methylphenidate and atomoxetine previously published reviews to provide one compre-
improve symptoms of hyperactivity and possibly hensive resource (with clear clinical utility) evaluat-
inattention in children with ASD (Patra et al., 2019; ing the following: (a) all available RCTs testing
Sturman et al., 2017) and (b) a lack of evidence for efficacy of pharmacological treatments for ADHD
improvement of ADHD symptoms in ASD with symptoms in children and youth with ASD, (b)
omega-3 fatty-acid supplements or gluten/casein- efficacy of medication treatment on secondary out-
free diets. comes relevant to general functioning, and (c) rates
In addition, the Autism Treatment Network (ATN) of adverse events with treatment. Evidence for effi-
Psychopharmacology Committee published two rec- cacy is distilled, and recommendations for best
ommended clinical pathways, one guiding symptom clinical practice are provided.

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 3

the inclusion criteria (e.g., whether studies including a placebo


Methods versus an active comparator group should be included) and
Protocol and registration were resolved through discussion among the research team
and further clarification of the criteria. Studies were included
Prior to conducting this review, we published our protocol on
in meta-analyses where data were available and where study
the International Prospective Register of Systematic Reviews
design and populations were comparable for pooling.
(PROSPERO; http://www.crd.york.ac.uk/prospero; protocol
number CRD42016052610). We adhered to the Preferred
Reporting Items for Systematic Review and Meta-Analyses Data extraction
(PRISMA) guidelines (Moher et al., 2009).
One author (RR) conducted a double data extraction using a
piloted data extraction sheet. Information on the study design,
Search strategy participants, interventions (i.e., doses and form of administra-
tion), methods, baseline characteristics, and results was
We searched MEDLINE, Embase, CINAHL, PsycINFO, ERIC,
extracted. Data on primary and secondary outcome measures,
Cochrane Library, Web of Science, ClinicalTrials.gov (www.c
including scales used, rater(s), follow-up, statistical analyses,
linicaltrials.gov), and the World Health Organization Interna-
and results, were also extracted. Corresponding authors of
tional Clinical Trials Registry Platform (WHO IRCTP; http://
included studies were contacted when missing data were
www.who.int/ictrp/en/) from 1992 onwards. The MEDLINE
encountered or for studies where point estimates were not
search strategy is available in the online supplement
provided for our primary outcome measures.
(Appendix S1). We conducted forward and backward citation
searching of included studies and related systematic reviews
(Barnard, Young, Pearson, Geddes, & O’Brien, 2002; Cohen Risk-of-bias assessment
et al., 2013; Deb et al., 2014; Fung et al., 2016; Ghanizadeh,
2013; Ghanizadeh, Tordjman, & Jaafari, 2015; Hirsch & Risk of bias was assessed using the Cochrane Risk of Bias
Pringsheim, 2016; Hurwitz, Blackmore, Hazell, Williams, & Assessment tool (Higgins & Green, 2011). Two authors (RR and
Woolfenden, 2012; Mahajan et al., 2012; Reichow et al., 2013; AB) independently rated each study as having low, high, or
Sharma & Shaw, 2012; Siegel & Beaulieu, 2012; Sochocky & unclear risk of bias for each domain: random sequence
Milin, 2013). We manually searched tables of contents of generation, allocation concealment, blinding of participants
relevant journals from 1992 onwards. We contacted authors of and personnel, blinding of outcome assessment, incomplete
abstracts or unpublished studies to determine whether studies outcome data, selective reporting, and other sources of bias,
had subsequently been published in a peer-reviewed journal. such as industry funding. For risk-of-bias domains where
Our search was completed in April 2018. assessment of risk of bias may vary by outcome (e.g., blinding
of outcome assessment), we assessed risk of bias in terms of
our primary outcomes of ADHD symptoms. Disagreements
Eligibility criteria and outcome measures were discussed (among RR, AB, KKA) until a consensus was
reached.
We considered RCTs including participants up to age 25 years
of age, diagnosed with a pervasive developmental disorder or
ASD according to the Diagnostic and Statistical Manual of Data synthesis and analysis
Mental Disorders, Fourth (DSM-IV) or Fifth editions (DSM-5),
or International Classification of Diseases, Tenth Revision For ADHD outcome synthesis, where more than one measure
(ICD-10) criteria. Eligible interventions included amphetamine for the same symptom domain was included, the stated
salts and methylphenidate-based stimulants, atomoxetine, primary outcome measure from each study was used for
alpha-2 adrenergic receptor agonists (guanfacine and cloni- meta-analyses. As in a previous review (Sturman et al., 2017),
dine), antipsychotics (all first- and second-generation antipsy- clinician or other investigator ratings were combined with
chotics listed in MEDLINE/Embase), tricyclic antidepressants teacher ratings in meta-analyses of ADHD symptoms, and
(all drugs under this category listed in MEDLINE/Embase), parent ratings were considered separately. For AEs, we pooled
bupropion, modafinil, or venlafaxine. We included studies that findings wherever possible, regardless of rater.
used placebo, behavioral therapies (e.g., parent training), or For crossover studies, we calculated standardized mean
any of the aforementioned medications as comparators. We differences (SMD) for continuous outcomes, including ADHD
excluded studies where one of the listed interventions was symptoms, using methods outlined in the Cochrane Handbook
used as an adjunctive treatment. We included studies evalu- for Systematic Reviews of Interventions (Higgins & Green,
ating our primary outcome of ADHD symptoms (hyperactivity/ 2011). For parallel studies, SMDs for continuous outcomes
impulsivity and inattention) at any follow-up time point and were calculated using Cohen’s d (Cohen, 1988). Effect sizes of
with any effect measure. Secondary outcomes included Clin- SMD = .2, .5, and .8 were considered to represent small,
ical Global Impression (CGI) rating scales (CGI-Improvement medium, and large effects, respectively (Cohen, 1988). For
[CGI-I] and CGI-Severity [CGI-S]), oppositional defiant disorder both crossover and parallel studies, we calculated mean
(ODD) symptoms, parent–child interaction, parent/caregiver differences (MD) where the same scale was used, indicating
stress, quality of life (QoL), cognitive functioning, and adaptive the absolute difference between groups on the scale used. For
functioning. Additional secondary outcomes were rates of crossover studies, we estimated or imputed correlation coef-
adverse events (AEs), serious AEs, and dropouts due to AEs. ficients in order to calculate standard errors that account for
the paired within-individual treatment comparisons (Elbourne
et al., 2002; Higgins & Green, 2011). Methods for estimating
Selection of studies and imputing correlation coefficients are detailed in the online
supplement (Appendix S2). We conducted sensitivity analyses
Retrieved citations were imported into Covidence for screening
examining the impact of different correlation coefficient values
(https://www.covidence.org/). One reviewer conducted title
on our findings. For dichotomous outcomes, such as AEs, risk
and abstract screening (RR), and two reviewers (RR and AB)
differences (RD) were estimated by calculating the proportion
each independently conducted full-text screening on every
of patients in each treatment and comparison group who
article retrieved. Articles were selected for inclusion at the full-
experienced the event/outcome and calculating the difference
text stage if they met our eligibility criteria. Disagreements
in proportions between groups. In studies where multiple
between reviewers (n = 64) were largely due to ambiguities in
fixed doses were administered, treatment effects across dose

© 2020 Association for Child and Adolescent Mental Health


4 Rebecca Rodrigues et al.

groups were combined into one average using established Lubetsky, 2000; Jahromi et al., 2009; Pearson et al.,
methods for parallel (Higgins & Green, 2011) and crossover 2013; Posey et al., 2007; Research Units on Pediatric
studies (Borenstein, Hedges, Higgins, & Rothstein, 2009).
Psychopharmacology (RUPP) Autism Network,
Continuous outcomes were pooled using the generic inverse
variance (for analyses with crossover studies) or inverse 2005), atomoxetine (4 studies; Table S3; Arnold
variance methods (for parallel studies), and dichotomous et al., 2006; Handen et al., 2015; Harfterkamp
outcomes were pooled using the Mantel-Haenszel method in et al., 2012, 2014; Lecavalier et al., 2018; McDougle
Review Manager, version 5.3. A random-effects model was et al., 2017; Smith et al., 2016; Tumuluru et al.,
applied to all pooled data. Effects with two-sided 95% confi-
2017; van der Meer et al., 2013), guanfacine (1
dence intervals (CI) excluding unity were considered statisti-
cally significant. study; Table S4; Scahill et al., 2015), aripiprazole (4
Heterogeneity was assessed using the I2 value with the studies; Table S5; Findling et al., 2014; Ichikawa
following cut-offs: 0%-25%=heterogeneity may not be impor- et al., 2017; Marcus et al., 2009; Owen et al., 2009;
tant; 25%–50% = moderate heterogeneity; 50%–75% =sub- Varni et al., 2012), risperidone (6 studies; Table S5;
stantial heterogeneity, and 75%-100%=considerable
Aman et al., 2008; Janssen Pharmaceutical K.K.,
heterogeneity (Higgins & Green, 2011). Post hoc analyses to
explore potential sources of heterogeneity were conducted, 2015; Kent et al., 2013; McDougle et al., 2005;
including a subgroup analysis of methylphenidate studies by Nagaraj, Singhi, & Malhi, 2006; Research Units on
dose, and pooling ADHD outcomes assessed using the same Pediatric Psychopharmacology (RUPP) Autism Net-
methods (i.e., same rater and scale). Given the small number of work, 2002; Shea et al., 2004; Troost et al., 2005,
studies in each intervention group (<10), neither publication
2006), and lurasidone (1 study; Table S5; Loebel
bias assessment nor risk-of-bias sensitivity analyses were
conducted. et al., 2016). Two studies were head-to-head trials of
aripiprazole versus risperidone (Table S5; Gha-
nizadeh, Sahraeizadeh, & Berk, 2014; Lamberti
Quality of evidence et al., 2016). An individual study investigating olan-
Quality of evidence of included studies was assessed for zapine versus haloperidol (Malone, Cater, Sheikh,
primary outcomes using the Grading of Recommendations Choudhury, & Delaney, 2001) and individual pla-
Assessment, Development and Evaluation (GRADE) Working cebo-controlled crossover trials of tianeptine (Nieder-
Group domains: study limitations, consistency of effect,
hofer, Staffen, & Mair, 2003) and venlafaxine
imprecision, indirectness, and publication bias. We rated the
quality of evidence as being high, moderate, low, or very low (Niederhofer, 2004) were also included. Given their
(Higgins & Green, 2011). Two authors (RR and AB) discussed small sample sizes (n ≤ 14), findings from these
the quality of evidence for ADHD outcomes within each domain three studies are presented in Tables S6–S12. No
and reached a consensus. GRADEpro software was used to studies examining amphetamines, clonidine, mod-
create summary of findings tables (https://gradepro.org/).
afinil, or bupropion met our inclusion criteria. Par-
ticipants across studies were primarily male (~80%).
Clinical interpretation of the evidence The mean age across studies ranged from 8 to
10 years. Intelligence quotient (IQ) of participants
Clinical recommendations and a figure depicting stepwise
suggestions for medication choice were drafted (by SHA) was not consistently reported across studies. Mean
following synthesis of the research evidence. This draft under- IQ of participants was 67 for methylphenidate stud-
went further review by M-CL. Drafts were then circulated to all ies (2 studies). Mean IQ of participants was 85 in
coauthors for discussion and revision. Where evidence gaps atomoxetine studies (3 studies). Studies of methyl-
were present, decisions were made regarding in text guidance
phenidate, guanfacine, atomoxetine, aripiprazole,
and each step of the stepwise medication choice pathway
based on collective clinical experience across coauthors (DAG, and risperidone reported inclusion of participants
PS, M-CL, EA, SHA) as well as consulting prior guidance in the with wide ranging intellectual ability (see Table S5
general ADHD treatment literature (i.e., without ASD). Pre- for reported data, including percentage of partici-
sented recommendations are based on consensus agreement pants reported to be in average IQ range, as well as
across coauthors.
borderline, mild/moderate, or severe range for intel-
lectual disability).
Risk of bias of included studies is summarized in
Results Figures S1 and S2. Risk of bias was unclear in
Selection of studies and study characteristics approximately half of studies for random sequence
generation, allocation concealment, and blinding
Our search retrieved 1,739 unique citations, of
domains. High risk of bias was most evident in
which 126 were reviewed as full-text (Figure 1).
‘other bias’ due to pharmaceutical funding (Heres
Twenty-five studies, published across 38 articles,
et al., 2006; Lundh, Lexchin, Mintzes, Schroll, &
met our inclusion criteria. Attempts to obtain miss-
Bero, 2017; Spielmans & Parry, 2010) for 2/4
ing data and unpublished reports are summarized in
atomoxetine studies (Arnold et al., 2006; Harfter-
Table S1. Characteristics of studies and participants
kamp et al., 2012), all aripiprazole studies (Findling
are summarized in Table 1; detailed summaries of
et al., 2014; Ichikawa et al., 2017; Marcus et al.,
individual study characteristics are available in the
2009; Owen et al., 2009), 3/6 risperidone studies
online supplement. Interventions from placebo-con-
(Janssen Pharmaceutical K.K., 2015; Kent et al.,
trolled studies included methylphenidate (4 studies;
2013; Shea et al., 2004), and the lurasidone study
Table S2; Ghuman et al., 2009; Handen, Johnson, &
(Loebel et al., 2016).

