Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

Handbook of Clinical Neurology, Vol.

165 (3rd series)


Psychopharmacology of Neurologic Disease
V.I. Reus and D. Lindqvist, Editors
https://doi.org/10.1016/B978-0-444-64012-3.00024-1
Copyright © 2019 Elsevier B.V. All rights reserved

Chapter 24

The psychopharmacology of autism spectrum disorder


and Rett syndrome

ANTONIO M. PERSICO*, ARIANNA RICCIARDELLO, AND FRANCESCA CUCINOTTA


Interdepartmental Program “Autism 0-90”, "Gaetano Martino" University Hospital, Messina, Italy

Abstract
Autism spectrum disorder (ASD) appears in early childhood and is characterized by persistent deficits in
communication and social interaction, as well as restricted interests, repetitive behaviors, and unusual sen-
sory issues. ASD can be idiopathic or syndromic, in the latter case representing one of the many manifes-
tations of a genetic disorder, such as Rett syndrome. Psychopharmacology cannot directly ameliorate core
autistic symptoms, but rather aims at treating comorbid disorders, such as ADHD, sleep disturbances, psy-
chomotor agitation and aggressiveness, seizures, and anxiety. Until the 1990s, it was mainly based on typ-
ical neuroleptics and tricyclic antidepressants, whereas second-generation antipsychotics later became
first-line drugs for these same indications. In general, selective serotonin reuptake inhibitors have not
proven effective in ASD patients. Psychostimulants are frequently prescribed, but display modest efficacy
and enhanced potential for side effects and noncompliance. Targeted patients benefit from mood stabilizers
and antiepileptic drugs. Experimental drug treatments for ASD include oxytocin and vasopressin, bume-
tanide, sulforaphane, nutritional supplements, memantine, and D-cycloserine. Work performed on syndro-
mic forms, represents an important source of information for experimental therapies of ASD and
knowledge of the unique mechanisms underlying autism in each individual patient may in the future
pave the path to personalized drug treatments.

corresponding to 1/59 children aged 8 in 2014 (Baio


INTRODUCTION
et al., 2018). The M:F ratio is between 3:1 and 4:1
Autism spectrum disorder (ASD) is a developmental dis- (Loomes et al., 2017). Female sex is associated with a
order that appears in early childhood and is characterized higher disease threshold, probably due to genetic,
by (A) persistent deficits in communication and social epigenetic, and hormonal protection (Werling, 2016).
interaction as well as (B) restricted interests, repetitive Approximately 90% of ASD cases are “idiopathic,”
behaviors, and unusual sensory issues (APA, 2013). whereas 10% are “syndromic,” i.e., ASD is part of a
These core symptoms are frequently associated with broader genetic syndrome (Persico and Napolioni,
emotional and behavioral disturbances, including anxi- 2013). From a biologic and, to some extent, clinical
ety, irritability, labile mood, inattention, hyperactivity, standpoint, Rett syndrome represents one of the best-
and sleep problems, which may lead to functional impair- studied examples of syndromic autism (Rett, 1966;
ment and significant emotional and economic burden for Amir et al., 1999).
caregivers (Simonoff et al., 2008). At present, evidence-based therapeutic interventions
The prevalence of ASD in the second half of the 1980s in ASD include a variety of approaches, such as behavior
stood at 2–5/10,000 children, posing autism as a rare dis- therapy, speech therapy, occupational and physical
ease (Fombonne, 2005). Thirty years later, in the United therapy, social skills training, special education, and
States the prevalence has risen to 168/10,000 children, vocational training, among other nonpharmacologic

*Correspondence to: Antonio M. Persico, Interdepartmental Program “Autism 0-90”, “Gaetano Martino” University Hospital, via
Consolare Valeria 1, I-98125 Messina, Italy. Tel: +39-090-221-3143, Fax: +39-090-293-0414, E-mail: apersico@unime.it
392 A.M. PERSICO ET AL.
interventions (Tonge et al., 2014; Wong et al., 2015). As mechanism, they enhance extracellular monoamine
yet, pharmacologic treatment is not directed to the core levels by reducing their uptake. TCAs, such as desipra-
symptoms of ASD, but rather to comorbid disorders mine, imipramine, nortriptyline, amitriptyline as well
and problem behaviors, such as irritability, anxiety, as buproprion, a dopamine reuptake blocker, have been
aggression, self-injurious behavior, ADHD, sleep disor- used to treat individuals with autism for comorbid anxi-
ders, and seizures. Until the end of the 1980s, typical neu- ety, obsessive–compulsive disorder (OCD), major
roleptics and tricyclic antidepressants (TCAs) were the depression, and attention deficit hyperactivity disorder
only two viable options to reduce behavioral symptoms (ADHD), but clinical response and side effect severity
in ASD. Currently, the array of drugs that have proven appear less favorable compared to the nonautistic popu-
their efficacy on disorders and symptoms comorbid with lation and warrant caution. Antagonizing multiple cho-
ASD has significantly risen, although the vast majority linergic, histaminergic, and adrenergic receptors, in
must be prescribed “off-label” (Persico et al., 2015). addition to monoamine transporters, TCAs frequently
For most medications, limited data are available regard- produce side effects, including dry mouth, constipation,
ing efficacy in autistic individuals, especially children urinary retention, blurred vision, sinus tachycardia,
and adolescents, owing to small sample sizes, diagnostic impaired cognition, sedation, impaired motor coordina-
heterogeneity, and methodological difficulties in orga- tion, weight gain, balance problems, orthostatic hypoten-
nizing multicenter trials (Persico et al., 2015). sion, cardiac toxicity, central nervous system (CNS)
This chapter provides an overview of psychopharma- depression, and seizures. Nonetheless, despite producing
cologic agents either approved or currently in use for more frequent and severe adverse effects than, for exam-
ASD and Rett syndrome. We then summarize innovative ple, fluoxetine or fluvoxamine, in healthy patients with-
pharmacologic agents undergoing clinical trials, aimed at out seizures and with normal cardiac function, TCAs are
translating our ever-increasing knowledge of the patho- generally safe and well tolerated.
physiology of these disorders into pharmacologic tools The predominantly 5-HT reuptake blocker most stud-
able to ameliorate the core symptoms of ASD. ied in ASD is clomipramine. Superior to placebo and to
desipramine (a predominantly NE reuptake blocker), clo-
mipramine was able to improve autistic symptoms,
TYPICAL NEUROLEPTICS AND
anger, and repetitive behaviors in 57% of ASD subjects
TRICYCLIC ANTIDEPRESSANTS
enrolled in a double-blind RCT (Gordon et al., 1993).
Neuroleptics have been the most widely studied class of However, progressively increasing doses of clomipra-
psychopharmacologic agents in autism over the past mine, up to a maximum of 250 or 5.0 mg/kg/day for
35 years. Starting in the 1960s, children and adolescents 5 weeks, showed no therapeutic advantage and resulted
with autism were enrolled in randomized controlled in severe side effects in an open trial on eight ASD chil-
trials (RCTs) of first-generation antipsychotics, espe- dren aged 3.5–8.7 years (Sanchez et al., 1996). An RCT,
cially haloperidol, thioridazine, and trifluoperizine. comparing the efficacy of clomipramine, haloperidol,
These agents were found to decrease hyperactivity, and placebo in 31 subjects with ASD, found no signifi-
stereotypic behaviors, social withdrawal, aggressive- cant difference between clomipramine and placebo on
ness, and temper outbursts. For example, the first stereotypy, irritability, and hyperactivity recorded using
RCT, enrolling 45 children aged 2–7 years with DSM- the Aberrant Behavior Checklist (ABC) (Remington
III autistic disorder, showed significant positive effects et al., 2001). Moreover, many participants receiving clo-
of haloperidol (0.25–4.0 mg/day) on global functioning mipramine discontinued treatment due to side effects or
and multiple behavioral problems, as measured with lack of efficacy. Nortriptyline, a predominantly NE reup-
Children’s Psychiatric Rating Scale (CPRS) and Clinical take blocker, may reduce hyperactivity, aggressiveness,
Global Impression (CGI) scores, but no improvement in and ritualized behavior (Kurtis et al., 1966), and be better
learning abilities was recorded (Anderson et al., 1989). tolerated than imipramine (Campbell et al., 1971).
The major limitation of these drugs is their side-effect Tianeptine, pharmacologically distinct from TCAs but
profile, ranging from relatively benign dry mouth and sharing at least some properties with this drug class,
sedation to severe parkinsonism, acute dystonia, tardive was able to reduce abnormal behaviors, such as inade-
dyskinesia, and neuroleptic malignant syndrome. Des- quate eye contact and speech abnormalities, significantly
pite these adverse effects, haloperidol and other neuro- more than placebo when rated by parents and teachers,
leptics remained the most prescribed drugs in ASD but clinician ratings did not show any significant dif-
until the 1990s. ference between tianeptine and placebo (Niederhofer
TCAs block several monoamine transporters, pri- et al., 2003). Adverse effects, including drowsiness
marily the serotonin (5-HT) and norepinephrine (NE) and reduced activity levels, were recorded in a sizable
transporters SERT and NET, respectively. Through this number of patients treated with tianeptine.
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 393
ATYPICAL ANTIPSYCHOTICS (0.175 mg/day for >45 kg) (Kent et al., 2013a). The side
effect profile of risperidone in individuals with autism is
Second-generation antipsychotics (SGAs), also known generally favorable. Neurologic side effects are rela-
as “atypical antipsychotics” emerged in the 1980s. This tively unusual, although acute extrapyramidal symptoms
new group of neuroleptics shares some effects with first- still occur. The most common side effect of risperidone in
generation neuroleptics, but differs in neurochemical autistic patients is also a metabolic syndrome, with
profile, effectiveness, and side effects. A shift toward increased appetite, weight gain, and, less frequently,
the use of SGAs was largely driven by their lower risk sedation. Direct comparison of haloperidol and risperi-
of extrapyramidal side effects, although they can pro- done in 30 children with ASD, aged 8–18 years, found
mote a metabolic syndrome, characterized by weight risperidone to be more effective than haloperidol in
gain, elevated lipid and prolactin serum levels, and pos- the treatment of behavioral symptoms, impulsivity,
sibly type 2 diabetes mellitus. In general, they have a dif- language skills, and impaired social relations (Miral
ferent receptor blocking profile, being less effective on et al., 2008). Finally, risperidone modulates important
dopamine D2 as compared to D1 receptors and also astroglial functions, possibly exerting antioxidant and
blocking many 5-HT receptor subtypes. Atypical anti- neuroprotective effects in ASD (Quincozes-Santos
psychotics include risperidone, paliperidone, aripipra- et al., 2009).