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 5

Figure 1 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flow diagram for study selection. ARI,
aripiprazole; ATX, atomoxetine; MPH, methylphenidate; RIS, risperidone. aTwenty-five studies were included that were published across
38 articles; b4 MPH studies were included that were published across 5 articles; c4 ATX studies were included that were published across
five articles

Evidence synthesis Methylphenidate


Findings for ADHD outcomes are summarized in Four studies provided data for ADHD outcomes
Figure 2. Forest plots for meta-analyses (Fig- (Ghuman et al., 2009; Handen et al., 2000; Pearson
ures S3–S5), GRADE summary of findings tables et al., 2013; RUPP, 2005). All four studies used a
(Tables S13–S19), secondary outcome findings from crossover design, and study duration was short
individual studies (Table S20), and sensitivity anal- (limited to 1–2 weeks). Three of four studies admin-
yses (Tables S22–S25) are presented in the online istered fixed doses (two to three dose levels). Treat-
supplement. ment effects were therefore averaged across each

© 2020 Association for Child and Adolescent Mental Health


6
Table 1 Characteristics of included RCTs and pooled samples

Methylphenidate Atomoxetine vs. Aripiprazole vs. Risperidone vs. Aripiprazole vs. Lurasidone vs. Guanfacine vs.
vs. placebo placebo placebo placebo risperidone placebo placebo

RCT features
No. RCTs 4 4 3 2 2 1 1
RCT design (no. RCTs) Crossover (4) Crossover (1), Parallel (3) Parallel (2) Parallel (2) Parallel (1) Parallel (1)
parallel (3)
Country (no. RCTs) United States (4) United States (3), United States (2), United States (1), Iran (1), Italy (1) United States (1) United States (1)
Netherlands (1) Japan (1) Canada (1)
Rebecca Rodrigues et al.

No. of sites 8 14 107 12 2 40 5


Industry funding 0/4 RCTs 2/4 RCTs 3/3 RCTs 1/2 RCTs 0/2 RCTs 1/1 RCT 0 /1 RCT
Setting Outpatient (3), Not reported Inpatient/ Outpatient (1), Outpatient (1), not Outpatient Outpatient
inpatient/ outpatient (1), inpatient/ reported (1)
outpatient (1) not reported (2) outpatient (1)
Treatment duration 1–2 weeks 6–10 weeks 8 weeks 8 weeks 8–24 weeks 6 weeks 8 weeks
Dose Range: Mean (SD): Mean (SD): Mean (SD): Mean (SD): 20 mg/day Mode:
0.125 to 0.50 mg/ 1.3 (0.3) mg/kg/ 8.8 (5.0) mg/day 1.7 (0.7) mg/day 6.0 (3.3) mg/day 3 mg/day
kg/dose daya aripiprazole, 1.4
(0.8) mg/day
risperidone
No. RCTs with ADHD 4 4 0 0 1 0 1
symptoms as primary
outcome
Participant features
N 117 237 408 180 103 150 62
Age, mean (SD), range 7.4 (2.2), 3–14 9.2 (2.7), 5–17 9.7 (3.1), 6–17 8.2 (2.6), 5–17 8.9 (3.5), 4–18 10.7 (3.0), 6–17 8.5 (2.3), 5–14
Male, n (%) 101 (86) 200 (84) 356 (87) 143 (79) 83 (81) 122 (81) 53 (85)
Diagnosis, n (%)
Autistic disorder 80 (68) 119 (50) 408 (100) 156 (87) 38 (65)b 150 (100) 51 (82)
Asperger’s 8 (7) 31 (13) 0 (0) 12 (7) 8 (13)b 0 (0) 2 (3)
PDD-NOS 27 (23) 85 (36) 0 (0) 11 (6) 9 (16)b 0 (0) 9 (15)
CDD 0 (0) 0 (0) 0 (0) 1 (1) 0 (0)b 0 (0) 0 (0)
No ASD 2 (2) 2 (1) 0 (0) 0 (0) 1 (2)b 0 (0) 0 (0)
IQ, mean (SD) 68.6 (31.0)c 86.3 (20.5) Not reported Not reported Not reported Not reported Not reported
Average, n (%) 1 (8)d Not reported 34 (37) 19 (12) 21 (34)
Borderline, n (%) 1 (8)d 22 (14)
Mild, n (%) 3 (23)d 32 (35)e 85 (54) 39 (63)
Moderate, n (%) 5 (38)d 14 (15)e
Severe, n (%) 3 (23)d 12 (13)e 31 (20)

ASD, autism spectrum disorder; CDD childhood disintegrative disorder; PDD-NOS, pervasive developmental disorder not otherwise specified; RCT, randomized controlled trial.
a
Pooled from 3/4 RCTs (Handen et al., 2015; Harfterkamp et al., 2012; McDougle et al., 2017) – Arnold et al., 2006 reported dose in mg/day (mean [standard deviation] final dose was 44.2
[21.9] mg/day).
b
Estimates include 1 of 2 RCTs (Ghanizadeh et al.2014).
c
Mean from 2 RCTs (RUPP2005 and Pearson et al.2013).
d
From 1 RCT (Handen et al., 2000).
e
1 of 3 RCTs reported IQ (Ichikawa et al.2017).

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 7

Figure 2 Effect sizes (pooled and individual studies) and quality of evidence for the effects of each intervention on ADHD symptoms. RCT
indicates randomized controlled trial; SMD standardized mean difference; CI confidence interval. aSMD not estimable due to reporting of
postintervention and change from baseline findings in each study, however, mean difference (MD) findings significant on the Aberrant
Behavior Checklist hyperactivity subscale (MD = 8.98, 95%CI = 12.02, 5.94, I2 = 20%, very low quality of evidence)

dose for these studies (Handen et al., 2000; Pearson consistently lower with low (as opposed to medium
et al., 2013; RUPP, 2005). Methylphenidate reduced or high-dose treatment; low = .11 to .21 mg/kg/
hyperactivity compared to placebo, with medium to dose, medium = .22 to .36 mg/kg/dose, high = .43
large effect sizes (parent-rated: SMD = .63, 95%CI to .6 mg/kg/dose; Figures S6-S8). In addition,
.95, .30, I2 = 60%, low quality of evidence; methylphenidate was associated with small improve-
teacher-rated: SMD = .81, 95%CI 1.43, .19, ments in ODD symptoms, though the 95%CI indi-
I2 = 86%, very low quality of evidence), and had cated a range of possible effects that crossed the line
small effects on inattention (parent-rated: of no effect (Figure S9), and large effects on CGI-I
SMD = .36, 95%CI .64, .07, I2 = 67%, low qual- and CGI-S scores (Figure S10).
ity of evidence; teacher-rated: SMD = .30, 95%CI
.49, .10, I2 = 57%, low quality of evidence).
Atomoxetine
Methylphenidate also had medium-sized effects on
total ADHD symptoms (parent-rated: SMD = .60, Four studies (6–10 weeks duration) compared the
95%CI .96, .23, I2 = 72%, very low quality of effects of atomoxetine versus placebo on ADHD
evidence; teacher-rated: SMD = .55, 95%CI 1.11, symptoms (Arnold et al., 2006; Handen et al.,
0.00, I2 = 92%, very low quality of evidence). Sub- 2015; Harfterkamp et al., 2012, 2014; McDougle,
group analysis indicated that effect sizes were 2017). Atomoxetine improved hyperactivity with

© 2020 Association for Child and Adolescent Mental Health


8 Rebecca Rodrigues et al.

small to medium effect sizes (parent-rated: Two aripiprazole studies used flexible dosing
SMD = .49, 95%CI .76, .23, I2 = 0%, low quality (Ichikawa et al., 2017; Owen et al., 2009), and one
of evidence; teacher-rated: SMD = .43, 95%CI had three fixed dose groups (Marcus et al., 2009);
.92, .06, I2 = 65%, low quality of evidence). Ato- therefore, we calculated a combined treatment effect
moxetine improved inattention with small to med- across the three dose groups for this study. Pooled
ium-sized effects shown (parent-rated: SMD = .54, findings from three studies of aripiprazole versus
95%CI .98, .09, I2 = 0%, low quality of evidence; placebo (Ichikawa et al., 2017; Marcus et al., 2009;
teacher/investigator-rated: SMD = .38, 95%CI Owen et al., 2009) showed that patients treated with
.75, .01, I2 = 40%, low quality of evidence). For aripiprazole experienced a decrease, with a medium
total ADHD symptoms, parent ratings from a single to large effect size, in parent-rated hyperactivity on
study demonstrated a medium to large effect the Aberrant Behavior Checklist (ABC) subscale
(SMD = .71, 95%CI 1.29, .13, I2 = N/A, low (SMD = .73, 95%CI .95, .51, I2 = 0%;
quality of evidence) and pooled teacher/investigator MD = 7.96, 95%CI 10.26, 5.66, I = 0%, low 2

ratings showed a small effect of atomoxetine quality of evidence). A randomized discontinuation


(SMD = .44, 95%CI .93, .06, I2 = 66%, very low study included children with ASD and irritability
quality of evidence). Removal of the crossover study that were responders to aripiprazole on irritability
from the pooled analysis did not change our findings outcomes. In this study, a decrease in ABC hyper-
(Table S24). In a double-blind extension, no signif- activity over 6–9 months was found in children who
icant difference was observed with atomoxetine were randomized to ongoing aripiprazole treatment
compared to placebo at 24 weeks, likely due to the compared to a switch to placebo (MD = 5.20, 95%
inclusion of only placebo responders in the compar- CI 10.20, .20, I2 = N/A; Findling et al., 2014).
ison group (Smith et al., 2016). Atomoxetine did not Aripiprazole treatment also improved emotional and
improve ODD symptoms (parent-rated SMD = .07, cognitive functioning on pediatric QoL scales; how-
95%CI .41, .27; teacher-rated SMD = .1, 95% ever, no effect was observed on social functioning or
CI .32, .13, Figure S11). A small effect of atomox- total scores (Figure S14). Aripiprazole treatment also
etine compared to placebo on the rate of positive reduced caregiver strain (Figure S15).
response on the CGI-I (very much or much improved) Risperidone improved parent-rated ABC hyperac-
was found (SMD = .19, 95%CI .05, .33, Figure S12). tivity in pooled findings from two studies (SMD = not
In cognitive tests assessing response inhibition, estimable; MD = 8.98, 95%CI 12.01, 5.94,
atomoxetine showed a small effect in improving I2 = 20%, very low quality of evidence; RUPP, 2002;
errors of commission (SMD = .36, 95%CI .74, Shea et al., 2004). We were unable to include three
.02), but not omission (Figure S13). similar studies in the meta-analysis because of
reporting differences; however, descriptive findings
indicated a significant improvement in hyperactivity
Alpha-2 agonists
with risperidone (Janssen Pharmaceutical K.K.,
In one available study (8 weeks duration; Scahill 2015; Kent et al., 2013; Nagaraj et al., 2006). Long-
et al., 2015), treatment with guanfacine immediate term findings from a double-blind discontinuation
release resulted in large effects on hyperactivity study in risperidone responders following a 24-week
(parent-rated: SMD = .93, 95%CI 1.45, .40, open-label trial showed no difference in hyperactivity
I2 = N/A, very low quality of evidence; investigator- scores in participants randomized to risperidone
rated: SMD = 1.35, 95%CI 1.91, .80, I2 = N/A, continuation (n = 12) versus placebo (n = 12) at
very low quality of evidence), inattention (parent- 32 weeks (Troost et al., 2005). We were unable to
rated: not measured; investigator-rated: pool available secondary outcomes (cognitive and
SMD = .81, 95%CI 1.33, .29, I2 = N/A, very adaptive functioning); see supplement for findings
low quality of evidence), and total ADHD symptoms (Table S20).
(parent-rated: not measured; investigator-rated: Individual studies that were limited in sample size
SMD = 1.20, 95%CI 1.75, .66, I2 = N/A, very (i.e., n = 59, 44, respectively) compared aripiprazole
low quality of evidence). Guanfacine improved CGI-I versus risperidone (Ghanizadeh et al., 2014; Lam-
response compared to placebo (RD = .41, 95%CI .2, berti et al., 2016) and lurasidone versus placebo
.61, and small effects were seen on some cognitive (Loebel et al., 2016). No significant difference was
measures (Table S20). observed on any ADHD outcome for either compar-
ison (very low quality of evidence; Figure 2).
Second-generation antipsychotics
Adverse events
With the exception of one trial of aripiprazole versus
risperidone (Lamberti et al., 2016), all antipsychotic In pooled findings from four studies (Ghuman et al.,
studies were designed to evaluate the treatment of 2009; B L Handen et al., 2000; Pearson et al., 2013;
significant irritability in children with ASD; hyper- RUPP, 2005), methylphenidate was associated with
activity was a secondary outcome and inattention an increased risk of dropout due to AEs (RD = .08,
was not evaluated. 95%CI .02, .17, I2 = 30%). No other drug was

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 9

associated with increased risk of discontinuation. No dose (.43–.6 mg/kg/dose) compared to medium
serious AEs were reported with methylphenidate. (.22–.36 mg/kg/dose) dose treatment. Uncertainty
Serious AEs were reported with atomoxetine (1 and heterogeneity in the effect estimates highlight
event; hospitalization for aggression; Arnold et al., the need for further research to determine superior-
2006), guanfacine (1 event; hospitalization for ity for high-dose treatment. Our findings are similar
aggression; Scahill et al., 2015), aripiprazole (2 to those in the recent Cochrane review of methyl-
events; 1 presyncope, 1 aggression; Marcus et al., phenidate treatment in ASD (Sturman et al., 2017).
2009), and lurasidone (5 events; 3 arm fractures, 1 In comparison with children with ADHD without
irritability, 1 appendicitis; Loebel et al., 2016). No ASD, the magnitude of methylphenidate effects on
serious AEs were reported with risperidone; how- total ADHD symptom scores may be lower in chil-
ever, there were 5 events reported as severe (1 dren with ASD (Storebø et al., 2015). Consistent with
hyperkinesia, 1 weight gain, 2 somnolence, 1 aggres- a prior systematic review (Mahajan et al., 2012), the
sive reaction with impaired concentration, and 1 current literature suggests that children with ASD
extrapyramidal disorder as a result of accidental may be more likely to discontinue short-term
overdose; Shea et al., 2004). methylphenidate treatment compared to placebo
Statistically significant findings from pooled and treatment. It is also important to note that AEs in
individual studies for specific AEs are summarized in reviewed studies may be underestimated compared
Table 2. Complete findings for all AEs are available to everyday practice as some trials excluded partic-
in Tables S26–S32. Methylphenidate was associated ipants that could not tolerate test doses of methyl-
with a higher risk of decreased appetite, sleep phenidate.
disturbance, and risk was higher with medium and Recently published studies examining efficacy of
high doses. Atomoxetine was associated with a atomoxetine in ASD enabled meta-analysis of the
higher risk of decreased appetite (RD = .21, 95%CI same (or greater, depending on the examined out-
.12, .31, I2 = 0%) and of abdominal discomfort come) number of studies for atomoxetine compared
(RD = .11, 95%CI .01, .22, I2 = 59%) and nausea to methylphenidate and including more participants.
(RD = .12, 95%CI .02, .25, I2 = 60%). A number of However, the overall quality of available evidence for
statistically significant AEs were observed with atomoxetine remains low. Our results were consis-
guanfacine (Table 2), however, the largest effects tent with those of Patra et al. (2019) showing
were observed for drowsiness (RD = .77, 95%CI .61, medium effects of atomoxetine on parent-rated inat-
.93, I2 = N/A) and fatigue (RD = .54, 95%CI .34, .74, tention. In contrast, effects on parent-rated hyper-
I2 = N/A). Second-generation antipsychotics were activity were smaller in the current study compared
associated with a number of adverse events, includ- to the previously published meta-analysis on this
ing gastrointestinal effects (vomiting, changes in topic (Patra et al., 2019). This difference may be due
appetite), drowsiness, tremor, drooling, and weight to the inclusion of two additional studies in our
gain (Table 2, Tables S29-S32). meta-analysis of atomoxetine effects on parent-rated
hyperactivity in ASD (Harfterkamp et al., 2012,
2014; McDougle, 2017), whereas our meta-analysis
Discussion of parent-rated inattention included the same stud-
We present an updated systematic review and meta- ies submitted to meta-analysis previously (Patra
analysis of the evidence for efficacy and tolerability of et al., 2019). Based on available evidence, effect
all pharmacological agents studied in RCTs for the sizes for treatment efficacy with atomoxetine were
treatment of ADHD symptoms in children and youth slightly smaller compared to methylphenidate and
with ASD. Our work is the first to synthesize the perhaps less clinically meaningful based on avail-
evidence across different medication classes using a able CGI-I data. One available study with a reason-
meta-analytic approach. With respect to efficacy, we able sample size, relative to other studies published
examined not only symptomatic improvement in across the ASD treatment literature (Ameis et al.,
inattention and hyperactivity, but also relevant 2018), provides support for the efficacy of guanfacine
functional outcomes. treatment on ADHD symptoms in children with
autism (Scahill et al., 2015).
Randomized, controlled data on the effects of
Summary of systematic review and meta-analysis
risperidone or aripiprazole versus placebo on hyper-
results
activity are limited to studies focused on treatment of
Current (low to very low quality) evidence, from significant irritability in children with ASD, where
studies of short duration (1–2 weeks), indicates that hyperactivity was a secondary outcome; therefore,
short-term methylphenidate treatment may have a results are exploratory and may not generalize to
clinically meaningful effect on parent and teacher- children with ASD and ADHD symptoms without
rated hyperactivity symptoms in school-age children significant irritability. Concerning metabolic, neuro-
with ASD and variable IQ. Subgroup analysis by logical, and other side effects are also reasons for
dose indicates lower effect sizes with low dose caution regarding the use of antipsychotic medica-
treatment. Observed effect sizes are larger for high tions for treatment of ADHD symptoms (Ameis et al.,

© 2020 Association for Child and Adolescent Mental Health


10 Rebecca Rodrigues et al.

Table 2 Summary of statistically significant adverse events from pooled and individual RCTs

Comparison Outcome No. RCTs and sample size RD (95% CI)a I2

Methylphenidate vs. placebo Decreased appetite 2 RCTs (90 patients) Low dose: 0.12 ( 0.15, 0.38) 84%
2 RCTs (101 patients) Medium dose: 0.23 (0.13, 0.33) 0%
2 RCTs (101 patients) High dose: 0.23 (0.11, 0.35) 14%
Sleep disturbance 2 RCTs (90 patients) Low dose: 0.09 (0.01, 0.17) 0%
2 RCTs (90 patients) Medium dose: 0.18 (0.09, 0.27) 0%
2 RCTs (74 patients) High dose: 0.15 (0.05, 0.25) 0%
Atomoxetine vs. placebo Decreased appetite 4 RCTs (301 patients) 0.21 (0.12, 0.31) 0%
Guanfacine vs. placebo Drowsiness 1 RCT (62 patients) 0.77 (0.61, 0.93) N/A
Fatigue 1 RCT (62 patients) 0.54 (0.34, 0.74) N/A
Decreased appetite 1 RCT (62 patients) 0.37 (0.17, 0.57) N/A
Emotional/tearful 1 RCT (62 patients) 0.31 (0.10, 0.51) N/A
Dry mouth 1 RCT (62 patients) 0.37 (0.18, 0.55) N/A
Irritability 1 RCT (62 patients) 0.27 (0.07, 0.47) N/A
Anxiety 1 RCT (62 patients) 0.27 (0.09, 0.44) N/A
Mid-sleep awakening 1 RCT (62 patients) 0.24 (0.05, 0.42) N/A
10-point drop in systolic BP 1 RCT (62 patients) 0.25 (0.02, 0.49) N/A
Aripiprazole vs. placebo Vomiting 3 RCTs (405 patients) 0.06 (0.00, 0.11) 0%
Decreased appetite 2 RCTs (308 patients) 0.07 (0.02, 0.12) 0%
Increased appetite 2 RCTs (313 patients) 0.07 (0.01, 0.14) 0%
EPS-related AEs 3 RCTs (405 patients) 0.07 (0.01, 0.12) 0%
Tremor 2 RCTs (313 patients) 0.10 (0.05, 0.14) 0%
Drooling 2 RCTs (313 patients) 0.09 (0.05, 0.13) 0%
Pyrexia 2 RCTs (313 patients) 0.08 (0.04, 0.13) 0%
Weight change (kg) 3 RCTs (405 patients) MD: 0.97 (0.54, 1.40) 0%
BMI change (kg/m2) 3 RCTs (405 patients) MD: 0.40 (0.18, 0.63) 0%
Risperidone vs. placebo Increased appetite 4 RCTs (314 patients) 0.25 (0.12, 0.38) 61%
Drowsiness 4 RCTs (314 patients) 0.38 (0.07, 0.68) 93%
Tremor 2 RCTs (179 patients) 0.11 (0.04, 0.18) 0%
Tachycardia 2 RCTs (179 patients) 0.11 (0.04, 0.19) 0%
Weight gain 3 RCTs (214 patients) 0.10 (0.03, 0.17) 0%
Weight change (kg) 3 RCTs (274 patients) MD: 1.45 (0.97, 1.93) 16%
Lurasidone vs. placebo Vomiting 1 RCT (148 patients) 0.14 (0.05, 0.23) N/A
Nasopharyngitis 1 RCT (148 patients) 0.08 (0.02, 0.14) N/A
Akathisia 1 RCT (148 patients) 0.06 (0.00, 0.12) N/A
Weight gain (kg) 1 RCT (148 patients) MD: 0.80 (0.25, 1.35) N/A

BMI, body mass index; BP, blood pressure; CI, confidence interval; EPS, extrapyramidal symptoms; MD, mean difference; RCT,
randomized controlled trial; RD, risk difference.
a
Positive risk difference favors intervention.