zole, olanzapine, clozapine, quetiapine, ziprasidone, Aripiprazole acts as an agonist, partial agonist, or
asenapine, sertindole, and zotepine. The FDA approved antagonist at dopamine D2 receptors, depending on cell
risperidone in 2006 and aripirazole in 2009 for the treat- type and function examined (Shapiro et al., 2003). It also
ment of irritability in ASD, including tantrums, aggres- acts as an inverse agonist at 5-HT2B receptors, as a partial
sion, and self-injury, whereas the European Medicine agonist at 5-HT1A, 5-HT2A, 5-HT2C, D3, and D4 recep-
Agency has only approved risperidone. tors, as an antagonist at 5-HT7, a1-adrenergic, and
Risperidone, a potent antagonist at dopamine D2 and H1-histamine receptors (Shapiro et al., 2003). This pecu-
serotonin 5-HT2A receptors, has been one of the most liar mechanism of action allows aripiprazole to act simul-
extensively studied medications in ASD. Several open taneously as an antagonist in hyperdopaminergic and as
studies have supported the efficacy, safety, and tolerabil- an agonist in hypodopaminergic brain regions or condi-
ity of risperidone in the treatment of autistic behaviors, tions. Several open-label studies have shown the tolera-
irritability, aggression, and hyperactivity (Findling bility and safety of aripiprazole therapy, as well as its
et al., 1997; McDougle et al., 1997; Nicolson et al., effectiveness for the treatment of hyperactivity and irri-
1998; Masi et al., 2001; Nishimura et al., 2003; tability in autistic children and adolescents (Stigler
Gagliano et al., 2004; Kent et al., 2013a,b; Lamberti et al., 2009; Marcus et al., 2011; Ishitobi et al., 2013;
et al., 2016). Multiple RCTs have also produced defini- Maloney et al., 2014; Lamberti et al., 2016; Ichikawa
tive evidence of efficacy on irritability and suggestive et al., 2018). Numerous RCTs have also provided strong
evidence for repetitive behaviors. Long-term efficacy evidence of efficacy in reducing irritability, hyperactiv-
of risperidone was reported in the 2005 RUPPAN study, ity, and stereotypic behavior in autistic children and ado-
in which an initial open-label 4-month treatment period lescents aged 6–17 years, both acutely and following
at an established effective dose was followed by an long-term treatment (Marcus et al., 2009; Owen et al.,
8-week double-blind, placebo-controlled withdrawal 2009; Aman et al., 2010; Robb et al., 2011; Findling
period; over 60% of youth relapsed while on placebo et al., 2014; Ichikawa et al., 2017). In addition to these
as compared to 12.5% on risperidone (RUPPAN, studies, an 8-week open-label trial conducted on 10 pre-
2005a). However, symptom improvement with risperi- schoolers with ASD, aged 3–6 years, reported a signifi-
done came at the expense of weight gain, which averaged cant reduction in irritability, stereotypic behavior, and
5.1 kg (RUPPAN, 2005a). Several double-blind RCTs hyperactivity (Habibi et al., 2015). Direct comparison
found risperidone effective in treating motor stereoty- of aripiprazole to risperidone in a 2-month double-blind
pies, hyperactivity, impulsivity, agitation, and aggression RCT showed similar efficacy in treating irritability for
in monotherapy (McDougle et al., 1998; McCracken the two drugs, as measured with the ABC-I, but a faster
et al., 2002; Malone et al., 2002; Shea et al., 2004; onset of action for aripiprazole (Ghanizadeh et al., 2014).
Troost et al., 2005; McDougle et al., 2005; Luby et al., The most frequent side effects include increased appetite
2006; Nagaraj et al., 2006; Pandina et al., 2007; Aman and weight gain, sedation, and psychomotor agitation,
et al., 2008; Kent et al., 2013a; Ghanizadeh et al., but side effects overall occurred less frequently com-
2014; Aman et al., 2015). Interestingly, in a double-blind pared to other SGAs, especially serious EKG and cardiac
RCT of 96 children, 5–17 years old, higher doses of ris- adverse events (De Hert et al., 2011; Ho et al., 2012).
peridone (1.75 mg/day for >45 kg) contributed to greater Children and adolescents tend to have more pronounced
change in the ABC-I as compared to lower doses side effects from aripiprazole when antipsychotic-naïve
394 A.M. PERSICO ET AL.
than if previously treated with another SGA, but drug adults who failed to respond to other first-line treatments
discontinuation rates are similar (10.8% vs 8.3%, (Zuddas et al., 1996; Chen et al., 2001; Lambrey et al.,
respectively) (Mankoski et al., 2013). 2010; Sahoo et al., 2017). It is used as second-line
Olanzapine use in ASD has been assessed only in one treatment because of potentially severe or even life-
small RCT (Hollander et al., 2006a), three prospective threatening side effects (especially agranulocytosis,
open-label trials (Potenza et al., 1999; Kemner et al., seizures, sedation, and cardiomyopathy), which require
2002; Fido and Al-Saad, 2008), and one open-label com- medical follow-up and regular blood tests for as long
parison with haloperidol (Malone et al., 2001). Hollander as the patient continues to take the medication. Quetia-
et al. (2006a) conducted an 8-week RCT on 11 autistic pine has produced contrasting results. Two small
children, aged 6–11 years, randomized to receive olanza- open-label studies found quetiapine not effective and
pine (mean dose 10 mg/d) or placebo. Significant poorly tolerated in children and adolescents with ASD
improvements were recorded in the primary outcome (Martin et al., 1999; Findling et al., 2004). However, a
measure (CGI-I), but not in secondary outcome measures retrospective study of 20 patients (aged 5–28 years)
(CY-BOCS and Overt Aggression Scale-Modified). who received a daily dose of quetiapine between
Common adverse effects included sedation and weight 25 and 600 mg over a period of 4–180 weeks showed
gain; rhinitis, “glazed eyes,” constipation, and insomnia modest improvements in maladaptive behavior, espe-
were also more frequent in olanzapine-treated children cially in CGI-S scores (Corson et al., 2004). Another ret-
compared to placebo. In a 6-week open-label study, rospective study suggests that 40%–60% of autistic
5/6 and 3/6 olanzapine- and haloperidol-treated ASD patients may benefit from quetiapine treatment
children, respectively, were rated as “responders” using (Hardan et al., 2005). Finally, an open-label trial of
the CGI-I (Malone et al., 2001). Collectively, these 11 adolescents aged 13–17 years found quetiapine well
results and those from open-trials indicate that olanza- tolerated and effective in treating aggressive behavior
pine may be beneficial, especially for irritability, to chil- and sleep disturbances, but less so in autistic behaviors
dren and adolescents who do not respond well to (Golubchik et al., 2011). Ziprasidone has an interesting
risperidone or aripiprazole. pharmacologic profile, characterized by: (a) strong
Lurasidone is a novel SGA with potent 5-HT2A, 5-HT2A receptor antagonism, (b) moderate antagonism
5-HT7, D2, and noradrenergic a1 receptor antagonism, at 5-HT1D, dopamine D2, noradrenergic a1, and hista-
as well as partial agonism at the 5-HT1A receptor mine H1 receptors, (c) antidepressant and anxiolytic
(Ishibashi et al., 2010). In addition to approved indica- effects through 5-HT1A receptor antagonism and
tions for adult schizophrenia and bipolar disorder, this norepinephrine reuptake inhibition. Ziprasidone
pharmacologic profile may also be associated with an (20–160 mg/day) can have beneficial effects for ASD
antidepressant effect and lesser side effects involving children and adolescents, without producing significant
weight gain and changes in metabolism. One recent mul- weight gain, metabolic changes, prolactin increase, or
ticenter double-blind RCT study enrolled 150 ASD other severe adverse effect (McDougle et al., 2002;
patients, aged 6–17 years, randomized to receive either Goforth and Rao, 2003; Duggal, 2007; Malone et al.,
lurasidone 20 mg/day (N ¼ 50), 60 mg/day (N ¼ 49), or 2007; Dominick et al., 2015). Side effects include QTc
placebo (N ¼ 51) for 6 weeks. Lurasidone was not signif- increases and occasional acute dystonic reactions
icantly superior to placebo in ABC Irritability or (Malone et al., 2007). The efficacy of ziprasidone on irri-
CY-BOCS scores at any dose, but produced significantly tability in youth with ASD may be somewhat lower com-
greater improvement at 20 mg/day over placebo using the pared to risperidone and aripiprazole, but it is well
CGI-I. Common side effects included nausea and drows- tolerated (Dominick et al., 2015). Paliperidone is the
iness, with only minor changes observed in weight and extended-release major active metabolite of risperidone.
metabolic parameters (Loebel et al., 2016). A case report Only two case reports and one 8-week open-label study
showed improvement in irritability, perseveration, and suggest that it may be effective on irritability in ASD
aggression in a 13-year-old autistic boy at an initial dose children and adolescents, at a mean final dosage of
of 10 mg/day, increased to 30 mg/day, without significant 7.1 mg/day (range 3–12 mg/day) (Stigler et al., 2010,
adverse effects (Millard et al., 2014). 2012; Kowalski et al., 2011). The drug is generally well
No RCT has been published to this date for clozapine, tolerated and side effects appear superimposable to those
quetiapine, ziprasidone, or newer SGAs, including pali- of risperidone, most frequently including weight gain
peridone, iloperidone, and asenapine. However, open tri- and increased prolactin levels (Stigler et al., 2012).
als report effectiveness, and these drugs are often used in There are no studies published to date on the use of
clinical practice. Clozapine has been reported to be effec- some newer SGAs, namely asenapine and iloperidone,
tive in improving hyperactivity and aggression in in ASD children and adolescents. These drugs are both
treatment-resistant autistic children, adolescents, and D2 and 5-HT2A receptors antagonists, approved by the
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 395
FDA in 2009 for the treatment of adults affected by hyperactivity, impulsivity, and sleep disturbance, are
schizophrenia. common in ASD children and adolescents, frequently
In summary, antipsychotics can be very useful for leading to noncompliance or to treatment interruption.
children and adolescents with autism to treat irritability, Fluvoxamine has been proven effective in treatment
psychomotor agitation, temper outbursts, or aggression of repetitive, maladaptive, and aggressive behaviors in
toward self or others. Prominent hyperactivity and some adults, but not in children and adolescents with
impulsivity may also respond better to neuroleptic treat- ASD, who tend to experience adverse effects very fre-
ment than to typical first-line psychostimulants, in low- quently (McDougle et al., 1996, 2000; Sugie et al.,
functioning autistic individuals or in patients who 2005). Also, citalopram is not more effective than pla-
develop greater agitation when administered psychosti- cebo on repetitive behaviors in children and adolescents
mulants. Once problem behaviors are reduced, behav- with ASD, and is more likely to be associated with
ioral and educational interventions with autistic adverse effects, including increased energy level, impul-
children and adolescents often develop greater efficacy. siveness, decreased concentration, hyperactivity, stereo-
SGAs are equally or more effective than typical neuro- typy, diarrhea, insomnia, dry skin or itching, and
leptics and safer in terms of side effect profile. Typical prolonged seizure (King et al., 2009). There are no RCTs
neuroleptics remain a viable option when ASD patients of paroxetine, sertraline, escitalopram. A case report pro-
are nonresponsive to at least two SGAs, noncompliant vides anecdotal evidence of efficacy in the treatment of
to several SGAs due to important side effects, or when repetitive behaviors, anxiety, and temper tantrums for
SGAs, including clozapine, are contraindicated due to paroxetine in a 7-year-old nonresponder to fluoxetine
documented medical reasons. and fluvoxamine (Posey et al., 1999). One open-trial pro-
vides some evidence of efficacy for paroxetine on self-
injurious behavior in intellectual disability (Davanzo
SELECTIVE SEROTONIN REUPTAKE
et al., 1998). A retrospective study of 37 adults with intel-
INHIBITORS
lectual/neurologic disabilities and autistic traits reported
To date, three selective serotonin reuptake inhibitors improvement in only 13 cases and a high incidence
(SSRIs) have received regulatory approval in Europe of side effects, including vomiting, psychomotor agita-
for use in the pediatric population: fluoxetine for moder- tion, drowsiness, hypomanic behavior, aggressiveness,
ate to severe major depressive episodes starting at age 8, insomnia, seizures, and skin rushes (Branford et al.,
and sertraline and fluvoxamine for OCD after 6 and 1998). The effectiveness of sertraline in ASD children
8 years of age, respectively (Persico et al., 2015). These and adults is mainly linked to anxiety disorder. One case
drugs raised initial hopes of clinical usefulness in ASD, series of nine autistic children, aged 6–12 years, docu-
based on their well-known efficacy on obsessive– ments improvement in anxiety, irritability, and agitation
compulsive symptoms and on 5-HT blood levels, found with clinical response to low doses (25–50 mg/day)
elevated in approximately 25% of autistic individuals appearing generally in 2–8 weeks and, frequently, wan-
(Gabriele et al., 2014). Contrary to this expectation, ing after a few months (Steingard et al., 1997). Hence,
results with SSRIs in ASD have been generally disap- short-term sertraline treatment can reduce behavioral
pointing. Overall, RCTs involving fluoxetine, fluvoxa- reactions associated with situational transitions or envi-
mine, fenfluramine, and citalopram provided modest ronmental changes in children with ASD, though bene-
and conflicting evidence of efficacy on repetitive behav- ficial effects may be temporary. An open-label study
iors and compulsions (Hollander et al., 2005, 2012; showed significant improvement in repetitive and
Williams et al., 2013; Mouti et al., 2014), but fluoxetine aggressive symptoms, but not in social deficits, in
has been shown to increase regional blood flow in the 24 of 42 (57%) adults with ASD (McDougle et al.