2013; Pagsberg et al., 2017). Nonetheless, pooled For risperidone, we were unable to include three
findings from available data indicate large effects of studies of similar design in the meta-analysis
treatment on parent-rated hyperactivity. because of missing data. Given the small number of
studies in each intervention and outcome group, we
were unable to assess publication bias with funnel
Limitations of our systematic review and meta-
plots and therefore cannot rule it out. In interpreting
analysis
findings from studies including parent and teacher-
We observed substantial heterogeneity in the meta- based outcomes, it is notable that although point
analysis of methylphenidate studies; however, inves- estimates for SMD were overall similar between
tigation of dose and rating scales suggests that the parent and teacher-based outcomes, precision was
heterogeneity may be partially explained by these consistently lower for teacher-based outcomes and
factors. For continuous outcomes from crossover the range of potential effects measured by the confi-
studies, we imputed correlation coefficients to calcu- dence interval crossed the line of no effect.
late SMDs and standard errors. Since imputed cor-
relation coefficients can impact the magnitude of
Evidence gaps and implications for future research
SMDs, use of this method could impact our findings.
However, results from our sensitivity analyses exam- Taken together, the quality of evidence is low to very
ining the impact of different correlation coefficient low for ADHD treatment studies in ASD. Evidence for
values (r = 0, .5, .8) did not change our findings. We long-term medication treatment is limited, and evi-
were unable to impute correlation coefficients for dence for effects on real-world function (e.g., adaptive,
binary outcomes and analyzed these data as inde- cognitive, academic) is minimal or altogether lacking
pendent groups; as a result, we likely underesti- in the current literature. As well, future studies
mated AEs with methylphenidate and atomoxetine. should evaluate the impact of pharmacotherapy on

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 11

academic performance and school adjustment, as this medication classes on ADHD symptoms as primary
is an important outcome for children and families that outcome in ASD (and five of the total available RCTs
has been overlooked in studies to date. examine three different medication classes, all of
A number of the available atomoxetine and which include samples of >60 participants; Posey
antipsychotic studies were industry supported. et al., 2007; Harfterkamp et al., 2012; Handen et al.,
Although we included industry funding in the 2015; Scahill et al., 2015; McDougle 2017). Nonethe-
‘other’ domain of the risk-of-bias assessment, a less, as the overall quality of the evidence base
recent review concluded that industry funding remains low, clinical opinions will differ on how
results in bias that cannot be explained (or much to lean on the limited data available in the ASD
accounted for) by standard risk-of-bias tools (Lundh population versus the much larger evidence base in
et al., 2017). Future long-term efficacy studies that the non-ASD, ADHD population. Here, we provide a
can provide information on whether initial treatment stepwise visual practical clinical guide to accompany
effects are maintained or enhanced in children with the current resource outlining baseline symptom
ASD with longer-term treatment are needed. Exam- evaluation, implementation of behavioral interven-
ination of whether clinical effects translate into tions, and subsequent medication treatment choice,
improvement in real-world functioning is also criti- if a medication is determined to be indicated by the
cal. Core outcome sets that include measures that treating clinician (Figure 3). The provided pathway
track functional impact (e.g., adaptive, executive offers guidance for medication choice across chil-
functioning) are needed to compare results across dren and youth with co-occurring ASD and ADHD.
studies and the impact of treatment on functional Of note, although the mean age for participants
outcome (McConachie et al., 2015). As further trials included in RCTs reviewed above was 8–10 years,
become available (which are needed across more many studies (across medication classes) included
pharmacological agents, e.g., amphetamines), net- adolescent participants (e.g., oldest participants
work meta-analysis may help to guide clinical deci- ranged from 12 to 18 years of age, Tables S1–S5).
sion-making by providing comparisons of different The provided pathway is not meant to take the place
interventions that have not been previously studied of other ADHD assessment and care guidelines. It
using head-to-head comparison trials (Cortese et al., does not list all clinical precautions requiring careful
2018; Tonin et al., 2018). evaluation prior to initiating a medication treatment
for ADHD symptoms, nor adverse effects experienced
with listed medication treatments. Rather, the path-
Practical guidance I: Clinical interpretation of way aims to provide a visual guide to accompany in
the synthesized evidence base text suggestions regarding medication choice.
The current recommendations are based on a con- Evaluation of best practices for ADHD symptom
sensus among coauthors that integrates (a) the assessment in ASD was beyond the scope of our
expanded empirical literature focused on medication synthesis of the treatment literature. Here, we
treatment of ADHD symptoms in ASD included in provide a brief summary of recommendations
our systematic review and meta-analysis, (b) evi- regarding evaluation of ADHD symptoms in ASD
dence from clinical guidelines and treatment studies but refer the reader to the 2012 ATN Symptom
of ADHD without ASD to support guidance when Evaluation Practice Pathway for further guidance
there are less/poor data to draw from in the ASD (Mahajan et al., 2012) as well as to a recent publi-
literature, and (c) clinical experience from expert cation from the United Kingdom ADHD Partnership
members of our research team (M-CL, DAG, EA, PS, summarizing consensus opinion focused on assess-
SHA). While the quality of the evidence for ADHD ment of ASD and ADHD and nonpharmacological
treatment studies in ASD (based on our evidence intervention approaches (Young et al., 2020). These
synthesis) remains low to very low, it is important to references provide clear, stepwise clinical guidance
note the advances that have been made in the on best practices for assessing ADHD symptoms in
literature since the publication of the 2012 ATN ASD, evaluation of medical and academic problems
Clinical Practice Pathways that provide clinical that may contribute to ADHD symptoms, and edu-
guidance for assessment and treatment of ADHD cational/behavioral interventions that should be
symptoms in ASD (Mahajan et al., 2012). For exam- implemented (particularly in the school setting) prior
ple, the 2012 ATN Practice Pathways were based on to initiating medication treatment (Mahajan et al.,
expert opinion, the general ADHD treatment litera- 2012; Young et al., 2020). These references are also
ture, and a systematic review of five RCTs examining in line with the 2018 NICE guidance for ADHD
efficacy of medication treatments on ADHD symp- medication management in children and youth in
toms in ASD as a primary outcome (Mahajan et al., the general population (5 years or older), with
2012). This included just one study (exploring effects respect to moving toward medication management
of methylphenidate) with a participant sample only when significant ADHD symptoms persist fol-
greater than twenty (Posey et al., 2007). We now lowing environmental modifications (National Insti-
have meta-analytic level evidence from eight different tute for Health & Care Excellence, 2018).
RCTs evaluating the efficacy of two different Environmental modifications for ADHD symptoms

© 2020 Association for Child and Adolescent Mental Health


12 Rebecca Rodrigues et al.

include strategies such as selection of seating in training plus placebo was beneficial over placebo
class to optimize engagement and reduce distrac- alone for the treatment of ADHD symptoms and
tion, reducing distraction through the use of head- noncompliance at week 10 of the double-blind RCT
phones/ear plugs or other personalized strategies (effect size range = 0.46–0.6; Handen et al., 2015). In
helpful to the individual, appropriate access to addition, combined treatment with parent training
teaching assistants to help with schoolwork, fre- and atomoxetine was also superior to the beneficial
quent breaks for physical movement paired with effects of atomoxetine alone on ADHD symptoms and
shorter duration of focus, and relaying expectations noncompliance, following an open-label extension of
using both verbal and visual or written supports the double-blind atomoxetine study (Smith et al.,
(National Institute for Health & Care Excellence, 2016). Thus, the available literature suggests that
2018). Recent evidence in children with ADHD in the psychosocial interventions can have a meaningful
general population indicates that functional out- impact on ADHD symptoms and noncompliance in
come may be better when treatment is initiated with ASD, as in the general ADHD population.
behavioral intervention, followed by medication, as
opposed to the other way around (Pelham et al.,
2016). Although studies that combine behavioral Practical guidance II: Clinical pathway
interventions with medication treatment did not recommendations
meet the inclusion criteria for our systematic review, First, given the quality of the available evidence base,
such studies are of interest for clinical practice. We a shared decision-making model between clinicians,
are aware of two clinical trials that compared the patients, and their caregivers/families is encouraged
effect of treatment with behavioral interventions plus that adjusts the pathway for a given child/youth
medication versus medication treatment alone on (including the order of medication class/preparation
ADHD and noncompliance symptoms in ASD (Aman chosen) to best support the patient and accommo-
et al., 2009; Handen et al., 2015). The study by date the approach to fit their socio-ecological con-
Aman and others compared the effect of combined texts (i.e., familial/caregiving factors, school setting,
treatment with manualized parent training plus etc.). The first phase of intervention begins with
risperidone to risperidone treatment alone on every- evaluation of ADHD symptoms using structured
day noncompliant behavior in children with ASD and questionnaires [e.g., Connor’s Rating Scale (Wolraich
significant irritability (n = 124). Parent training con- et al., 2003), SWAN rating scale (Brites, Salgado-
sisted of 11, 60–90 min treatment sessions, deliv- Azoni, Ferreira, Lima, & Ciasca, 2015)]. Additional
ered individually to parents and focused on the use use of a structured measure such as the Clinical
of visual schedules, positive reinforcement, teaching Global Impression (CGI) Scale provides an easily
functional communication, and adaptive skills. implemented baseline measure of a patient’s global
Combined treatment was superior to risperidone functioning prior to initiating a medication treatment
treatment alone on the ABC hyperactivity/noncom- (using the CGI-Severity, CGI-S) that can also be used
pliance scale (effect size, d = 0.55) with larger effect to track progress in later phases of the pathway
sizes found by treatment week 24 (compared to week (using the CGI-Improvement, CGI-I, version of the
16; Aman et al., 2009; Handen et al., 2013). The scale (Busner & Targum, 2007). Medical evaluation
study by Handen et al. (2015) that was included in to assess for clinical conditions suggestive of the
our quantitative synthesis, based on comparison of need for consultation from other health profession-
atomoxetine to placebo treatment (Handen et al., als (e.g., cardiology, based on the presence of base-
2015), also included combined parent training plus line cardiovascular risk prior to initiating medication
atomoxetine and parent training plus placebo arms treatment) is essential (Subcommittee on Attention-
(using the same manualized approach as in Aman Deficit/Hyperactivity Disorder et al., 2011; Warren
et al., 2009). While this study did not find a bene- et al., 2009). Baseline documentation of vital signs
ficial effect of atomoxetine plus parent training and symptoms that can be exacerbated with ADHD
compared to atomoxetine treatment alone, parent medication treatment or experienced as side effects

Figure 3 Medication choice pathway for children and youth with ADHD and ASD. (1) Is clinically significant ADHD present using
structured questionnaires?: for example, Connor’s Rating Scale, SWAN, etc (reference: Wolraich et al., 2003; https://www.ncbi.nlm.
nih.gov/pubmed/23363973); Clinical Global Impressions-Severity (CGI-S) to measure baseline symptom severity (https://www.ncbi.nlm.
nih.gov/pmc/articles/PMC2880930/); (2) Evaluate baseline symptoms, co-occuring diagnoses, medical conditions. Document baseline
symptoms: sleep, appetite, any symptoms of psychosis, tics, mood lability, other psychiatric diagnoses. Document blood pressure, heart
rate, height, weight. Document/address medical conditions as per Warren et al., 2009 (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC2776560/) AAP 2011 Guidelines for ADHD (https://pediatrics.aappublications.org/content/pediatrics/128/5/1007.full.pdf) and Mahajan
et al., 2012 (https://www.ncbi.nlm.nih.gov/pubmed/?term=23118243); (3) Quick assessment and evaluation every 2 weeks: for example,
ADHD assessment—structured questionnaires; vital signs: heart rate, blood pressure, height, weight, sleep, appetite, mood, GI effects,
tics, medication compliance, side effects (note 4); Clinical Global Impressions-Improvement (CGI-I) guidelines (https://www.ncbi.nlm.
nih.gov/pmc/articles/PMC2880930/); (4) Intolerable side effects: for example, hypertension, tachycardia, extreme weight loss or gain,
insomnia, suicidal ideation, extreme mood swings

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 13

Figure 3. Medication Choice Pathway for Children and Youth with ADHD and ASD

Evaluate:
Baseline symptoms, co- Behavioural
Evaluate: Yes occuring diagnoses, Supports: Yes
Screening Phase
Is clinically significant
medical conditions, Are recommended
(~0-8 Weeks)
ADHD present on clinical Medication
contraindications to behavioural supports Choice Pathway
assessment & structured
medication treatment in place?
questionnaires?
(1) (2)

No
No 8 Week
Behavioural
Exit Pathway Intervention If clinically significant ADHD symptoms persist, consider:

if ADHD symptoms disappear

Start First-line Treatment with


Stimulant Monotherapy
1st choice: methylphenidate
monotherapy (immediate or extended
release)

Quick Assessment and


Evaluation, every 2 weeks
e.g CGI-I >= 3 e.g. CGI-I = 1 or 2
(3)
(~8-23 Weeks)
First -Line
Treatment

Partial or No Response
Side Effects Limit Ongoing
Full Response
Treatment [consider (4)]

consider monotherapy with


Second-line treatment option: Continuation of dosage and
atomoxetine, alpha-2 agonist behavioural supports;
Switch to another (guanfacine or clonidine, consider switching to less
methylphenidate preparation, immediate or extended frequent follow-up
Titrate
release) or amphetamine
to effective dose
(when inadequate response or
mild-to-moderate side effects
limit first-line methylphenidate
treatment). Choice based on
patient/family/environment/
context of care factors

Quick Assessment and


Evaluation, every 2 weeks e.g. CGI-I= 1 or 2
e.g. CGI-I>= 3
(3)
(~23-38 Weeks)

Side Effects Limit Ongoing


Second-Line

Partial or No Response Full Response


Treatment

Treatment [consider (4)]

Continuation of dosage
Switch to another second- and behavioural supports;
Titrate
line option not previously consider switching to less
to effective dose
used frequent follow-up