anterior cingulate gyrus and orbitofrontal cortex 1998). The most common adverse effects were weight
(Buchsbaum et al., 2001). The antianxiety effects of flu- gain, anxiety, and psychomotor agitation. There is only
oxetine may also explain the occasional observation of one open-label study on 28 ASD children treated with
some improvement in (a) core ASD symptoms, including escitalopram, reporting significant improvement in
an increase in eye contact, social initiation and respon- CGI-I ratings and in ABC scores for irritability, lethargy,
siveness, and repertoire of interests; (b) irritability and stereotypy, hyperactivity, and inappropriate speech.
problem behaviors, including decreased tantrums and Dose-related adverse effects, notably irritability and
aggression toward self and others; (c) attention deficit hyperactivity, occurred at doses above 10 mg/day in
(initiating, shifting, and sustaining attention), leading 78% of cases (Owley et al., 2005).
altogether to better social adaptation and inclusion In summary, SSRIs are definitely more effective in
(Hollander et al., 2012). However, improvement is neurotypical individuals suffering from OCD and
often clinically marginal and side effects, especially depression than in autistic individuals affected by
396 A.M. PERSICO ET AL.
obsessive and anxious–depressive symptoms. Second, reducing inappropriate sexual behavior associated with
they appear relatively more effective and better tolerated ASD (Nguyen and Murphy, 2001; Albertini et al.,
in adult autistic individuals, as compared to ASD chil- 2006; Coskun and Mukaddes, 2008; Coskun et al.,
dren and adolescents. Nonetheless, a trial with an SSRI 2009). The efficacy of trazodone, a heterocyclic antide-
should be considered whenever compulsive behavior pressant, has been the object of only a few case reports
and anxiety are posing major hurdles to developmental, involving ASD children. One case report showed a
educational, and social progress. A valid trial should last reduction of aggression at a dose of 150 mg/day in
10–12 weeks but, contrary to nonautistic patients, shorter divided doses (Gedye, 1991); another case study
trials (i.e., 1–3 weeks) may suffice and prevent subse- reported beneficial effects of trazodone on sleeping in
quent behavioral activation. Low SSRI doses should an 8-year-old autistic boy also treated with fluvoxamine
be prescribed with frequent clinical monitoring of target (Parker and Hartman, 2002); priapism was described as a
symptoms and potential side effects. dose-dependent side effect in a 13-year-old autistic boy
(Kem et al., 2002).
SEROTONIN AND NOREPINEPHRINE
ANTIANXIETY MEDICATIONS
REUPTAKE INHIBITORS AND OTHER
ANTIDEPRESSANT MEDICATIONS Antianxiety agents such as benzodiazepines have been
relatively understudied in ASD. They are clinically use-
Among serotonin and norepinephrine reuptake inhibitors
ful at times in the treatment of anxiety, particularly as
(SNRIs), more data are available for venlafaxine, which
add-on to an SSRI. However, behavioral side effects such
inhibits the reuptake of serotonin, norepinephrine, and,
as disinhibition, crying, and irritability limit their use.
to a lesser extent, dopamine (Ellingrod et al., 1994). Sig-
Instead, two RCTs have been performed on buspirone,
nificant improvement in repetitive behaviors, restricted
a 5-HT1A receptor agonist. An 8-week RCT involving
interests, social deficits, communication and language,
40 ASD children and adolescents showed that low-dose
inattention, and hyperactivity were reported in an open,
buspirone is more effective than placebo as add-on
retrospective study involving 10 autistic children,
to risperidone for treating irritability (Ghanizadeh
adolescents, and young adults (Hollander et al., 2000).
and Ayoobzadehshirazi, 2015). At a mean dose of
Clinical response was observed at low doses
6.7 mg/day, 13/16 (81.2%) buspirone-treated patients
(6.25–50 mg/day, mean daily dose 24.37 mg) and the
showed a 30% decline in irritability scores compared
drug was well tolerated. Another open trial reported
to 7/18 (38.9%) patients in the placebo group. Contrary
beneficial effects of low-dose venlafaxine on self-injury
to buspirone alone, which tends to decrease appetite and
and hyperactivity in two 17-year-old autistic boys
body weight, the most common adverse effect adding
and in a 23-year-old autistic woman (Carminati et al.,
buspirone onto risperidone was increased appetite, as
2006). A recent RCT found no convincing evidence of
well as drowsiness and fatigue. Another recent multicen-
efficacy, contrasting six patients receiving venlafaxine
ter RCT enrolled 166 ASD preschoolers aged 2–6 years
(18.75 mg/day), along with their usual treatment (zuclo-
randomized to 24 weeks of 2.5/5.0 mg of buspirone twice
penthixol and/or clonazepam), with seven patients who
daily vs placebo (Chugani et al., 2016). No difference in
received placebo plus usual care (Carminati et al.,
ADOS Composite Total Score among the 3 treatment
2016). At the end of the study, global improvement
groups was recorded at 24 weeks, but the ADOS
was observed in 33% and 71% of venlafaxine- and
Restricted and Repetitive Behavior score showed a
placebo-treated patients, respectively, with irritability
time-by-treatment effect: 2.5-mg buspirone produced
improving equally in the entire sample. Only marginal
significant improvement, whereas placebo and 5.0-mg
evidence of lower cumulative doses of clonazepam and
buspirone yielded no change. Adverse events did not dif-
zuclopenthixol in the venlafaxine-treated group was
fer significantly among treatment groups. This study thus
obtained. Mirtazapine, a TCA, acting as an antagonist
supports low-dose buspirone treatment (2.5 mg/day) as
at a2 adrenergic and at multiple serotonin receptors,
an adjunct therapy to target restrictive and repetitive
improved aggressiveness, self-injury, irritability, hyper-
behaviors in conjunction with behavioral interventions.
activity, anxiety, depression, and insomnia in 9 of
26 (34.6%) ASD subjects, according to one open-label
ANTIEPILEPTICS AND MOOD
study (Posey et al., 2001). Mirtazapine did not improve
STABILIZERS
core social and communication deficits. Adverse effects
were minimal and included increased appetite, irritabil- Antiepileptic drugs (AEDs) have been extensively
ity, and transient sedation. Other case reports suggest employed in ASD as mood stabilizers and to attenuate
efficacy for this drug, at a dose ranging from 5 to problem behaviors, but only a limited number of open-
30 mg/day, not only on aggressiveness but also in label studies and RCTs have been performed in autistic
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 397
children and adolescents (Hirota et al., 2014; significantly lower (Wang et al., 2017). EEG normaliza-
Canitano, 2015). tion was significantly more frequent in the treatment
Valproic acid was found to attenuate irritability in an group (Wang et al., 2017).
initial open trial including 14 young ASD patients (with Topiramate is an interesting drug, because of its
and without seizures) (Hollander et al., 2001). Three sub- weight-containing and mood-stabilizing effects, but the
sequent RCTs enrolling 12–30 ASD subjects yielded few studies performed to date have yielded conflicting
contrasting results (Hellings et al., 2005; Hollander results. An 18-month open-trial involving 10 ASD chil-
et al., 2006b, 2010). Hellings et al. (2005) found no sig- dren and adolescents undergoing weight gain with SGA
nificant difference in ABC irritability scores, with high treatment reported modest variation in weight excess and
interindividual variability and large placebo effect. On frequent adverse behavioral effects, dampening enthusi-
the contrary, using more severe symptomatic inclusion asm for topiramate add-on use (Canitano, 2005). Simi-
criteria to reduce interindividual variability, Hollander larly, limited efficacy and frequent side effects were
et al. (2010) showed a significant advantage of sodium reported in another case series (Mazzone and Ruta,
divalproex over placebo. Furthermore, Hollander et al. 2006). However, an 8-week RCT involving 40 autistic
(2006b) reported significant improvement in repetitive children, 4–12 years old, randomly allocated to receive
behaviors in a small RCT involving 13 autistic children. either topiramate + risperidone (Group A) or placebo
Valproate was also found to attenuate the irritability asso- + risperidone (Group B), showed beneficial effects for
ciated with fluoxetine treatment in ASD significantly topiramate on irritability, stereotypic behaviors, hyperac-
better than placebo (Anagnostou et al., 2006). tivity, and/or noncompliance (Rezaei et al., 2010). Lamo-
Levetiracetam binds to the synaptic vesicle glycopro- trigine, an AED mainly active in reducing voltage-gated
tein SV2A, inhibiting presynaptic calcium channels and Na+ currents, showed no efficacy on irritability and
reducing neurotransmitter release. This pharmacologic social behavior in an RCT involving 27 autistic children
action limits nerve signal conduction across synapses aged 3–11 years old (Belsito et al., 2001). The use of
(Lynch et al., 2004; Vogl et al., 2012). The use of levetir- oxcarbazepine for irritability and agitation in ASD has
acetam in ASD is controversial due to several studies been addressed in only 1 case series, involving
reporting major behavioral side effects. Levetiracetam 30 patients, aged 5–21 years, taking on average 2 other
was initially found to improve attention, hyperactivity, psychotropic medications, including SGAs, SSRIs,
emotional lability, and aggressiveness in an open trial alpha agonists, and divalproex (Douglas et al., 2013).
involving six drug-naive ASD boys (Rugino and Fourteen patients (47%) achieved a CGI-I score of
Samsock, 2002). Wasserman et al. (2006) compared “much improved,” while another 10 patients (33%) were
levetiracetam to placebo in a sample of 20 ASD children, “improved”; 7 patients (23%) had a clinically significant
aged 5–17 years, and found no evidence of efficacy on adverse effect leading to drug discontinuation. Finally, a
global functioning, irritability, repetitive behaviors, recent case report points toward possible beneficial
impulsivity, and hyperactivity. The most relevant side effects of low-dose phenytoin (5 mg/day for over
effect was aggression. Levetiracetam was also found 18 months) on verbal and nonverbal communication in
ineffective for controlling stereotyped movements in a 19-year-old autistic young man treated with stimulant
patients with ASD (Rajapakse and Pringsheim, 2010). medication since early childhood (Bird, 2015).
Importantly, one case report documents an autistic Lithium is a “classical” mood stabilizer also used in
regression, characterized by stereotypic behaviors and autistic individuals with comorbid recurrent affective
severe social and communicative dysfunction after disorder and/or mood instability, especially in the pres-
administration of levetiracetam in a 6-year-old girl with ence of mania, euphoria, or elevated mood. There are
cerebral palsy, whose main symptoms were mild intellec- currently no RCTs of lithium in children and adolescents
tual disability and focal epilepsy. This regression sub- with ASD. Two older case reports document the efficacy
sided after drug discontinuation, suggesting that of lithium therapy, the first one at blood levels
levetiracetam may occasionally provoke ASD-like >1.0 mEq/L on two 4-year-old autistic children with
behavioral adverse effects (Camacho et al., 2012). hyperactivity resistant to stimulant drugs and with cycli-
Finally, 70 children with ASD and subclinical epilepti- cal mood swings (Kerbeshian et al., 1987); the second
form discharges (SEDs) identified by EEG were ran- case report describes a beneficial response to lithium in
domly divided into two equal groups treated either two individuals with ASD whose symptoms included
with levetiracetam (60 mg/kg/day) and educational train- agitation, self–injurious behavior, aggression, and
ing (treatment group) or with educational training only insomnia (Steingard and Biederman, 1987). In a more
(control). At the 6-month follow-up, PEP-3 scores were recent retrospective chart review of 30 children and ado-
significantly higher for the treatment group compared lescents with ASD treated with lithium because of
to controls, whereas CARS and ABC scores were comorbid bipolar disorder, 43% were rated as improved
398 A.M. PERSICO ET AL.
on CGI-Irritability scores and 71% of patients with two possibly more in children than in adults. However,
or more pretreatment mood disorder symptoms were response rates tend to be somewhat lower, and adverse
rated as improved (Siegel et al., 2014). Patients most effects more frequent, in autistic individuals compared
improved were those with mania and elevated mood. to typically developing children with ADHD
Mean lithium blood level was 0.70 mEq/L, and the mean (RUPPAN, 2005b). Interindividual variability in
duration of lithium treatment was 29.7 days. At least one response to MPH by autistic children may be influenced
adverse effect was reported by 47% of patients, most by multiple monoaminergic gene variants (McCracken
commonly vomiting (13%), tremor (10%), fatigue et al., 2014). Six RCTs of MPH in children with ASD
(10%), irritability (7%), or enuresis (7%) (Siegel have been performed to date. In the initial RUPPAN
et al., 2014). study (2005b), 66 of 72 children, aged 5–14 years, with
A very interesting case report describes the efficacy of a DSM-IV diagnosis of Autistic Disorder or Pervasive
lithium as a rescue therapy for regression and catatonia and Developmental Disorder-Not Otherwise Specified
autistic traits in a 21-year-old man and in a 17-year-old girl (PDD-NOS) were randomized to a double blind cross-
with Phelan–McDermid syndrome, due to chr 22q13.33 over study of MPH vs placebo. A significant improve-
deletion involving the SHANK3 gene (Serret et al., ment was recorded on the ABC hyperactivity subscale,
2015). The former patient, unresponsive to multidrug ther- with 49% of participants responding at all MPH dose
apies including antipsychotics and other psychotropic increments and maximum benefits at medium-high doses
drugs, responded to 1500 mg/day of lithium, yielding (0.25 and 0.5 mg/kg per dose, respectively). Improve-
blood levels of 0.8 mEq/L. Lithium successfully corrected ment in hyperactivity/impulsivity and inattention, with-
a severe regression characterized by increasing psycho- out significant worsening of stereotypic or oppositional
motor agitation, aggressiveness, impulsivity, urinary, and defiant behavior, was later confirmed in the same sample
bowel incontinence, promoting cognitive and social using other outcome measures, such as SNAP-IV and
improvement over the subsequent 3-month period. The CY-BOCS scores (Posey et al., 2007), with improvement
latter patient underwent a period of severe behavioral again most evident at 0.25–0.50 mg/kg doses. The most
regression and catatonia. One year of lithium treatment common adverse effects of MPH included irritability,
(1000mg/day, lithium blood level 0.7 mEq/L), in associa- appetite loss, insomnia, and emotional outbursts. Similar
tion with a low dose of clonazepam (0.2 mg/day), spurred results were obtained in other RCTs involving autistic
improvement in motor and verbal abilities allowing the children (Quintana et al., 1995; Handen et al., 2000;
patient to recover to similar levels of functioning as Jahromi et al., 2009). Dosage for ASD children ranges
before regression. In both cases lithium was well tolerated. from 7.5 to 50 mg/day, sometimes divided and often
Another case report is on an adult male with Phelan– dosed by weight (0.3–0.6 mg/kg/day); for preschool chil-
McDermid syndrome whose mood and behavior dren ranges between 5 and 20 mg/day in divided doses
responded to a combined treatment with lithium and olan- (Doyle and McDougle, 2012). A 4-week RCT conducted
zapine, following resistance to various combinations of by Pearson et al. (2013) in 24 ASD children, with mean
SGAs and AEDs (Egger et al., 2017). age 8.8 years and comorbid ADHD, demonstrated ben-
Future prospective studies of lithium are needed to eficial effects on hyperactivity, with the combination
assess whether lithium strictly benefits comorbid bipolar of extended-release MPH in the morning and
symptoms or improves some ASD-specific symptoms. immediate-release MPH in the afternoon. This study is
especially interesting, because this combination is fre-
quently applied in clinical practice. In a recently pub-
STIMULANTS AND a2 ADRENERGIC
lished RCT (Kim et al., 2017), 27 subjects, aged 5–17
AGONISTS
years with ASD and ADHD, were randomized to receive
The comorbidity between ASD and ADHD, now recog- either low (10 mg/day) or medium (30 mg/day) doses of
nized by DSM-5, is present in 30%–40% of autistic indi- an extended-release liquid formulation of MPH. Signif-
viduals (Berenguer-Forner et al., 2015). This high icant decreases in irritability and hyperactivity ABC
incidence and the negative impact of inattention, hyper- scores were recorded more frequently in the medium
activity, and impulsivity on treatment programs and on dose group (mean dose ¼ 20.28 mg/day). Also, rebound
social adjustment, have spurred interest in the use of psy- hyperactivity and aggression at the end of the day were
chostimulants in ASD patients with comorbid ADHD. significantly lower only in the medium-dose group.
Methylphenidate (MPH) acts as a dopamine and, to a In several studies, and also in clinical practice, intel-
lesser extent, as a norepinephrine reuptake blocker. It lectual disability and lower IQ tend to be associated with
is the psychostimulant most prescribed for children with lower response rates to MPH and with more side effects
ADHD. In general, MPH is known to be moderately (Aman et al., 2003). However, at least 1 RCT involving
efficacious in improving ADHD symptoms in ASD, 122 children and adolescents with severe ADHD and
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 399
intellectual disability (including many with comorbid norepinephrine reuptake blocker, in reducing depressive
ASD) found MPH (0.5/1.0/1.5 mg/kg/day) significantly and inattentive/hyperactive symptoms (Golubchik et al.
superior to placebo and no correlation between IQ 2013). A significant, yet modest, decrease in the severity
or ASD symptoms and clinical response (Simonoff of depressive and ADHD symptoms assessed with
et al., 2013). CDRS and ADHD-RS, respectively, was recorded.
Atomoxetine is a selective dopamine and norepineph- A significant positive correlation was found between
rine reuptake inhibitor, approved for the treatment of changes in depression and ADHD severity scores. Most
ADHD in children, adolescents, and adults starting at patients reported minimal or tolerable side effects.
6 years of age. A 4-week RCT performed on 97 ASD Clonidine is a selective a2 adrenergic and
individuals with comorbid ADHD, aged 6–17, found imidazoline-1 receptor agonist, approved by the FDA
some evidence of efficacy for atomoxetine (1.2 mg/kg/ in 2010 for the treatment of ADHD in pediatric patients
day) on hyperactivity, stereotyped behaviors, inappro- starting at age 6. Two RCTs provided evidence of effi-
priate speech, and fear of change, but no significant cacy on impulsivity, hyperarousal, and self-stimulating
global improvement using the CGI; side effects included behavior in ASD children and adults (Fankhauser
nausea, decreased appetite, fatigue, and early morning et al., 1992; Jaselskis et al., 1992). This drug is also effec-
awakenings (Harfterkamp et al., 2012). A subsequent tive on hyperactivity, aggressiveness, mood instability,
open-label follow-up study conducted on 88 patients irritability, and on nighttime awakenings, improving
taking the drug for 28 additional weeks showed impro- sleep quality (Ming et al., 2008). In these studies, drug
vement in hyperactivity, impulsivity, and inattention dosage ranges from 0.1 to 0.2 mg/day and side effects
(Harfterkamp et al., 2013). Adverse events, including included drowsiness, sedation, hypertension, and
gastrointestinal symptoms, irritability, tinnitus, mood decreased activity. Guanfacine is a central a2 adrenergic
swings, and sedation, were generally mild and tended agonist. A retrospective study of 80 ASD children and
to diminish over time, especially nausea and fatigue. adolescents, aged 8–18 years, reported improvement in
Dosages generally ranged from 1.2 to 1.4 mg/kg/day. hyperactivity, inattention, insomnia, and tics (Posey
Another recent RCT in 128 ASD children (ages 5–14) et al., 2004a). An open trial found 12 out of 25 (48%)
lasting 6 weeks for dose adjustment based on clinical ASD children, either nonresponders or intolerant to
response (up to a maximum of 1.8 mg/kg/day), followed MPH, responded well to guanfacine (Scahill et al.,
by dose maintenance for an additional 4 weeks, con- 2006). The efficacy of this drug in improving hyperactiv-
firmed that: (a) the most frequent side effects are ity in ASD children was then confirmed by two RCT
decreased appetite and fatigue, (b) most side effects do studies (Handen et al., 2008; Scahill et al., 2015). Dos-
not raise concern and last 4 weeks of less, and ages ranged between 0.25 and 9 mg/day, often in divided
(c) unlike typically developing children, ASD children doses. The most frequent side effects included irritability,
did not display serious adverse events, including QTc drowsiness, decreased energy, sleep disturbance, consti-
changes or suicidal ideation (Tumuluru et al., 2017). pation, headache, and nocturnal enuresis (Handen et al.,
An interesting recent RCT demonstrates the effective- 2008; Scahill et al., 2015).
ness of atomoxetine over placebo, underscoring at the
same time the usefulness of parent training in the context
FUTURE PERSPECTIVES IN THE
of an effective multidisciplinary treatment approach. In
PHARMACOTHERAPY OF AUTISM
particular, the combination of atomoxetine + parent train-
SPECTRUM DISORDER
ing (ATX + PT) produced higher response rates with
lower atomoxetine doses compared to the doses pre- The lack of psychoactive drugs able to ameliorate the
scribed in the “atomoxetine alone” group. Meanwhile, “core” symptoms of ASD is currently felt as perhaps
ATX + PT and ATX alone were superior to placebo the main unmet need in pediatric neuropsychophar-
+ PT and to placebo alone, respectively, in improving macology (Persico et al., 2015). For this reason, an
ADHD symptoms (Handen et al., 2015). A subsequent impressive effort is being put forth by academia and
24-week extension study involving 117 children, aged pharmaceutical companies to move the field of autism
5–15, out of the 128 (91%) acute-trial participants, con- psychopharmacology forward. No other behavioral
firmed the efficacy of atomoxetine, providing further evi- disorder of childhood and adolescence is the target of
dence of the greater benefits obtained by combining as many ongoing clinical trials listed on the NIH web
pharmacologic and psychosocial treatments over either site https://clinicaltrials.gov/. Some experimental drugs
treatment alone, both in terms of treatment outcome derive from animal and human research on monogenic
and patient compliance (Smith et al., 2016). forms of syndromic autism, for example, Rett syndrome,
An open-label trial of 11 children with ASD explored fragile X, tuberous sclerosis, and Phelan–McDermid
the safety and efficacy of reboxetine, a selective syndrome, among many. Other drugs derive from
400 A.M. PERSICO ET AL.
cellular, animal, molecular, and human studies of idio- Both oxytocin and vasopressin have a short half-live
pathic ASD, whose pathogenetic underpinnings are of around 3 min in plasma and 20 min in CSF, where they
beginning to be relatively well understood. Drugs are inactivated by placental leucine aminopeptidase
derived from research on syndromic forms were initially (LNPEP) (Gazis, 1978). Therefore, only through the
spurring high expectations, because in mouse models nasal route is it possible to ensure that a substantial
they displayed a surprising degree of effectiveness when amount of the active ingredient reaches the CNS, given
administered not only prenatally, but also postnatally or its peptide nature and half-life; in fact, if this therapy was
even in adult animals. Unfortunately, these drugs have administered parenterally, only 1% of these neuropep-
not met their primary endpoints in human RCTs, once tides would reach the CNS unmodified.
again proving that animal and cellular models can indeed Based on these data, intranasal oxytocin and vaso-
contribute preliminary but never conclusive evidence of pressin, as well as the AVPR1a antagonist RG7314,
efficacy, given the exquisite complexity of the are under scrutiny. Intranasal oxytocin, certainly the most
human brain. studied compound in current experimental ASD pharma-
On the other hand, among drugs derived from patho- cology, yields insufficient evidence of efficacy on core
genetic studies of idiopathic ASD, some may display ASD symptoms, according to two recent meta-analyses
measurable efficacy on core ASD symptoms, although including 17 and 12 RCTs, respectively (Keech et al.,
with different levels of evidence. We hereby summarize 2017; Ooi et al., 2017). Given the genetic variability of
the most studied and/or the most promising pharma- the OXTR gene and anatomical differences in nasal cav-
cologic agents in the experimental pipeline for the ity influencing oxytocin absorption into the CNS, this
treatment of core autistic symptoms: interindividual variability in response to intranasal
oxytocin in ASD is not surprising.