Quick Assessment and


Evaluation, every 2 weeks
e.g. CGI-I >=3 (3) e.g. CGI-I= 1 or 2
(~32+ Weeks)
Final Phase

Partial or No Response Side Effects Limit Ongoing


Treatment [consider (4)]

Full Response
Reassessment, Need
for Complex Care

© 2020 Association for Child and Adolescent Mental Health


14 Rebecca Rodrigues et al.

of treatment is critical for later evaluation of tolera- 2009). One study found that this risk is greater with
bility of a given medication, once initiated (i.e., sleep, amphetamines than methylphenidate (2.83 episodes
appetite, presence of tics, psychosis, suicidality, per 1,000 person-years vs. 1.78 episodes per 1000
mood dysregulation/irritability, blood pressure, person-years, hazard ratio = 1.65 [95%CI 1.31–2.09];
heart rate, weight, height; Storebø et al., 2015). Moran et al., 2019). The evidence is mixed, however,
Based on the present updated systematic review and with other studies finding that methylphenidate is not
meta-analysis, and consistent with prior guidance, associated with an increased risk of psychosis in
the current evidence base supports first-line methyl- ADHD and may even have a protective effect (Hollis
phenidate monotherapy, based on efficacy evidence et al., 2019; Man et al., 2016). As ASD itself is an
in ASD as well as general ADHD treatment guidance independent risk factor for later psychotic disorders
(Cortese et al., 2018; Mahajan et al., 2012; National in youth and adulthood (Lai et al., 2019), clinicians
Institute for Health & Care Excellence, 2018; Padilha should be aware of the potential risk for psychosis
et al., 2018). As recommended previously, system- with stimulant treatment and carefully document risk
atic and cautious titration by small increments with for psychosis at baseline (e.g., individual and family
frequent reassessment and dose adjustment and history of psychosis or mania/hypomania).
more frequent monitoring for treatment of ADHD Previous guidance for treatment of ADHD symp-
symptoms may improve tolerability in children and toms in ASD has suggested that when inadequate
youth with ASD (Mahajan et al., 2012; National response or mild-to-moderate side effects limit ongo-
Institute for Health & Care Excellence, 2018). The ing treatment with methylphenidate preparations, an
optimal dose level will aim to maximize therapeutic amphetamine preparation is recommended as the
benefits while producing minimal adverse events. next medication choice (Mahajan et al., 2012). It is
Although more evidence is available for short than important to note that expert opinion may differ at
long-acting methylphenidate in the ASD population, this decision point regarding the next best medica-
preparation choice may be tailored to practical tion choice after a failed methylphenidate trial (i.e.,
needs, such as the patient’s and family’s prefer- when treatment with methylphenidate is not effective
ences, the need for increased flexibility with short- or side effects preclude continued use). The current
acting or dosing convenience with longer-acting resource considered (a) potential for reduced tolera-
preparations (National Institute for Health & Care bility to stimulants in children and youth with ASD
Excellence, 2018). and ADHD (compared to ADHD alone), (b) increased
For the general management of ADHD, a trial of evidence of efficacy for other medication classes
amphetamines (specifically Lisdexamphetamine) is (atomoxetine and alpha-2 agonist) in children and
recommended following inadequate response to a 6- youth with ASD and ADHD since the prior medication
week trial of methylphenidate (National Institute for choice pathway (Mahajan et al., 2012) was published
Health & Care Excellence, 2018). As yet, there are no (though evidence quality remains low to very low),
RCT data supporting the use of amphetamines for and (c) lack of studies examining efficacy and toler-
management of ADHD in children/youth with ASD. In ability of amphetamines in children and youth with
children with ADHD without ASD, the evidence base ASD and ADHD balanced with evidence of efficacy
indicates that amphetamine treatment has large but reduced tolerability in the non-ASD, ADHD
effects on ADHD symptoms, and moderate effects on population (Cortese et al., 2018; Padilha et al.,
academic performance and similar side effect profiles 2018). Based on these considerations, it is our expert
as with methylphenidate (Cortese et al., 2018; Padilha consensus opinion that second-line treatment
et al., 2018; Punja et al., 2016). While monotherapy options for children and youth with ASD and ADHD
with amphetamines may be considered in ASD, it is include both nonstimulant medication treatment
important for clinicians to be aware that network options with atomoxetine or an alpha-2 agonist or a
meta-analyses comparing ADHD treatments in the trial of amphetamine treatment (when inadequate
ADHD without ASD literature suggest that overall response or mild-to-moderate side effects limit ongo-
tolerability is lower with amphetamines compared to ing treatment with methylphenidate preparations).
methylphenidate (Cortese et al., 2018; Padilha et al., Updated evidence provides more confidence in
2018), particularly for sleep disturbance, appetite choosing a trial of either monotherapy with atomox-
disturbance, and irritability (Padilha et al., 2018). etine or guanfacine as a second-line option following
This needs to be considered alongside indication from a failed methylphenidate trial. Although head-to-
the current evidence base that tolerability to methyl- head comparison trials of guanfacine and atomox-
phenidate is lower in ASD than in the ADHD without etine are not available in the ADHD and ASD
ASD population (McCracken, 2004). In addition, population, drawing from one head-to-head trial
although higher rates of serious side effects with and network meta-analysis from the ADHD without
stimulant treatments have not been shown consis- ASD literature, tolerability may be lower with guan-
tently in RCTs (Punja et al., 2016; Storebø et al., 2018; facine (including with the extended release prepara-
Storebø et al., 2015), in general, stimulants have been tion) or amphetamine treatment than with
found to be associated with a small risk of psychotic atomoxetine (Cortese et al., 2018). A prior network
symptoms in children with ADHD (Mosholder et al., meta-analysis comparing different ADHD

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 15

medications on a variety of safety outcomes (e.g., Appendix S2. Methods for data synthesis and analysis.
sleep, decreased appetite, and behavior events such Table S1. Attempts to contact study authors for
as irritability, anxiety, aggression) provides helpful unpublished or missing data and outcomes.
clinical information regarding which medication may Table S2. Characteristics of included studies compar-
be the best choice for a given child based on their ing methylphenidate vs. placebo.
particular circumstance or side effect sensitivity Table S3. Characteristics of included studies compar-
Padilha et al.,2018. Clinicians will need to evaluate ing atomoxetine vs. placebo.
the current evidence base with families, consider Table S4. Characteristics of the included alpha-2
agonist study.
personalized child, familial and environmental (e.g.,
Table S5. Characteristics of included antipsychotic
school) factors, the context of care (e.g., ability to
studies.
monitor response and tolerability with frequent
Table S6. Characteristics of included small, single
follow-ups), side effect, administration and discon- studies.
tinuation profiles of each second-line option and Table S7. Summary of results for the effect of olanza-
ultimately undertake a shared decision-making pro- pine, tianeptine, and venlafaxine on ADHD symptoms.
cess to support choice among second-line options for Table S8. Summary of findings for olanzapine vs.
a given patient. haloperidol.
Of note, second-line options from the ATN medica- Table S9. Summary of findings for tianeptine vs.
tion choice pathway (Mahajan et al., 2012) include placebo.
atomoxetine and either guanfacine or clonidine as Table S10. Summary of findings for venlafaxine vs.
second-line alpha-2 adrenergic agonists. There are no placebo.
trials of clonidine in children with both ADHD and Table S11. Adverse events from small, single studies
ASD. Although selectivity for alpha-2 adrenergic sub- (olanzapine vs. haloperidol, tianeptine vs. placebo, and
types differs with clonidine versus guanfacine, based venlafaxine vs. placebo).
on the general ADHD literature, these medications Table S12. Sensitivity analysis for different correlation
may have similar efficacy and tolerability (Hirota, coefficient values for ADHD outcomes from the individ-
ual tianeptine and venlafaxine studies.
Schwartz, & Correll, 2014; with increased risk for
Table S13. Summary of findings for methylphenidate
sedation, somnolence and fatigue, hypotension and
vs. placebo.
bradycardia with immediate or extended release ver-
Table S14. Summary of findings for atomoxetine vs.
sions of either medication), though hypotension and placebo.
bradycardia may be more common with immediate Table S15. Summary of findings for guanfacine vs.
release clonidine, and QTc prolongation with extended placebo.
release guanfacine (Hirota et al., 2014). Therefore, if Table S16. Summary of findings for aripiprazole vs.
guanfacine is not available, or an extended release placebo.
formulation of either alpha-2 agonists are not avail- Table S17. Summary of findings for risperidone vs.
able (for easier administration), or cost prohibits the placebo.
use of one formulation, clonidine could be used as a Table S18. Summary of findings for aripiprazole vs.
second-line treatment option, in place of guanfacine. risperidone.
Given risk of rebound hypertension, tachycardia, and Table S19. Summary of findings for lurasidone vs.
arrhythmias with abrupt discontinuation, alpha-2 placebo.
agonists should be tapered gradually prior to discon- Table S20. Summary of findings for secondary out-
tinuation (Scahill et al., 2015). comes from single studies.
Importantly, as there is limited evidence with Table S21. Summary of results for ADHD symptoms
respect to the risk and benefit of long-term mainte- measured on the same scale.
nance, careful evaluation of the clinical impact of Table S22. Sensitivity analysis of different correlation
coefficient values for outcomes from methylphenidate
treatment on ADHD symptoms, global functioning,
studies.
and quality of life across settings (e.g., school, home,
Table S23. Sensitivity analysis for pooled atomoxetine
community) is required. Therefore, ongoing evalua- ADHD findings, in which the teacher-rated outcomes
tion of the need for continuous pharmacological (from Handen et al.10) were removed from the teacher/
treatment has to be an integral component of ongoing investigator-rated ADHD outcomes.
care of children and youth with ASD and ADHD with Table S24. Sensitivity analysis for pooled atomoxetine
careful consideration of the potential benefits weighed ADHD findings, in which the crossover study (Arnold
against the limitations of safety/efficacy data. et al.9) was removed.
Table S25. Sensitivity analysis for different correlation
coefficient values for outcomes including the atomox-
Supporting information etine crossover study9.
Additional supporting information may be found online Table S26. Adverse events with methylphenidate ver-
in the Supporting Information section at the end of the sus placebo.
article: Table S27. Adverse events with atomoxetine versus
placebo.
Appendix S1. Medline search strategy.

© 2020 Association for Child and Adolescent Mental Health


16 Rebecca Rodrigues et al.

Table S28. Adverse events with guanfacine versus Figure S12. Forest plot for meta-analysis of the effect of
placebo. atomoxetine on obtaining a positive responses on the
Table S29. Adverse events with aripiprazole versus Clinical Global Impression-Improvement (CGI-I) scale.
placebo. Figure S13. Forest plot for meta-analysis of effects of
Table S30. Adverse events with risperidone versus atomoxetine on cognitive tasks assessing response
placebo. inhibition including the continuous performance task.
Table S31. Adverse events with aripiprazole versus (A) Errors of commission and (B) errors of omission.
risperidone. Figure S14. Forest plot for meta-analysis of the effect of
Table S32. Adverse events with lurasidone versus aripiprazole on pediatric quality of life.
placebo. Figure S15. Forest plot for meta-analysis of the effect of
Figure S1. Review authors’ judgments about each risk- aripiprazole on caregiver strain, measured with the
of-bias item, presented as percentages across all Caregiver Strain Questionnaire.
included studies.
Figure S2. Review authors’ judgments about each risk-
of-bias item for each included study. Acknowledgements
Figure S3. Forest plots for meta-analysis of the effects This work was supported in part by two Academic
of methylphenidate on ADHD symptoms, including (A) Scholars Awards from the Department of Psychiatry,
hyperactivity, (B) inattention, and (C) total ADHD University of Toronto (to S.H.A. and M-C.L.), by the
symptoms. O’Brien Scholars Program, Slaight Family Child and
Figure S4. Forest plots for meta-analysis of the effects Youth Mental Health Innovation Fund and The Cather-
of atomoxetine on ADHD symptoms, including (A) ine and Maxwell Meighen Foundation via the CAMH
hyperactivity, (B) inattention, and (C) total ADHD Foundation (to S.H.A. and M-C.L.), and an Ontario
symptoms. Mental Health Foundation New Investigator Fellowship
Figure S5. Forest plots for meta-analysis of the effects (to S.H.A.). E.A. is supported by funding from CIHR,
of antipsychotics on hyperactivity, including aripipra- NIH, DoD, Ontario Brain Institute, Brain Canada,
zole, (A) standardized mean difference and (B) mean Genome Canada, Ontario Research Fund, Azrieli Foun-
difference, and (C) risperidone, mean difference. dation, Autism Speaks, HRSA, Canada Research Chairs
Figure S6. Subgroup analysis of parent- and teacher- Program, and Dr. Sims Chair in Autism. The authors
rated hyperactivity by methylphenidate dose. thank John Costella, Associate Librarian at Western
Figure S7. Subgroup analysis of parent- and teacher- University, for his guidance in developing the search
rated inattention by methylphenidate dose. strategy. The authors also thank Caroline Kassee and
Figure S8. Subgroup analysis of parent- and teacher- Dr. Amanda Sawyer for their help with developing the
rated total ADHD symptoms by methylphenidate dose. medication choice pathway figure. E.A. has received
Figure S9. Forest plot for meta-analysis of the effects of consultation fees from Roche, in kind support from
methylphenidate on oppositional defiant disorder AMO Pharma, and editorial honorarium from Wiley. The
(ODD) symptoms. remaining authors have declared that they have no
competing or potential conflicts of interest.
Figure S10. Forest plots for meta-analysis of the effect
of methylphenidate on Clinical Global Impression
(CGI)-Improvement (CGI-I) and CGI-Severity (CGI-S),
including (A) standardized mean difference and (B) Correspondence
mean difference. Stephanie H. Ameis, Centre for Addiction and Mental
Figure S11. Forest plots for meta-analysis of the effect Health, 80 Workman Way, Toronto, ON, M6J 1H4, USA;
of atomoxetine on oppositional defiant disorder (ODD) Email: Stephanie.ameis@camh.ca
symptoms.