Oxytocin and vasopressin Bumetanide


Oxytocin and arginine-vasopressin, synthesized by the Bumetanide is a selective diuretic for Cl, acting as a
magnocellular neurons of the supraoptic nucleus and high affinity antagonist for the NKCC1 transporter
the magnocellular and parvocellular neurons of the para- (Feit, 1981). The transmembrane Cl gradient is pro-
ventricular hypothalamic nucleus, in addition to duced by the activity of two cotransporters, NKCC1
“classic” hormonal functions, also act as neurotransmit- importing Cl into the cell, and KCC2 extruding Cl into
ters, released in extrahypothalamic regions. Dense the extracellular space (Payne et al., 2003). In the adult
expression of oxytocin receptors (OXTR) is present in brain, Cl is more concentrated in the extracellular fluid,
regions critical to social cognition, attachment, empathy, due to KCC2 being more expressed than NKCC1, so that
and motivation, such as the anterior cingulate cortex, the the opening of GABA-A channels produces Cl entry
temporo-parietal junction, the insula, and the nucleus into the neuron and hyperpolarization. Instead, during
accumbens, while AVPR1a receptors are expressed in prenatal and early postnatal life, NKCC1 is more
the hippocampus and the amygdala (Kirsch et al., expressed than KCC2, inverting the Cl gradient and
2005; Knafo et al., 2008; Insel, 2010; Meyer- causing depolarization when GABA-A channels open
Lindenberg et al., 2011; Wigton et al., 2015). The (Ben Ari et al., 2007). In neurodevelopmental disorders
“social role,” mediated by OXTR and the AVPR1a recep- like autism, intellectual disability, and epilepsy, this
tors for oxytocin and vasopressin, respectively, was immature configuration often persists at later ages also,
discovered while studying the differences in attachment contributing to the excess of excitation over inhibition
between prairie and mountain voles, two different present in ASD and causing the so-called “paradoxical
rodents species, which show strong and weak attach- effect” of benzodiazepines, i.e., a pharmacologic induc-
ment, respectively, linked to differential brain OXTR tion of psychomotor agitation instead of anxiolysis and
and AVPR1a gene expression in limbic regions (Carter sedation (Marrosu et al., 1987). Bumetanide aims to
et al., 1995; Insel, 2010; Johnson and Young, 2015; reduce this imbalance, by promoting Cl outflow from
Johnson et al., 2016). Oxytocin and vasopressin thus excitable neurons through renal excretion.
act, in females and males, respectively, as critical medi- One open-label study conducted on eight teenagers
ators of maternal, social, and mating behaviors (Lucht with ASD initially provided promising results. Bumeta-
et al., 2009; Insel, 2010; Johnson and Young, 2015). nide led to improvement in the ability to recognize and
Not surprisingly, much evidence supports the contribu- characterize the emotions expressed by the face follow-
tion of a dysfunctional oxytocin–vasopressin system to ing an activation of the brain regions involved in facial
the pathogenesis of ASD (Jacob et al., 2007; Gregory recognition and social and emotional perception, such
et al., 2009; Yamasue et al., 2009; Zhang et al., 2017). as the fusiform gyrus and the inferior occipital gyrus
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 401
(Hadjikhani et al., 2015). Two subsequent RCTs pro- studies, suggesting beneficial effects on social interac-
duced significant improvement of autistic behaviors tion and sensory self-stimulation (Rossignol and Frye,
measured using the CARS, SRS, and CGI; parents 2014b). Two RCTs published to date have given contra-
defined their children as “present,” with a greater capac- dictory results, but the two studies differ substantially in
ity for integration and communication with the environ- many regards. A study on 121 children and teenagers
ment, signaling few side effects (Lemonnier et al., 2012, with ASD did not provide evidence of efficacy, although
2017). Maximum efficacy was recorded at the dose of tolerability was found to be reassuring (Aman et al.,
1 mg twice a day, maximum safety at 0.5 mg b.i.d. The 2017). On the contrary, the addition of memantine to ris-
most frequent adverse events were hypokalemia, enure- peridone in 40 autistic children showed further improve-
sis, dehydration, appetite loss, and muscle weakness ment in irritability, stereotypies, and hyperactivity
(Lemonnier et al., 2017). (Ghaleiha et al., 2013). Additional studies are ongoing
to test memantine effects on memory, communication
Sulforaphane and spoken language, motor skills, problem behaviors,
and cognition in ASD.
Sulforaphane is a natural compound especially present in
broccoli sprouts, endowed with antioxidant and antineur-
oinflammatory effects. An initial RCT showed sizable Arbaclofen (STX209)
improvement in ABC scores for irritability, lethargy, ste- The known imbalance between excitatory and inhibitory
reotypies, and hyperactivity, as well as in SRS scores for neurotransmission, as well as studies clarifying the com-
awareness, communication, motivation, and mannerisms plex interface between GABA-B, mGluR5 and AMPA
(Singh et al., 2014). Beneficial effects on social interac- receptors in the pathophysiology of fragile-X syndrome,
tion, aberrant behaviors, and verbal communication were have created interest in arbaclofen, the prodrug precursor
recorded using the CGI-I. The substance was very well of the GABA-B receptor agonist R-baclofen (Silverman
tolerated and this substance is currently being tested in et al., 2015), but having greater stability and
several ongoing RCTs (Singh et al., 2014). manageability.
In a first open study, 32 autistic children and teenagers
Glutamatergic drugs: Memantine and received 10 mg of arbaclofen a day (6–11 years) or 10 mg
D-cycloserine three times a day (12–17 years). A significant decrease
D-Cycloserine acts as a partial agonist of the glycine in irritability and social withdrawal, paired with an
binding site at NMDA receptors. By modulating glycine increase in motivational drive towards social interaction,
action as a cotransmitter at the NMDA receptor, this drug was observed in approximately half of the sample
is predicted to promote longer channel openings with (Erickson et al., 2014). A subsequent RCT, however,
increased Ca2+ entry, ultimately enhancing connectivity did not achieve its primary outcome (social withdrawal
and consolidating environmentally induced plastic mod- ABC scores) (Veenstra-VanderWeele et al., 2017),
ifications in excitatory circuits. An initial single-blind although secondary outcomes on severity, assessed with
pilot study in 10 autistic patients showed a significant the CGI-S and the VABS-II, gave a positive result. Side
improvement in social withdrawal (Posey et al., effects were mild (emotional lability and drowsiness). As
2004b). Two subsequent RCTs (Urbano et al., 2014, methodological limitations may have had a negative
2015; Minshawi et al., 2016; Wink et al., 2017) have impact on this study, further studies are ongoing.
shown significant clinical improvement in core ASD
symptoms (Urbano et al., 2015) as well as in stereotyped Supplements and probiotics
behaviors (Urbano et al., 2014), with a synergistic effect
A variety of supplements have been clinically assessed,
in behavioral intervention, fully compatible with the
under the assumption that ASD children may benefit
activity-mediated mechanism of action of this drug.
from natural substances endowed with antioxidant, anti-
The only frequent side effect was a modest increase in
inflammatory, energy-stimulating, or sleep inducing
irritability.
properties. Although the methodological quality of RCTs
Memantine acts as a noncompetitive low-affinity
in natural medicine does not always meet minimum stan-
NMDA receptor antagonist (Chen et al., 1992;
dards of reliability, it is worth briefly listing the major
Kornhuber et al., 1989). Its short-lived receptor blockade
areas of interest, as some clinically useful applications
kinetics prevents negative effects on learning and mem-
may well come forth in the near future.
ory (Lipton, 2006; Danysz and Parsons, 2012). Its bene-
ficial effects on synaptogenesis and neural networking, ● Antioxidants: N-acetylcysteine (NAC), carnosine,
demonstrated in different animal and cellular models, vitamin B12, folinic acid, methyl-B12, glutathione.
led to several prospective and retrospective open-label These substances should help counteract the
402 A.M. PERSICO ET AL.
accumulation of reactive oxygen species (ROS) pre- (Kempuraj et al., 2008; Kritas et al., 2013). At least
sent in ASD individuals (Kern and Jones, 2006). three open-label studies from the same group have
RCTs have provided conflicting results for most of yielded promising results in ASD children and ado-
these substances and primary outcome measures do lescents, especially in eye contact, attention, social
not usually show a statistically significant effect. deficits, problem behaviors, and adaptive skills
Interestingly, folinic acid may improve language, (Theoharides et al., 2012; Taliou et al., 2013;
attention, and stereotypical behaviors specifically Tsilioni et al., 2015). These results still await confir-
in ASD children carrying autoantibodies capable of mation by RCTs. Finally, o-3 fatty acids are potent
binding or blocking the FRa receptor (Frye et al., antiinflammatory and immunoregulatory agents
2013; Ramaekers et al., 2013). Hence, targeted used in the treatment of a wide variety of diseases
patient populations may well benefit from these associated with chronic inflammation. According
compounds. to a recent meta-analysis of 6 international RCTs,
● Energizers: Q10 ubiquinol and L-carnitine. Coen- including 194 autistic patients, the data indicate an
zyme Q10 is a key element in the mitochondrial improvement in hyperactivity, lethargy, and stereo-
respiratory chain. Q10 ubiquinone is reduced to types, but no effects on global functioning or on
Q10 ubiquinol in the liver, and the latter represents socialization and communication deficits (Cheng
the active compound, able to exert an important anti- et al., 2017).
oxidant action and to stimulate energy metabolism. ● Sleep-inducing agents: melatonin. Sleep distur-
At least two RCTs have assessed the effect of Q10 bances are especially frequent in ASD and may be
ubiquinone in 20 and 55 children with ASD, aged related to reduced melatonin synthesis and enhanced
between 5 and 16 years, and shown improvements kynurenine pathway activation due to neuroinflam-
on the Parental Global Impressions-Revised scale mation (Gevi et al., 2016). Melatonin, quite possibly
as well as in subscales for hyperactivity, opposition, the most widely prescribed compound for the man-
and receptive language. Unfortunately Q10 ubiqui- agement of autistic children, may thus represent
none was administered in the context of multivitamin more a “substitution” therapy than a pharmacologic
and mineral supplements such NAC, choline, and intervention. Seven RCTs have proven melatonin
inositol, complicating the interpretation of these effective in ameliorating total sleep duration and
results (Adams and Holloway, 2004; Adams et al., sleep onset latency (Rossignol and Frye, 2014a).
2011). Carnitine allows the transport of fatty acids Similar improvements were recorded using
across the inner mitochondrial membrane, stimulating prolonged-release melatonin, according to one
the production of ATP. Blood levels of carnitine would recent RCT involving 125 ASD children and adoles-
be reduced in some patients with ASD (Filipek et al., cents (Gringras et al., 2017).