Key points

 Clinically significant attention-deficit/hyperactivity disorder (ADHD) symptoms are common and impairing in
children and youth with autism spectrum disorder (ASD).
 This systematic review and meta-analysis sought to update and integrate the available evidence regarding
the pharmacological treatment of ADHD in ASD.
 Methylphenidate, atomoxetine, and guanfacine had small to large effects on hyperactivity and inattention
in children with autism spectrum disorder, based on low/very low quality of evidence.
 Indirect, low quality evidence indicated aripiprazole and risperidone were efficacious in reducing hyperactive
symptoms.
 Current evidence for treatment of ADHD symptoms in ASD is limited; however, methylphenidate remains the
first-line treatment option and emerging evidence for atomoxetine and guanfacine indicates these
treatments may reduce ADHD symptoms in ASD.

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 17

Cohen, D., Raffin, M., Canitano, R., Bodeau, N., Bonnot, O.,
References Perisse, D., . . . & Laurent, C. (2013). Risperidone or arip-
Aman, M.G., Hollway, J.A., McDougle, C.J., Scahill, L., Tier- iprazole in children and adolescents with autism and/or
ney, E., McCracken, J.T., . . . & Posey, D.J. (2008). Cognitive intellectual disability: A Bayesian meta-analysis of efficacy
effects of risperidone in children with autism and irritable and secondary effects. Research in Autism Spectrum Disor-
behavior. Journal of Child and Adolescent Psychopharma- ders, 7, 167–175.
cology, 18, 227–236. Cohen, J. (1988). In L. Erlbaum (Ed.), Statistical power
Aman, M.G., McDougle, C.J., Scahill, L., Handen, B., Arnold, analysis for the behavioral sciences (2nd edn). Hillsdale,
L.E., Johnson, C., . . . & Wagner, A. (2009). Medication and NJ: Lawrence Erlbaum Associates.
parent training in chidlren with pervasive developmental Cortese, S., Adamo, N., Del Giovane, C., Mohr-Jensen, C.,
disorders and serious behaviour problems: Results from a Hayes, A.J., Carucci, S., . . . & Cipriani, A. (2018). Compar-
randomized clinicial trial. Journal of the Americal Academy ative efficacy and tolerability of medications for attention-
Child and Adolescent Psychiatry, 48, 1143–1154. deficit hyperactivity disorder in children, adolescents, and
Ameis, S.H., Corbett-Dick, P., Cole, L., & Correll, C.U. (2013). adults: A systematic review and network meta-analysis. The
Decision making and antipsychotic medication treatment for Lancet Psychiatry, 5, 727–738.
youth With autism spectrum disorders: Applying guidelines Cortese, S., Castelnau, P., Morcillo, C., Roux, S., & Bonnet-
in the real world. The Journal of Clinical Psychiatry, 74, Brilhault, F. (2012). Psychostimulants for ADHD-like symp-
1022–1024. toms in individuals with autism spectrum disorders. Expert
Ameis, S.H., Kassee, C., Corbett-Dick, P., Cole, L., Dadhwal, Review of Neurotherapeutics, 12, 461–473.
S., Lai, M.-C., . . . & Correll, C.U. (2018). Systematic review Craig, F., Margari, F., Legrottaglie, A.R., de Giambattista, C., &
and guide to management of core and psychiatric symptoms Margari, L. (2016). A review of executive function deficits in
in youth with autism. Acta Psychiatrica Scandinavica, 138, autism spectrum disorder and attention-deficit/hyperactiv-
379–400. ity disorder. Neuropsychiatric Disease and Treatment, 12,
Ameis, S.H., Lerch, J.P., Taylor, M.J., Lee, W., Viviano, J.D., 1191–1202.
Pipitone, J., . . . & Anagnostou, E. (2016). A diffusion tensor Dalsgaard, S., Nielsen, H.S., & Simonsen, M. (2013). Five-fold
imaging study in children with ADHD, autism spectrum increase in national prevalence rates of attentiondeficit/
disorder, OCD, and matched controls: Distinct and non- hyperactivity disorder medications for children and adoles-
distinct white matter disruption and dimensional brain- cents with autism spectrum disorder, attention-deficit/hy-
behavior relationships. American Journal of Psychiatry, 173, peractivity disorder, and other psychiatric disorders: A
1213–1222. Danish registe. Journal of Child and Adolescent Psychophar-
Ameis, S.H., & Szatmari, P. (2015). Common psychiatric macology, 23, 432–439.
comorbidities and their assessment. In E. Anagnostou & J. Deb, S., Farmah, B., Arshad, E., Deb, T., Roy, M., & Unwin, G.
Brian (Eds.), Clinician’s manual on autism spectrum disorder (2014). The effectiveness of aripiprazole in the management
(pp. 19–32). Cham, Switzerland: Springer International of problem behaviour in people with intellectual disabilities,
Publishing Switzerland. developmental disabilities and/or autistic spectrum disor-
Aoki, Y., Yoncheva, Y.N., Chen, B., Nath, T., Sharp, D., Lazar, der: A systematic review. Research in Developmental Dis-
M., . . . & Di Martino, A. (2017). Autism spectrum disorder abilities, 35, 711–725.
and attention deficit/hyperactivity disorder: A categorical Di Martino, A., Zuo, X.N., Kelly, C., Grzadzinski, R., Mennes,
and dimensional investigation of white matter structure. M., Schvarcz, A., . . . & Milham, M.P. (2013). Shared and
JAMA Psychiatry, 74, 1120–1128. distinct intrinsic functional network centrality in autism and
Arnold, L.E., Aman, M.G., Cook, A.M., Witwer, A.N., Hall, K.L., attention-deficit/hyperactivity disorder. Biological Psychia-
Thompson, S., & Ramadan, Y. (2006). Atomoxetine for try, 74, 623–632.
hyperactivity in autism spectrum disorders: Placebo-con- Elbourne, D.R., Altman, D.G., Higgins, J.P., Curtin, F., Wor-
trolled crossover pilot trial. Journal of the American Academy thington, H., & Vail, A. (2002). Meta-analyses involving
of Child and Adolescent Psychiatry, 45, 1196–1205. cross-over trials: Methodological issues. International Jour-
Barnard, L., Young, A., Pearson, J., Geddes, J., & O’Brien, G. nal of Epidemiology, 31, 140–149.
(2002). A systematic review of the use of atypical antipsy- Findling, R.L., Mankoski, R., Timko, K., Lears, K., McCartney,
chotics in autism. Journal of Psychopharmacology, 16, 93– T., McQuade, R.D., . . . & Sheehan, J.J. (2014). A randomized
101. controlled trial investigating the safety and efficacy of
Bora, E., & Pantelis, C. (2016). Meta-analysis of social cogni- aripiprazole in the long-term maintenance treatment of
tion in attention-deficit/hyperactivity disorder (ADHD): pediatric patients with irritability associated with autistic
Comparison with healthy controls and autistic spectrum disorder. Journal of Clinical Psychiatry, 75, 22–30.
disorder. Psychological Medicine, 46, 699–716. Frazier, T.W., Shattuck, P.T., Narendorf, S.C., Cooper, B.P.,
Borenstein, M., Hedges, L.V., Higgins, J.P., & Rothstein, H. Wagner, M., & Spitznagel, E.L. (2011). Prevalence and
(2009). Introduction to meta-analysis. Chichester, UK: John correlates of psychotropic medication use in adolescents
Wiley and Sons Ltd. with an autism spectrum disorder with and without care-
Brites, C., Salgado-Azoni, C.A., Ferreira, T.L., Lima, R.F., & giver-reported attentiondeficit/ hyperactivity disorder. Jour-
Ciasca, S.M. (2015). Development and applications of the nal of Child and Adolescent Psychopharmacology, 21, 571–
SWAN rating scale for assessment of attention deficit 579.
hyperactivity disorder: A literature review. Brazilian Journal Fung, L.K., Mahajan, R., Nozzolillo, A., Bernal, P., Krasner, A.,
of Medical and Biological Research, 48, 965–972. Jo, B., . . . & Hardan, A.Y. (2016). Pharmacologic treatment of
Busner, J., & Targum, S.D. (2007). The clinical global impres- severe irritability and problem behaviors in Autism: A
sions scale: Applying a research tool in clinical practice. systematic review and meta-analysis. Pediatrics, 137,
Psychiatry, 4, 28–37. S124–S135.
Christensen, D.L., Braun, K.V.N., Baio, J., Bilder, D., Charles, Ghanizadeh, A. (2013). Atomoxetine for treating ADHD symp-
J., Constantino, J.N., . . . & Yeargin-Allsopp, M. (2018). toms in autism: A systematic review. Journal of Attention
Prevalence and characteristics of autism spectrum disorder Disorders, 17, 635–640.
among children aged 8 years — Autism and developmental Ghanizadeh, A., Sahraeizadeh, A., & Berk, M. (2014). A head-
disabilities monitoring network, 11 sites, United States, to-head comparison of aripiprazole and risperidone for
2012. MMWR Surveillance Summaries, 65, 1–23. safety and treating autistic disorders, a randomized double