2004), and 1 RCT involving 34 ASD children aged ● Gut-modifying agents. Probiotics are live microor-
3–10 years showed significant improvement in core ganisms that can stabilize the mucosal barrier by
autistic symptoms with supplementation, reporting increasing the expression of mucin, reducing exces-
only minimal side effects, such as irritability and sive bacterial growth, stimulating mucosal immunity
intestinal discomfort (Geier et al., 2011). (secretory IgA), and synthesizing antioxidant sub-
● Neuroinflammation and autoimmunity: minocycline, stances (Fond et al., 2015). The high prevalence of
luteolin, and quercetin, omega-3 fatty acids. Neu- gastrointestinal disorders in ASD, the significantly
roinflammation is an active mechanism in the CNS skewed gut microbiome found in many autistic chil-
of autistic individuals from relatively early ages dren, and the interesting results obtained in animal
(Vargas et al., 2005; Garbett et al., 2008). Also, auto- models with probiotics (Buie et al., 2010; Hsiao
immunity appears to enhance ASD severity in some et al., 2013; Tomova et al., 2015) led to interest in
patients (Piras et al., 2014). Minocycline, a semisyn- testing probiotics for clinical efficacy in ASD. To
thetic derivative of tetracyclines with neuroprotec- date, evidence of their efficacy is still limited to sev-
tive and antiinflammatory effects, has not shown eral open trials reporting benefits at the gastrointes-
significant efficacy in a pilot open-label study tinal and/or behavioral level (Adams et al., 2011;
(Pardo et al., 2013). A subsequent RCT of minocy- Kaluzna-Czaplinska and Blaszczyk, 2012; Grossi
cline as add-on therapy to risperidone has yielded et al., 2016; Kang et al., 2017; Shaaban et al., 2017).
modest improvements (Ghaleiha et al., 2016). Luteo-
lin and quercetin, flavonoid compounds that are sec-
RETT SYNDROME
ondary metabolites of plants and present in various
foods, are endowed with powerful antioxidant and The pharmacologic therapy of Rett syndrome is
antiinflammatory actions, especially on mast cells described here, because this monogenic disorder
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 403
represents an important example of syndromic autism. control. Often, RTT comes with many associated comor-
Rett syndrome (RTT, OMIM #312750) is an X-linked bidities, which complicate patient management and
neurodevelopmental disorder, which mostly affects require a multidisciplinary intervention (Tarquinio
females, although a few cases of RTT males with an extra et al., 2015). Cognitive decline, breathing dysfunction,
X-chromosome or somatic mosaicism have also been epileptic seizures, spasticity, scoliosis, and reduced
described (Moog et al., 2003). Occurring in growth are some of the most frequent symptoms that
1:10,000–15,000 live female births, it accounts for up emerge at the time of developmental regression.
to 10% of severe intellectual disability of genetic origin Recent projections estimate that RTT patients will live
in females (Burd et al., 1991; Armstrong, 1997). In clas- into adulthood (Kirby et al., 2010). It is thus necessary to
sical RTT, an initial period of normal neurologic and provide a complete assessment of the symptoms that
physical development is followed by partial or complete most frequently occur in these patients, in order to choose
loss of acquired purposeful hand skills and speech, the best therapeutic options. Our improved understand-
accompanied by gait abnormalities and stereotypic hand ing of the pathophysiologic underpinnings of RTT has
movements. Several stages are observed clinically: stag- stimulated research into better treatment for comorbid
nation (age 6–18 months), rapid regression (age 1–4 symptoms and the development of new compounds that
years), pseudostationary (age 2–potentially life), and late are potentially able to boost compensatory mechanisms.
motor deterioration (age 10–life). Consequently an RTT We now briefly review pharmacologic interventions
diagnosis is excluded in the presence of grossly abnormal targeting neurologic and psychiatric symptoms associ-
psychomotor development observed during the first six ated with RTT, as well as the main drugs currently
months of life, a history of brain injury secondary to undergoing clinical trials for this syndrome.
trauma, neurometabolic diseases, or severe neurologic
infections (Neul et al., 2010). Other frequent symptoms
The pharmacology of sleep, breathing,
include breathing dysfunction, seizures, spasticity, scoli-
psychiatric, and behavioral disturbances
osis, sleep disturbance, and reduced growth (Chapleau
et al., 2013). Atypical forms include the preserved speech Sleep disturbances are relatively common in RTT, reach-
variant initially described by Zappella et al. (2001), char- ing over 60% of cases (Anderson et al., 2014). Before
acterized by constant intellectual disability, quite often prescribing treatments to regulate circadian rhythmicity,
obesity, and disappearance of hand washing stereotypic clinicians should exclude abdominal pain, gastroesoph-
behaviors after the first few years of life. Rare cases of ageal reflux, and airway obstruction as possible contrib-
isolated hypotonia and developmental delay or utors. RTT patients typically suffer from initial and/or
Angelman-like clinical presentations have been reported intermediate insomnia, i.e., difficulty falling asleep and
(Watson et al., 2001; Heilstedt et al., 2002) frequent awakenings during the night, respectively
RTT is caused mainly by de novo mutations in the (Piazza et al., 1990). Appropriate sleep hygiene is impor-
methyl-CpG-binding protein 2 (MECP2) gene, located tant in RTT patients, but pharmacologic treatments can
on the Xq28 chromosome (Amir et al., 1999). MECP2 be helpful. The most frequently prescribed sleep inducer
plays an important role in neuronal development, den- is melatonin, which has been found to ameliorate also
dritic branching, and brain morphology, because of its total sleep duration and sleep quality in a small 4-week
ability to bind methylated DNA and to orchestrate epige- RCT trial on nine RTT young girls (McArthur and
netic control over transcription by modulating chromatin Budden, 1998). Also L-carnitine may improve sleep
structure in neuronal cells (Lombardi et al., 2015). duration and quality, according to 1 case report of carni-
Recent studies establish that approximately 95% of clas- tine deficiency in a 5-year-old Rett girl (Plochl et al.,
sical RTT cases and 75% of atypical RTT cases have 1996) and 1 open trial involving 21 Rett girls (Ellaway
mutations in MECP2 (Percy et al., 2007; Neul et al., et al., 2001).
2008; Gold et al., 2018). The remaining cases are due Psychiatric symptoms are also emerging as a major
to other genes associated with RTT, including cyclin- concern in RTT, because they are frequent and often
dependent kinase-like 5 (CDKL5), Forkhead box protein difficult to diagnose. These include anxiety, mood distur-
G1 (FOXG1), myocyte-specific enhancer factor 2C bances, and disruptive behaviors. Mood and anxiety dis-
(MEF2C), and transcription factor 4 (TCF4) (Armani orders may be slightly more prevalent in RTT individuals
et al., 2012; Gold et al., 2018). aged 26–30 years, compared to younger and older age
groups (Anderson et al., 2014). According to three case
reports (G€okben et al., 2012; Brook and Usman, 2015;
Therapeutic approaches
Ohno et al., 2016), the most effective drugs used to man-
There are currently no specific pharmacologic therapies age anxiety, mood disturbances, self-aggressiveness,
for RTT patients, only those directed at symptomatic and sleep apneas are SSRI antidepressants, often in
404 A.M. PERSICO ET AL.
association with the 5-HT1A agonist buspirone or similar gallbladder disease (Motil et al., 2012; Freilinger et al.,
compounds (G€ okben et al., 2012; Brook and Usman, 2014). Several nonpharmacologic methods can be used
2015; Ohno et al., 2016). In RTT rodent models, these to aid or prevent these symptoms, but proton pump inhib-
compounds interestingly induce MECP2 gene expres- itors and the H2 receptor blocker ranitidine remain a
sion, regulate CO2 chemosensitivity, and modulate the viable option, provided drug–drug interactions and the
hypothalamic–pituitary–adrenal axis, which is fre- need for vitamin D treatment are carefully considered
quently altered in RTT patients (Hesketh et al., 2005; (Chapleau et al., 2013).
Cassel et al., 2006; Toward et al., 2013). Breathing dis-
orders, including hyperventilation, apnea, breath hold- Adaptive and behavioral training
ing, and air swallowing, are much more frequent while
The regression phase of RTT typically encompasses loss
awake, but may also occur during sleep and deserve a
of previously acquired language skills and of purposeful
trial with the above-mentioned serotoninergic agents,
hand use, increasing difficulties in motor abilities (dys-
while the tricyclic antidepressant desipramine has proven
praxia), and intellectual disability. Hence, behavioral
clinically ineffective in an RCT involving 36 RTT girls
training based on the general principles of operant
(Mancini et al., 2017).
conditioning represents one of the two major areas of
intervention with RTT patients. The second area is repre-
The pharmacology of epilepsy
sented by alternative communication strategies. Similar
As many as 60%–80% of RTT patients develop seizures to patients diagnosed with ASD, during the regression
that evolve into epilepsy, typically at the end of the period RTT patients display deficits in social contact
regression period or soon afterward. RTT is not associ- and experience communication dysfunction. However,
ated with any specific seizure type. In a substantial pro- social deficits and eye contact progressively improve
portion of individuals with RTT, epilepsy is considered in the following years, allowing nonverbal communica-
the major clinical concern, especially in reference to tion with caregivers, provided some training and facilita-
treatment strategies (Glaze et al., 2010; Nissenkorn tion is given. The development of novel augmentative
et al., 2015). AED treatment is currently based on the communication technologies has allowed otherwise non-
general epilepsy literature and on clinical experience, verbal RTT patients to engage with others and to express
since prospective studies are difficult to perform, given themselves (Townend et al., 2016), promoting social
the relatively low prevalence of this rare disease adaptation of RTT young adults into their families and
(Huppke et al., 2007; Krajnc et al., 2011; Pintaudi enjoyment of interpersonal contact (Haas, 1988). Addi-
et al., 2015). AEDs shown to be effective in seizure man- tional cognitive approaches employ nonverbal training
agement and control include valproate (VPA), carbamaz- aimed at teaching basic and complex emotion recogni-
epine (CBZ), and oxcarbamazepine, with additional case tion as part of a cognitive rehabilitation program
reports supporting lamotrigine (LTG), levetiracetam, and (Fabio et al., 2013). Also motor learning supplemented
topiramate (Krajnc, 2015). Many RTT patients present with social, cognitive, and other sensory experiences
serious adverse events and/or suffer from drug-resistant has been proven useful (Rodger et al., 2018).
seizures. VPA may be most effective in younger girls,
with satisfactory seizure control in 40% of the patients Experimental pharmacologic treatments
aged <5 years and in 19% of the girls aged 5–9 years,
Following the discovery of MeCP2 roles in RTT, many
while CBZ may be the most effective AED in patients
research projects aimed at identifying potential therapeu-
15 years or older, followed by LTG, which is prescribed
tic targets in RTT have been conducted. At the moment,
less frequently due to nutritional and behavioral side
the most achievable strategy for treating “classical” RTT
effects (Vignoli et al., 2017). The prevalence of drug-
seems to target events downstream from the MeCP2 def-
resistant epilepsy in RTT patients reaches approximately
icit (Kaufmann et al., 2016). Following this approach, as
30%, as occurs with other newly diagnosed epilepsies,
seen for ASD, ongoing trials are testing omega-3 fatty
and another 20%–40% become refractory to treatment
acids, to reduce oxidative stress (Leoncini et al., 2011);
over time (Krajnc, 2015).
folinic acid to restore methylation capacity (Hagebeuk
et al., 2011); lovastatin (NCT02563860) and drugs like
The pharmacology of gastrointestinal
EPI-743 and triheptanoin to improve metabolic and
dysfunction
mitochondrial function, respectively; glatiramer acetate
Gastrointestinal problems are common and often severe and fingolimod, synaptic regulators, and BDNF-
in RTT patients. They include gastroesophageal reflux, enhancers; dextromethorphan and ketamine (Chapleau
air swallowing with abdominal distention, chronic con- et al., 2013; Gupta et al., 2014), NMDA receptor antag-
stipation, and abdominal pain, occasionally due to onists possibly able to counteract the excess NMDA
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 405
receptor density documented in MeCP2 knockout mice In the past, the clinical benefits of psychotropic drugs
(Blue et al., 2011), and the excess glutamate present in were often discovered accidentally and the mechanism
the CSF of young RTT girls (Hamberger et al., 1992). responsible for these improvements was studied post
Bromocriptine and desipramine have also been studied hoc. At present, psychopharmacology is increasingly
for their effect on monoamine neurotransmitters based on knowledge of the molecular pathophysiology
(Zappella et al., 1990), based on clinical signs of underlying the behavioral disorder, using animal and cel-
impaired dopaminergic activity in RTT (Humphreys lular models. This is especially evident in monogenic
and Barrowman, 2016). Initially among the most prom- syndromic forms of autism, although the greater com-
ising compounds, IGF1 (mecasermin) is a pleiotropic plexity of the human brain at times prevents the success-
growth factor with critical roles in synapse development, ful outcomes observed in animal models to be paralleled
maturation, and maintenance. IGF1 is able in vitro to by similar clinical outcomes in autistic patients. The
counteract the most evident functional outcome of the experimental use of neurons obtained from human
loss of MeCP2 at the neuronal level, namely loss of den- induced pluripotent stem cells (iPSC) represents the cur-
drite ramifications, decreased number of dendritic spines rent frontier of the individualized functional characteri-
and reduced synaptic contacts. One open trial involving zation and in vitro identification of the drugs that
10 Rett girls treated with IGF1 twice a day subcutane- might be most effective in reverting the negative conse-
ously for 20–24 weeks, compared to 10 untreated quences of a genetic mutation or chromosomal anomaly,
patients, examined IGF-1 for efficacy, safety, and toler- analyzed in the context of the whole genome of the
ability (Pini et al., 2016). A statistically significant patient (Kathuria et al., 2017). Targeted psychopharma-
improvement in several measures of Rett severity and cology has now reached the stage of large phase III trials,
social adjustment was obtained, but changes were far less and studies are underway with many promising com-
impressive than those recorded in rodent models. Also, pounds. Meanwhile, the careful use of currently avail-
considering the high costs and the need to administer able psychoactive drugs, though not directly effective
IGF1 subcutaneously, this approach deserves further upon core autistic symptoms, can ameliorate problem
evidence from large RCTs before its efficacy can be behaviors, sleep disturbances, and comorbid disorders,
considered proven. facilitating integration in school and work environments,
enhancing the efficacy of behavioral intervention, and
improving significantly the quality of life of patients
CONCLUSIONS and their families. This, in and of itself, is a clinical target
very much worth pursuing.