© 2020 Association for Child and Adolescent Mental Health


18 Rebecca Rodrigues et al.

blind clinical trial. Child Psychiatry and Human Develop- Hirsch, L.E., & Pringsheim, T. (2016). Aripiprazole for autism
ment, 45, 185–192. spectrum disorders (ASD). In T. Pringsheim (Ed.), Cochrane
Ghanizadeh, A., Tordjman, S., & Jaafari, N. (2015). Aripipra- database of systematic reviews. Chichester, UK: John Wiley
zole for treating irritability in children & adolescents with & Sons, Ltd.
autism: A systematic review. Indian Journal of Medical Hollingdale, J., Woodhouse, E., Young, S., Fridman, A., &
Research, 142, 269. Mandy, W. (2019). Autistic spectrum disorder symptoms in
Ghirardi, L., Brikell, I., Kuja-Halkola, R., Freitag, C.M., children and adolescents with attention-deficit/hyperactiv-
Franke, B., Asherson, P., . . . & Larsson, H. (2018). The ity disorder: A meta-analytical review. Psychological Medi-
familial co-aggregation of ASD and ADHD: Register-based cine, 18, 1–14.
cohort study. Molecular Psychiatry, 23, 257–262. Hollis, C., Chen, Q., Chang, Z., Quinn, P.D., Viktorin, A.,
Ghirardi, L., Pettersson, E., Taylor, M.J., Freitag, C.M., Lichtenstein, P., . . . & Larsson, H. (2019). Methylphenidate
Franke, B., Asherson, P., . . . & Kuja-Halkola, R. (2019). and the risk of psychosis in adolescents and young adults: A
Genetic and environmental contribution to the overlap population-based cohort study. The Lancet Psychiatry, 6,
between ADHD and ASD trait dimensions in young adults: 651–658.
A twin study. Psychological Medicine, 49, 1713–1721. Houghton, R., Liu, C., & Bolognani, F. (2018). Psychiatric
Ghuman, J.K., Aman, M.G., Lecavalier, L., Riddle, M.A., comorbidities and psychotropic medication use in autism: A
Gelenberg, A., Wright, R., . . . & Fort, C. (2009). Randomized, matched cohort study with ADHD and general population
placebocontrolled, crossover study of methylphenidate for comparator groups in the United Kingdom. Autism
attention-deficit/hyperactivity disorder symptoms in Research, 11, 1690–1700.
preschoolers with developmental disorders. Journal of Child Hurwitz, R., Blackmore, R., Hazell, P., Williams, K., &
and Adolescent Psychopharmacology, 19, 329–339. Woolfenden, S. (2012). Tricyclic antidepressants for autism
Gillberg, C. (2010). The ESSENCE in child psychiatry: Early spectrum disorders (ASD) in children and adolescents. In R.
symptomatic syndromes eliciting neurodevelopmental clin- Hurwitz (Ed.), Cochrane database of systematic reviews.
ical examinations. Research in Developmental Disabilities, Chichester, UK: John Wiley and Sons Ltd.
31, 1543–1551. Ichikawa, H., Mikami, K., Okada, T., Yamashita, Y., Ishizaki,
Handen, B.L., Aman, M.G., Arnold, L.E., Hyman, S.L., Tumu- Y., Tomoda, A., . . . & Tadori, Y. (2017). Aripiprazole in the
luru, R.V., Lecavalier, L., . . . & Smith, T. (2015). Atomox- treatment of irritability in children and adolescents with
etine, parent training, and their combination in children autism spectrum disorder in Japan: A randomized, double-
with autism spectrum disorder and attention-deficit/hyper- blind, placebo-controlled study. Child Psychiatry and
activity disorder. Journal of the American Academy of Child Human Development, 48, 796–806.
and Adolescent Psychiatry, 54, 905–915. Jahromi, L.B., Kasari, C.L., McCracken, J.T., Lee, L.S.-Y.,
Handen, B.L., Johnson, C.R., Butter, E.M., Lecavalier, L., Aman, M.G., McDougle, C. J., . . . & Posey, D.J. (2009).
Scahill, L., Aman, M.G., . . . & Mulick, J.A. (2013). Use of a Positive effects of methylphenidate on social communication
direct observational measure in a trial of risperidone and and self-regulation in children with pervasive developmental
parent training in children with pervasive developmental disorders and hyperactivity. Journal of Autism and Develop-
disorders. Journal of Developmental and Physical Disabili- mental Disorders, 39, 395–404.
ties, 25, 355–371. James, S., Montgomery, P., & Williams, K. (2011). Omega-3
Handen, B.L., Johnson, C.R., & Lubetsky, M. (2000). Efficacy fatty acids supplementation for autism spectrum disorders
of methylphenidate among children with autism and symp- (ASD). Cochrane Database of Systematic Reviews, 11,
toms of attention-deficit hyperactivity disorder. Journal of CD007992.
Autism and Developmental Disorders, 30, 245–255. Janssen Pharmaceutical K.K. (2015). A study to evaluate the
Harfterkamp, M., Buitelaar, J.K., Minderaa, R.B., van de Loo- efficacy and safety of risperidone (R064766) in children and
Neus, G., van der Gaag, R.-J., & Hoekstra, P.J. (2014). adolescents with irritability associated with autistic disor-
Atomoxetine in autism spectrum disorder: No effects on der. Available from: https://clinicaltrials.gov/ct2/show/
social functioning; some beneficial effects on stereotyped NCT01624675. Accessed 1 April 2018.
behaviors, inappropriate speech, and fear of change. Journal Jobski, K., H€ ofer, J., Hoffmann, F., & Bachmann, C. (2017).
of Child and Adolescent Psychopharmacology, 24, 481–485. Use of psychotropic drugs in patients with autism spectrum
Harfterkamp, M., van de Loo-Neus, G., Minderaa, R.B., van der disorders: A systematic review. Acta Psychiatrica Scandi-
Gaag, R.-J., Escobar, R., Schacht, A., . . . & Hoekstra, P.J. navica, 135, 8–28.
(2012). A randomized double-blind study of atomoxetine Kamimura-Nishimura, K., Froehlich, T., Chirdkiatgumchai, V.,
versus placebo for attention-deficit/hyperactivity disorder Adams, R., Fredstrom, B., & Manning, P. (2017). Autism
symptoms in children with autism spectrum disorder. spectrum disorders and their treatment with psychotropic
Journal of the American Academy of Child and Adolescent medications in a nationally representative outpatient sam-
Psychiatry, 51, 733–41. ple: 1994–2009. Annals of Epidemiology, 27, 448–453.e1.
Heres, S., Davis, J., Maino, K., Jetzinger, E., Kissling, W., & Kent, J.M., Kushner, S., Ning, X., Karcher, K., Ness, S., Aman,
Leucht, S. (2006). Why olanzapine beats risperidone, risperi- M., . . . & Hough, D. (2013). Risperidone dosing in children
done beats quetiapine, and quetiapine beats olanzapine: An and adolescents with autistic disorder: A double-blind,
exploratory analysis of head-to-head comparison studies of placebo-controlled study. Journal of Autism and Develop-
second-generation antipsychotics. American Journal of Psy- mental Disorders, 43, 1773–1783.
chiatry, 163, 185–194. Lai, M.-C., Kassee, C., Besney, R., Bonato, S., Hull, L., Mandy,
Higgins, J., & Green, S. (Eds.). (2011). Cochrane Handbook for W., . . . & Ameis, S.H. (2019). Prevalence of co-occurring
Systematic Reviews of Interventions (Version 5.). The mental health diagnoses in the autism population: A
Cochrane Collaboration. Available from: www.handbook.c systematic review and meta-analysis. Lancet Psychiatry, 6,
ochrane.orgAccessed 1 April 2018. 819–829.
Hirota, T., Schwartz, S., & Correll, C.U. (2014). Alpha-2 Lai, M.-C., Lombardo, M.V., & Baron-Cohen, S. (2014).
agonists for attention-deficit/hyperactivity disorder in Autism. The Lancet, 383, 896–910.
youth: A systematic review and meta-analysis of monother- Lamberti, M., Siracusano, R., Italiano, D., Alosi, N., Cucinotta,
apy and add-on trials to stimulant therapy. Journal of the F., Di Rosa, G., . . . & Gagliano, A. (2016). Head-to-head
American Academy of Child and Adolescent Psychiatry, 53, comparison of aripiprazole and risperidone in the treatment
153–173. of ADHD symptoms in children with autistic spectrum

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 19

disorder and ADHD: A pilot, open-label, randomized con- Millward, C., Ferriter, M., Calver, S.J., & Connell-Jones, G.G.
trolled study. Pediatric Drugs, 18, 319–329. (2008). Gluten- and casein-free diets for autistic spectrum
Lecavalier, L., Pan, X., Smith, T., Handen, B.L., Arnold, L.E., disorder. Cochrane Database of Systematic Reviews, 2,
Silverman, L., . . . & Aman, M.G. (2018). Parent stress in a CD003498.
randomized clinical trial of atomoxetine and parent training Moher, D., Liberati, A., Tetzlaff, J., Altman, D.G., Altman, D.,
for children with autism spectrum disorder. Journal of Antes, G., . . . & Stewart, L.A. (2009). Preferred reporting
Autism and Developmental Disorders, 48, 980–987. items for systematic reviews and meta-analyses: The
Lionel, A.C., Tammimies, K., Vaags, A.K., Rosenfeld, J.A., Ahn, PRISMA statement. PLoS Medicine, 6, e1000097.
J.W., Merico, M., . . . & Scherer, S. (2014). Disruption of the Moran, L.V., Ongur, D., Hsu, J., Castro, V.M., Perlis, R.H., &
ASTN2/TRIM32 locus at 9q33.1 is a risk factor in males for Schneeweiss, S. (2019). Psychosis with methylphenidate or
autism spectrum disorders, ADHD and other neurodevelop- amphetamine in patients with ADHD. New England Journal
mental phenotypes. Human Molecular Genetics, 23, 2752– of Medicine, 380, 1128–1138.
2768. Mosholder, A.D., Gelperin, K., Hammad, T.A., Phelan, K., &
Loebel, A., Brams, M., Goldman, R.S., Silva, R., Hernandez, D., Johann-Liang, R. (2009). Hallucinations and other psy-
Deng, L., . . . & Findling, R. (2016). Lurasidone for the chotic symptoms associated with attention-deficit/hyperac-
treatment of irritability associated with autistic disorder. tivity disorder drugs in children. Pediatrics, 123, 611–616.
Journal of Autism and Developmental Disorders, 46, 1153– Nagaraj, R., Singhi, P., & Malhi, P. (2006). Risperidone in
1163. children with autism: Randomized, placebo-controlled, dou-
Lundh, A., Lexchin, J., Mintzes, B., Schroll, J.B., & Bero, L. ble-blind study. Journal of Child Neurology, 21, 450–455.
(2017). Industry sponsorship and research outcome. National Institute for Health and Care Excellence (2013).
Cochrane Database of Systematic Reviews, 2, MR000033. Autism spectrum disorder in under 19s: Support and
Mahajan, R., Bernal, M.P., Panzer, R., Whitaker, A., Roberts, management (CG170). Available from: https://www.nice.
W., Handen, B., . . . & Veenstra-VanderWeele, J. (2012). org.uk/guidance/cg170 [last accessed 14 November 2018].
Clinical practice pathways for evaluation and medication National Institute for Health and Care Excellence (2018).
choice for attention-deficit/hyperactivity disorder symptoms Attention deficit hyperactivity disorder: Diagnosis and man-
in autism spectrum disorders. Pediatrics, 130, S125–S138. agement (NG87). Available from: https://www.nice.org.uk/
Malone, R.P., Cater, J., Sheikh, R.M., Choudhury, M.S., & guidance/ng87. Accessed 1 April 2018.
Delaney, M.A. (2001). Olanzapine versus haloperidol in Niederhofer, H. (2004). Venlafaxine has modest effects in
children with autistic disorder: An open pilot study. Journal autistic children. Therapy, 1, 87–90.
of the American Academy of Child and Adolescent Psychia- Niederhofer, H., Staffen, W., & Mair, A. (2003). Tianeptine: A
try, 40, 887–894. novel strategy of psychopharmacological treatment of chil-
Man, K.K.C., Coghill, D., Chan, E.W., Lau, W.C.Y., Hollis, C., dren with autistic disorder. Human Psychopharmacology,
Liddle, E., . . . & Wong, I.C.K. (2016). Methylphenidate and 18, 389–393.
the risk of psychotic disorders and hallucinations in chil- Owen, R., Sikich, L., Marcus, R.N., Corey-Lisle, P., Manos, G.,
dren and adolescents in a large health system. Translational McQuade, R.D., . . . & Findling, R. (2009). Aripiprazole in the
Psychiatry, 6, e956. treatment of irritability in children and adolescents with
Marcus, R.N., Owen, R., Kamen, L., Manos, G., McQuade, autistic disorder. Pediatrics, 124, 1533–40.
R.D., Carson, W.H., & Aman, M.G. (2009). A placebo- Padilha, S.C.O.S., Virtuoso, S., Tonin, F.S., Borba, H.H., &
controlled, fixed-dose study of aripiprazole in children and Pontarolo, R. (2018). Efficacy and safety of drugs for
adolescents with irritability associated with autistic disor- attentiondeficit/ hyperactivity disorder in children and ado-
der. Journal of the American Academy of Child and Adoles- lescents: A network meta-analysis. European Child and
cent Psychiatry, 48, 1110–1119. Adolescent Psychiatry, 27, 1335–1345.
Martin, J., Cooper, M., Hamshere, M.L., Pocklington, A., Pagsberg, A.K., Tarp, S., Glintborg, D., Stenstrøm, A.D., Fink-
Scherer, S.W., Kent, L., . . . & Thapar, A. (2014). Biological Jensen, A., Correll, C.U., & Christensen, R. (2017). Acute
overlap of attention-deficit/hyperactivity disorder and aut- antipsychotic treatment of children and adolescents with
ism spectrum disorder: Evidecne from copy number vari- schizophrenia-spectrum disorders: A systematic review and
ants. Journal of the American Academy of Child and network metaanalysis. Journal of the American Academy of
Adolescent Psychiatry, 53, 761–770. Child and Adolescent Psychiatry, 56, 191–202.
McConachie, H., Parr, J.R., Glod, M., Hanratty, J., Livingstone, Palladino, V.S., McNeill, R., Reif, A., & Kittel-Schneider, S.
N., Oono, I.P., . . . & Williams, K. (2015). Systematic review of (2019). Genetic risk factors and gene–environment interac-
tools to measure outcomes for young children with autism tions in adult and childhood attention-deficit/hyperactivity
spectrum disorder. Health Technology Assessment, 19, 1– disorder. Psychiatric Genetics, 29, 63–78.
506. Patra, S., Nebhinani, N., Viswanathan, A., & Kirubakaran, R.
McCracken, J.T. (2004). Safety issues with drug therapies for (2019). Atomoxetine for attention deficit hyperactivity disor-
autism spectrum disorders. The Journal of Clinical Psychi- der in children and adolescents with autism: A systematic
atry, 66, 32–37. review and meta-analysis. Autism Research, 12, 542–552.
McDougle, C.J. (2017). Effectiveness of atomoxetine in treating Pearson, D.A., Santos, C.W., Aman, M.G., Arnold, L.E., Casat,
ADHD symptoms in children and adolescents with autism. C.D., Mansour, R., . . . & Cleveland, L.A. (2013). Effects of
Available from: https://clinicaltrials.gov/ct2/show/ extended release methylphenidate treatment on ratings of
NCT00498173 [last accessed 12 January 2018]. attention-deficit/hyperactivity disorder (ADHD) and associ-
McDougle, C.J., Hollway, J., Scahill, L., Koenig, K., Aman, M., ated behavior in children with autism spectrum disorders
McGough, J., . . . & Vitiello, B. (2005). Risperidone for the and ADHD symptoms. Journal of Child and Adolescent
core symptom domains of autism: Results from the study by sychopharmacology, 23, 337–51.
the Autism Network of the Research Units on Pediatric Pelham, W.E., Fabiano, G.A., Waxmonsky, J.G., Greiner, A.R.,
Psychopharmacology. American Journal of Psychiatry, 162, Gnagy, E.M., Pelham, W.E., . . . & Murphy, S.A. (2016).
1142–1148. Treatment sequencing for childhood ADHD: A multiple-
Miller, M., Musser, E.D., Young, G.S., Olson, B., Steiner, R.D., randomization study of adaptive medication and behavioral
& Nigg, J.T. (2019). Sibling recurrence risk and cross- interventions. Journal of Clinical Child and Adolescent
aggregation of attention-deficit/hyperactivity disorder and Psychology, 45, 396–415.
autism spectrum disorder. JAMA Pediatrics, 173, 147–152. Polderman, T.J., Hoekstra, R.A., Posthuma, D., & Larsson, H.
(2014). The co-occurrence of autistic and ADHD dimensions