With very few exceptions, the current pharmacotherapy
of childhood neuropsychiatric disorders has been
REFERENCES
directly derived from adult psychiatry. This poses major
problems when drugs and dosages prescribed for schizo- Adams JB, Holloway C (2004). Pilot study of a moderate dose
phrenia or bipolar disorder are applied to patients with multivitamin/mineral supplement for children with autistic
ASD, given their greater sensitivity to psychoactive spectrum disorder. J Altern Complement Med 10:
drugs and the lower doses required. Also, adverse events 1033–1039.
and paradoxical effects tend to be more frequent com- Adams JB, Audhya T, McDonough-Means S et al. (2011).
Effect of a vitamin/mineral supplement on children and
pared to neurotypical individuals suffering from other
adults with autism. BMC Pediatr 11: 111.
psychiatric or behavioral disorders. Sensory issues and Albertini G, Polito E, Sara M et al. (2006). Compulsive mas-
motor coordination deficits must also be taken into turbation in infantile autism treated by mirtazapine. Pediatr
account in prescribing psychoactive drugs: liquid forms Neurol 34: 417–418.
may be mandatory in small children with major swallow- Aman MG, Buican B, Arnold LE (2003). Methylphenidate
ing deficits but pills may be easier to administer to ASD treatment in children with borderline IQ and mental retar-
children with strong olfactory hypersensitivity or highly dation: analysis of three aggregated studies. J Child
fixed visuo/olfactory sensory rituals. Finally, the state- Adolesc Psychopharmacol 13: 29–40.
ment “once you have seen one autistic child, you have Aman MG, Hollway JA, McDougle CJ et al. (2008). Cognitive
seen one autistic child” is apt in reference to psychophar- effects of risperidone in children with autism and
macologic therapy. Interindividual variability in drug irritable behavior. J Child Adolesc Psychopharmacol 18:
227–236.
response and tolerability is huge and the psychopharma-
Aman MG, Kasper W, Manos G et al. (2010). Line-item anal-
cology of ASD requires highly individualized psycho- ysis of the Aberrant Behavior Checklist: results from two
pharmacologic tailoring to fit each single patient. No studies of aripiprazole in the treatment of irritability asso-
disorder can be more distant from a therapeutic “copy- ciated with autistic disorder. J Child Adoles
and-paste” approach than ASD. Psychopharmacol 20: 415–422.
406 A.M. PERSICO ET AL.
Aman M, Rettiganti M, Nagaraja HN et al. (2015). Buchsbaum M, Hollander E, Haznedar M et al. (2001). Effect of
Tolerability, safety, and benefits of risperidone in children fluoxetine on regional cerebral metabolism in autistic spec-
and adolescents with autism: 21-month follow-up after trum disorders: a pilot study. Int J Neuropsychopharmacol 4:
8-week placebo-controlled trial. J Child Adolesc 119–125.
Psychopharmacol 25: 482–493. Buie T, Campbell DB, Fuchs 3rd GJ et al. (2010). Evaluation,
Aman MG, Findling RL, Hardan AY et al. (2017). Safety and diagnosis, and treatment of gastrointestinal disorders in
efficacy of memantine in children with autism: random- individuals with ASDs: a consensus report. Pediatrics
ized, placebo-controlled study and open-label extension. 125 (Suppl. 1): S1–18.
J Child Adolesc Psychopharmacol 27: 403–412. Burd L, Randall T, Martsolf JT et al. (1991). Rett syndrome
American Psychiatric Association AY (Ed.), (2013). symptomatology of institutionalized adults with mental
Diagnostic and statistical manual of mental disorders, fifth retardation: comparison of males and females. Am
ed.American Psychiatric Association, Washington, DC. J Ment Retard 95: 596–601.
Amir RE, Van den Veyver IB, Wan M et al. (1999). Rett syn- Camacho A, Espı́n JC, Nuñez N et al. (2012). Levetiracetam-
drome is caused by mutations in X-linked MECP2, encod- induced reversible autistic regression. Pediatr Neurol 47:
ing methyl-CpG-binding protein 2. Nat Genet 23: 185–188. 65–67.
Anagnostou E, Esposito K, Soorya L et al. (2006). Divalproex Campbell M, Fish B, Shapiro T et al. (1971). Imipramine in
versus placebo for the prevention of irritability associated preschool autistic and schizophrenic children. J Autism
with fluoxetine treatment in autism spectrum disorder. Child Schizophr 1: 267–282.
J Clin Psychopharmacol 26: 444–446. Canitano R (2005). Clinical experience with topiramate to
Anderson LT, Campbell M, Adams P et al. (1989). The effects counteract neuroleptic induced weight gain in 10 individ-
of haloperidol on discrimination learning and behavioral uals with autistic spectrum disorders. Brain Dev 27:
symptoms in autistic children. J Autism Dev Disord 19: 228–232.
227–239. Canitano R (2015). Mood stabilizers in children and adoles-
Anderson A, Wong K, Jacoby P et al. (2014). Twenty years of cents with autism spectrum disorders. Clin
surveillance in Rett syndrome: what does this tell us? Neuropharmacol 38: 177–182.
Orphanet J Rare Dis 9: 87. Carminati GG, Deriaz N, Bertschy G (2006). Low-dose venla-
Armani R, Archer H, Clarke A et al. (2012). Transcription fac- faxine in three adolescents and young adults with autistic
tor 4 and myocyte enhancer factor 2C mutations are not disorder improves self-injurious behavior and attention
common causes of Rett syndrome. Am J Med Genet A deficit/hyperactivity disorders (ADHD)-like symptoms.
158A: 713–719. Prog Neuropsychopharmacol Biol Psychiatry 30: 312–315.
Armstrong DD (1997). Review of Rett syndrome. Carminati GG, Gerber F, Darbellay B et al. (2016). Using ven-
J Neuropathol Exp Neurol 56: 843–849. lafaxine to treat behavioral disorders in patients with
Baio J, Wiggins L, Christensen DL et al. (2018). Prevalence of autism spectrum disorder. Prog Neuropsychopharmacol
autism spectrum disorder among children aged 8 years— Biol Psychiatry 65: 85–95.
autism and developmental disabilities monitoring network, Carter CS, DeVries AC, Getz LL (1995). Physiological sub-
11 sites, United States, 2014. MMWR Surveill Summ 67: strates of mammalian monogamy: the prairie vole model.
1–23. Neurosci Biobehav Rev 19: 303–314.
Belsito KM, Law PA, Kirk KS et al. (2001). Lamotrigine ther- Cassel S, Carouge D, Gensburger C et al. (2006). Fluoxetine
apy for autistic disorder: a randomized, double-blind, and cocaine induce the epigenetic factors MeCP2 and
placebo-controlled trial. J Autism Dev Disord 31: 175–181. MBD1 in adult rat brain. Mol Pharmacol 70: 487–492.
Ben Ari Y, Gaiarsa JL, Tyzio R et al. (2007). GABA: a pioneer Chapleau CA, Lane J, Pozzo-Miller L et al. (2013). Evaluation
transmitter that excites immature neurons and generates of current pharmacological treatment options in the
primitive oscillations. Physiol Rev 87: 1215–1284. management of Rett syndrome: from the present to
Berenguer-Forner C, Miranda-Casas A, Pastor-Cerezuela G future therapeutic alternatives. Curr Clin Pharmacol 8:
et al. (2015). Comorbidity of autism spectrum disorder 358–369.
and attention deficit with hyperactivity. A review study. Chen HS, Pellegrini JW, Aggarwal SK et al. (1992). Open
Rev Neurol 60 (Suppl. 1): S37–S43. channel block of N-methyl-D-aspartate (NMDA) responses
Bird PD (2015). The treatment of autism with low dose phenyt- by memantine: therapeutic advantage against NMDA
oin: a case report. J Med Care Rep 9: 8. receptor-mediated neurotoxicity. J Neurosci 12:
Blue ME, Kaufmann WE, Bressler J et al. (2011). Temporal 4427–4436.
and regional alterations in NMDA receptor expression in Chen NC, Bedair HS, McKay B et al. (2001). Clozapine in the
Mecp2-null mice. Anat Rec (Hoboken) 294: 1624–1634. treatment of aggression in an adolescent with autistic dis-
Branford D, Bhaumik S, Naik B (1998). Selective serotonin order. J Clin Psychiatry 62: 479–480.
re-uptake inhibitors for the treatment of perseverative Cheng YS, Tseng PT, Chen YW et al. (2017). Supplementation
and maladaptive behaviours of people with intellectual dis- of omega 3 fatty acids may improve hyperactivity, leth-
ability. J Intellect Disabil Res 42(Pt 4): 301–306. argy, and stereotypy in children with autism spectrum dis-
Brook E, Usman M (2015). SSRIs in Rett syndrome. Aust N Z orders: a meta-analysis of randomized controlled trials.
J Psychiatry 49: 667–668. Neuropsychiatr Dis Treat 13: 2531–2543.
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 407
Chugani DC, Chugani HT, Wiznitzer M et al. (2016). Efficacy Fabio RA, Castelli I, Marchetti A et al. (2013). Training com-
of low-dose buspirone for restricted and repetitive behavior munication abilities in Rett syndrome through reading and
in young children with autism spectrum disorder: a writing. Front Psychol 4: 911.
randomized trial. J Pediatr 170: 45–53. Fankhauser M, Karumanchi V, German M et al. (1992).
Corson AH, Barkenbus JE, Posey DJ et al. (2004). A double-blind, placebo controlled study of the efficacy of
A retrospective analysis of quetiapine in the treatment of transdermal clonidine in autism. J Clin Psychiatry 53: 77–82.
pervasive developmental disorders. J Clin Psychiatry 65 Feit PW (1981). Bumetanide—the way to its chemical
(11): 1531–1536. structure. J Clin Pharmacol 21: 531–536.
Coskun M, Mukaddes NM (2008). Mirtazapine treatment in a Fido A, Al-Saad S (2008). Olanzapine in the treatment of
subject with autistic disorder and fetishism. J Child behavioral problems associated with autism: an open-label
Adolesc Psychopharmacol 18: 206–209. trial in Kuwait. Med Princ Pract 17: 415–418.
Coskun M, Karakoc S, Kircelli F et al. (2009). Effectiveness of Filipek PA, Juranek J, Nguyen MT et al. (2004). Relative car-
mirtazapine in the treatment of inappropriate sexual behav- nitine deficiency in autism. J Autism Dev Disord 34:
iors in individuals with autistic disorder. J Child Adolesc 615–623.
Psychopharmacol 19: 203–206. Findling RL, Maxwell K, Wiznitzer M (1997). An open
Danysz W, Parsons CG (2012). Alzheimer’s disease, beta- clinical trial of risperidone monotherapy in young
amyloid, glutamate, NMDA receptors and memantine— children with autistic disorder. Psychopharmacol Bull 33:
searching for the connections. Br J Pharmacol 167: 155–159.
324–352. Findling RL, McNamara NK, Gracious BL et al. (2004).