© 2020 Association for Child and Adolescent Mental Health


20 Rebecca Rodrigues et al.

in adults: An etiological study in 17, 770 twins. Transla- Stergiakouli, E., Smith, G.D., Martin, J., Skuse, D.H., Viecht-
tional Psychiatry, 4, e435. bauer, W., Ring, S.M., . . . & St. Pourcain, B. (2017). Shared
Posey, D.J., Aman, M.G., McCracken, J.T., Scahill, L., Tierney, genetic influences between dimensional ASD and ADHD
E., Arnold, L.E., . . . & McDougle, C.J. (2007). Positive effects symptoms during child and adolescent development. Molec-
of methylphenidate on inattention and hyperactivity in ular Autism, 8, 18.
pervasive developmental disorders: An analysis of secondary Storebø, O.J., Pedersen, N., Ramstad, E., Kielsholm, M.L.,
measures. Biological Psychiatry, 61, 538–544. Nielsen, S.S., Krogh, H.B., . . . & Gluud, C. (2018). Methyl-
Punja, S., Shamseer, L., Hartling, L., Urichuk, L., Vandermeer, phenidate for attention deficit hyperactivity disorder (ADHD)
B., Nikles, J., & Vohra, S. (2016). Amphetamines for in children and adolescents - Assessment of adverse events
attention deficit hyperactivity disorder (ADHD) in children in non-randomised studies. Cochrane Database of System-
and adolescents. Cochrane Database of Systematic Reviews, atic Reviews, 5, CD012069.
2, CD009996. Storebø, O.J., Ramstad, E., Krogh, H.B., Nilausen, T.D.,
Reichow, B., Volkmar, F.R., & Bloch, M.H. (2013). Systematic Skoog, M., Holmskov, M., . . . & Gluud, C. (2015). Methyl-
review and meta-analysis of pharmacological treatment of phenidate for children and adolescents with attention deficit
the symptoms of attention-deficit/hyperactivity disorder in hyperactivity disorder (ADHD). Cochrane Database of Sys-
children with pervasive developmental disorders. Journal of tematic Reviews, 11, CD009885.
Autism and Developmental Disorders, 43, 2435–2441. Sturman, N., Deckx, L., & van Driel, M.L. (2017). Methylphe-
Research Units on Pediatric Psychopharmacology (RUPP) nidate for children and adolescents with autism spectrum
Autism Network (2002). Risperidone in children with autism disorder. Cochrane Database of Systematic Reviews, 2017,
and serious behavioral problems. The New England Journal CD011144.
of Medicine, 347, 314–321. Subcommittee on Attention-Deficit/Hyperactivity Disorder,
Research Units on Pediatric Psychopharmacology (RUPP) Steering Committee on Quality Improvement and Manage-
Autism Network (2005). Randomized, controlled, crossover ment (2011). ADHD: Clinical practice guideline for the
trial of methylphenidate in pervasive developmental disor- diagnosis, evaluation, and treatment of attention-deficit/
ders with hyperactivity. Archives of General Psychiatry, 62, hyperactivity disorder in children and adolescents. Pedi-
1266–1274. atrics, 128, 1007–1022.
Sandin, S., Lichtenstein, P., Kuja-Halkola, R., Hultman, C., Tonin, F.S., Steimbach, L.M., Mendes, A.M., Borba, H.H.,
Larsson, H., & Reichenberg, A. (2017). The heritability of Pontarolo, R., & Fernandez-Llimos, F. (2018). Mapping the
autism spectrum disorder. JAMA, 318, 1182–1184. characteristics of network meta-analyses on drug therapy: A
Scahill, L., McCracken, J.T., King, B.H., Rockhill, C., Shah, B., systematic review. PLoS One, 13, e0196644.
Politte, L., . . . & McDougle, C.J. (2015). Extended-release Troost, P.W., Althaus, M., Lahuis, B.E., Buitelaar, J.K.,
guanfacine for hyperactivity in children with autism spec- Minderaa, R.B., & Hoekstra, P.J. (2006). Neuropsychological
trum disorder. American Journal of Psychiatry, 172, 1197– effects of risperidone in children with pervasive develop-
1206. mental disorders: A blinded discontinuation study. Journal
Sharma, A., & Shaw, S.R. (2012). Efficacy of risperidone in of Child and Adolescent Psychopharmacology, 16, 561–573.
managing maladaptive behaviors for children with autistic Troost, P.W., Lahuis, B.E., Steenhuis, M.-P., Ketelaars, C.E.J.,
spectrum disorder: A meta-analysis. Journal of Pediatric Buitelaar, J.K., van Engeland, H., . . . & Hoekstra, P.J.
Health Care, 26, 291–299. (2005). Longterm effects of risperidone in children with
Shea, S., Turgay, A., Carroll, A., Schulz, M., Orlik, H., Smith, autism spectrum disorders: A placebo discontinuation
I., & Dunbar, F. (2004). Risperidone in the treatment of study. Journal of the American Academy of Child and
disruptive behavioral symptoms in children with autistic Adolescent Psychiatry, 44, 1137–44.
and other pervasive developmental disorders. Pediatrics, Tumuluru, R.V., Corbett-Dick, P., Aman, M.G., Smith, T.,
114, E634–E641. Arnold, L.E., Pan, X., . . . & Handen, B. (2017). Adverse
Siegel, M., & Beaulieu, A.A. (2012). Psychotropic medications events of atomoxetine in a double-blind placebo-controlled
in children with autism spectrum disorders: A systematic study in children with autism. Journal of Child and Adoles-
review and synthesis for evidence-based practice. Journal of cent Psychopharmacology, 27, 708–714.
Autism and Developmental Disorders, 42, 1592–1605. van der Meer, J.M., Harfterkamp, M., van de Loo-Neus, G.,
Sikora, D.M., Vora, P., Coury, D.L., & Rosenberg, D. (2012). Althaus, M., de Ruiter, S.W., Donders, A.R.T., . . .& Rom-
Attention-deficit/hyperactivity disorder symptoms, adaptive melse, N.N.J. (2013). A randomized, double-blind compar-
functioning, and quality of life in children with autism ison of atomoxetine and placebo on response inhibition and
spectrum disorder. Pediatrics, 130, S91–S97. interference control in children and adolescents with autism
Smith, T., Aman, M.G., Arnold, L.E., Silverman, L.B., Lecav- spectrum disorder and comorbid attention-deficit/hyperac-
alier, L., Hollway, J., . . . & Handen, B. (2016). Atomoxetine tivity disorder symptoms. Journal of Clinical Psychopharma-
and parent training for children with autism and attention- cology, 33, 824–827.
deficit/hyperactivity disorder: A 24-week extension study. Varni, J.W., Handen, B.L., Corey-Lisle, P.K., Guo, Z., Manos,
Journal of the American Academy of Child and Adolescent G., Ammerman, D.K., . . . & Mankoski, R. (2012). Effect of
Psychiatry, 55, 868–876.e2. aripiprazole 2 to 15 mg/d on health-related quality of life in
Sochocky, N., & Milin, R. (2013). Second generation antipsy- the treatment of irritability associated with autistic disorder
chotics in Asperger’s disorder and high functioning autism: in children: A post hoc analysis of two controlled trials.
A systematic review of the literature and effectiveness of Clinical Therapeutics, 34, 980–992.
meta-analysis. Current Clinical Pharmacology, 8, 370–379. Warren, A.E., Hamilton, R.M., B elanger, S.A., Gray, C., Gow,
Soke, G.N., Maenner, M.J., Christensen, D., Kurzius-Spencer, R.M., Sanatani, S., . . . & Schachar, R. (2009). Cardiac risk
M., & Schieve, L.A. (2018). Prevalence of co-occurring assessment before the use of stimulant medications in
medical and behavioral conditions/symptoms among 4- children and youth: A joint position statement by the
and 8-year-old children with autism spectrum disorder in Canadian Paediatric Society, the Canadian Cardiovascular
selected areas of the United States in 2010. Journal of Society, and the Canadian Academy of Child and Adolescent
Autism and Developmental Disorders, 48, 2663–2676. Psychiatry. The Canadian Journal of Cardiology, 25, 625–
Spielmans, G.I., & Parry, P.I. (2010). From evidence-based 630.
medicine to marketing-based medicine: Evidence from inter- Willcutt, E.G. (2012). The prevalence of DSM-IV attention-
nal industry documents. Journal of Bioethical Inquiry, 7, 13– deficit/hyperactivity disorder: A meta-analytic review. Neu-
29. rotherapeutics, 9, 490–499.

© 2020 Association for Child and Adolescent Mental Health


Treating ADHD symptoms in autism 21

Wolraich, M.L., Lambert, W., Doffing, M.A., Bickman, L., deficit/hyperactivity disorder and autism spectrum disorder
Simmons, T., & Worley, K. (2003). Psychometric properties based upon expert consensus. BMC Medicine, 18, 146.
of the Vanderbilt ADHD diagnostic parent rating scale in a https://doi.org/10.1186/s12916-020-01585-y. Accessed 1
referred population. Journal of Pediatric Psychology, 28, April 2018.
559–568.
Young, S., Hollingdale, J., Absoud, M., Bolton, P., Branney, P., Accepted for publication: 3 July 2020
Colley, W., . . . & Woodhouse, E. (2020). Guidance for
identification and treatment of individuals with attention

© 2020 Association for Child and Adolescent Mental Health

You might also like