Davanzo PA, Belin TR, Widawski MH et al. (1998). Paroxetine Quetiapine in nine youths with autistic disorder. J Child
treatment of aggression and self-injury in persons with men- Adolesc Psychopharmacol 14: 287–294.
tal retardation. Am J Ment Retard 102: 427–437. Findling RL, Mankoski R, Timko K et al. (2014).
De Hert M, Dobbelaere M, Sheridan EM et al. (2011). A randomized controlled trial investigating the safety
Metabolic and endocrine adverse effects of second- and efficacy of aripiprazole in the long-term maintenance
generation antipsychotics in children and adolescents: a treatment of pediatric patients with irritability associated
systematic review of randomized, placebo controlled trials with autistic disorder. J Clin Psychiatry 75: 22–30.
and guidelines for clinical practice. Eur Psychiatry 26: Fombonne E (2005). Epidemiology of autistic disorder and
144–158. other pervasive developmental disorders. J Clin
Dominick K, Wink LK, McDougle CJ et al. (2015). Psychiatry 66 (Suppl. 10): 3–8.
A retrospective naturalistic study of ziprasidone for irrita- Fond G, Boukouaci W, Chevalier G et al. (2015). The "psycho-
bility in youth with autism spectrum disorder. J Child microbiotic”: targeting microbiota in major psychiatric dis-
Adolesc Psychopharmacol 25: 397–401. orders: a systematic review. Pathol Biol 63: 35–42.
Douglas JF, Sanders KB, Benneyworth MH et al. (2013). Brief Freilinger M, B€ ohm M, Lanator I et al. (2014). Prevalence, clin-
report: retrospective case series of oxcarbazepine for irrita- ical investigation, and management of gallbladder disease in
bility/agitation symptoms in autism spectrum disorder. Rett syndrome. Dev Med Child Neurol 56: 756–762.
J Autism Dev Disord 43: 1243–1247. Frye RE, Sequeira JM, Quadros EV et al. (2013). Cerebral
Doyle CA, McDougle CJ (2012). Pharmacologic treatments folate receptor autoantibodies in autism spectrum disorder.
for the behavioral symptoms associated with autism spec- Mol Psychiatry 18: 369–381.
trum disorders across the lifespan. Dialogues Clin Neurosci Gabriele S, Sacco R, Persico AM (2014). Blood serotonin levels
14: 263–279. in autism spectrum disorder: a systematic review and meta-
Duggal HS (2007). Ziprasidone for maladaptive behavior and analysis. Eur Neuropsychopharmacol 24: 919–929.
attention-deficit/hyperactivity disorder symptoms in Gagliano A, Germanò E, Pustorino G et al. (2004).
autistic disorder. J Child Adolesc Psychopharmacol 17: Risperidone treatment of children with autistic disorder:
261–263. effectiveness, tolerability, and pharmacokinetic implica-
Egger JIM, Verhoeven WMA, Groenendijk-Reijenga R et al. tions. J Child Adolesc Psychopharmacol 14: 39–47.
(2017). Phelan-McDermid syndrome due to SHANK3 Garbett K, Ebert PJ, Mitchell A et al. (2008). Immune tran-
mutation in an intellectually disabled adult male: success- scriptome alterations in the temporal cortex of subjects
ful treatment with lithium. BMJ Case Rep 2017. https://doi. with autism. Neurobiol Dis 30: 303–311.
org/10.1136/bcr-2017-220778. Gazis D (1978). Plasma half-lives of vasopressin and oxytocin
Ellaway CJ, Peat J, Williams K et al. (2001). Medium-term analogs after iv injection in rats. Proc Soc Exp Biol Med
open label trial of L-carnitine in Rett syndrome. Brain 158: 663–665.
Dev 23: S85–S89. Gedye A (1991). Trazodone reduced aggressive and self-
Ellingrod VL, Perry PJ (1994). Venlafaxine: a heterocyclic injurious movements in a mentally handicapped male
antidepressant. Am J Hosp Pharm 51: 3033–3046. patient with autism. J Clin Psychopharmacol 11: 275–276.
Erickson CA, Veenstra-Vanderweele JM, Melmed RD et al. Geier DA, Kern JK, Davis G et al. (2011). A prospective
(2014). STX209 (arbaclofen) for autism spectrum disor- double-blind, randomized clinical trial of levocarnitine to
ders: an 8-week open-label study. J Autism Dev Disord treat autism spectrum disorders. Med Sci Monit 17:
44: 958–964. PI15–23.
408 A.M. PERSICO ET AL.
Gevi F, Zolla L, Gabriele S et al. (2016). Urinary metabolo- Haas RH (1988). The history and challenge of Rett syndrome.
mics of young Italian autistic children supports abnormal J Child Neurol 3: S3–S5.
tryptophan and purine metabolism. Mol Autism 7: 47. Habibi N, Dodangi N, Nazeri A (2015). Efficacy and safety of
Ghaleiha A, Asadabadi M, Mohammadi MR et al. (2013). aripiprazole for treatment of irritability in children with
Memantine as adjunctive treatment to risperidone in chil- autistic disorder: an open-label study. Iran J Med Sci 40:
dren with autistic disorder: a randomized, double-blind, 548–549.
placebo-controlled trial. Int J Neuropsychopharmacol 16: Hadjikhani N, Z€ urcher NR, Rogier O et al. (2015). Improving
783–789. emotional face perception in autism with diuretic bumeta-
Ghaleiha A, Alikhani R, Kazemi MR et al. (2016). nide: a proof-of-concept behavioral and functional brain
Minocycline as adjunctive treatment to risperidone in imaging pilot study. Autism 19: 149–157.
children with autistic disorder: a randomized, double-blind Hagebeuk EE, Koelman JH, Duran M et al. (2011). Clinical
placebo-controlled trial. J Child Adolesc Psychopharmacol and electroencephalographic effects of folinic acid treat-
26: 784–791. ment in Rett syndrome patients. J Child Neurol 26:
Ghanizadeh A, Ayoobzadehshirazi A (2015). A randomized 718–723.
double-blind placebo-controlled clinical trial of adjuvant Hamberger A, Gillberg C, Palm A et al. (1992). Elevated CSF
buspirone for irritability in autism. Pediatr Neurol 52: glutamate in Rett syndrome. Neuropediatrics 23: 212–213.
77–81. Handen BL, Johnson CR, Lubetsky M (2000). Efficacy of
Ghanizadeh A, Sahraeizadeh A, Berk M (2014). A head-to- methylphenidate among children with autism and symp-
head comparison of aripiprazole and risperidone for safety toms of attention-deficit hyperactivity disorder. J Autism
and treating autistic disorders, a randomized double blind Dev Disord 30: 245–255.
clinical trial. Child Psychiatry Hum Dev 45: 185–192. Handen BL, Sahl R, Hardan AY (2008). Guanfacine in chil-
Glaze DG, Percy AK, Skinner S et al. (2010). Epilepsy and the dren with autism and/or intellectual disabilities. J Dev
natural history of Rett syndrome. Neurology 74: 909–912. Behav Pediatr 29: 303–308.
Goforth HW, Rao MS (2003). Improvement in behaviour and Handen BL, Aman MG, Arnold LE et al. (2015). Atomoxetine,
attention in an autistic patient treated with ziprasidone. parent training, and their combination in children with
Aust N Z J Psychiatry 37: 775–776. autism spectrum disorder and attention-deficit/
G€
okben S, Ardıç UA, Serdaroğlu G (2012). Use of buspirone hyperactivity disorder. J Am Acad Child Adolesc
and fluoxetine for breathing problems in Rett syndrome. Psychiatry 54: 905–915.
Pediatr Neurol 46: 192–194. Hardan AY, Jou RJ, Handen BL (2005). Retrospective study of
Gold WA, Krishnarajy R, Ellaway C et al. (2018). Rett syn- quetiapine in children and adolescents with pervasive
drome: a genetic update and clinical review focusing on developmental disorders. J Autism Dev Disord 35:
comorbidities. ACS Chem Neurosci 9: 167–176. 387–391.
Golubchik P, Sever J, Weizman A (2011). Low-dose Harfterkamp M, van de Loo-Neus G, Minderaa R et al. (2012).
quetiapine for adolescents with autistic spectrum dis- A randomized double blind study of atomoxetine versus
order and aggressive behavior: open-label trial. Clin placebo for attention-deficit/hyperactivity disorder symp-
Neuropharmacol 34: 216–219. toms in children with autism spectrum disorder. J Am
Golubchik P, Sever J, Weizman A (2013). Reboxetine treat- Acad Child Adolesc Psychiatry 51: 733–741.
ment for autistic spectrum disorder of pediatric patients Harfterkamp M, Buitelaar J, Minderaa R et al. (2013).
with depressive and inattentive/hyperactive symptoms: Long-term treatment with atomoxetine for attention-
an open label trial. Clin Neuropharmacol 36: 37–41. deficit/hyperactivity disorder symptoms in children and
Gordon CT, State RC, Nelson JE et al. (1993). A double-blind adolescents with autism spectrum disorder: an open-label
comparison of clomipramine, desipramine and placebo in extension study. J Child Adolesc Psychopharmacol 23:
the treatment of Autistic Disorder. Arch Gen Psychiatry 194–199.
50: 441–447. Heilstedt HA, Shahbazian MD, Lee B (2002). Infantile hypo-
Gregory SG, Connelly JJ, Towers AJ et al. (2009). Genomic tonia as a presentation of Rett syndrome. Am J Med Genet
and epigenetic evidence for oxytocin receptor deficiency 111: 238–242.
in autism. BMC Med 7: 62. Hellings JA, Weckbaugh M, Nickel EJ et al. (2005). A double-
Gringras P, Nir T, Breddy J et al. (2017). Efficacy and safety of blind, placebo- controlled study of valproate for aggression
pediatric prolonged-release melatonin for insomnia in chil- in youth with pervasive developmental disorders. J Child
dren with autism spectrum disorder. J Am Acad Child Adolesc Psychopharmacol 15: 682–692.
Adolesc Psychiatry 56: 948–957. Hesketh S, Jessop DS, Hogg S et al. (2005). Differential
Grossi E, Melli S, Dunca D et al. (2016). Unexpected improve- actions of acute and chronic citalopram on the rodent
ment in core autism spectrum disorder symptoms after hypothalamic-pituitary-adrenal axis response to acute
long-term treatment with probiotics. SAGE Open Med restraint stress. J Endocrinol 185: 373–382.
Case Rep 4: 2050313X16666231. Hirota T, Veenstra-Vanderweele J, Hollander E et al. (2014).
Gupta SV, Ewen R, Bibat J et al. (2014). Pharmacokinetics and Antiepileptic medications in autism spectrum disorder: a
preliminary assessment of efficacy of dextromethorphan systematic review and meta-analysis. J Autism Dev
for the treatment of Rett syndrome. Ann Neurol 76: S246. Disord 44: 948–957.
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 409
Ho JG, Caldwell RL, McDougle CJ et al. (2012). The effects Ishitobi M, Kosaka H, Takahashi T et al. (2013). Effectiveness
of aripiprazole on electrocardiography in children with and tolerability of switching to aripiprazole from
pervasive developmental disorders. J Child Adolesc risperidone in subjects with autism spectrum disorders: a
Psychopharmacol 22: 277–283. prospective open-label study. Clin Neuropharmacol 36:
Hollander E, Kaplan A, Cartwright C et al. (2000). 151–156.
Venlafaxine in children, adolescents, and young adults Jacob S, Brune CW, Carter CS et al. (2007). Association of the
with autism spectrum disorders: an open retrospective oxytocin receptor gene (OXTR) in Caucasian children and
clinical report. J Child Neurol 15: 132–135. adolescents with autism. Neurosci Lett 417(1): 6–9.
Hollander E, Dolgoff-Kaspar R, Cartwright C et al. (2001). Jahromi L, Kasari C, McCracken J et al. (2009). Positive
An open trial of divalproex sodium in autism spectrum effects of methylphenidate on social communication and
disorders. J Clin Psychiatry 62: 530–534. self-regulation in children with pervasive developmental
Hollander E, Phillips A, Chaplin W et al. (2005). A placebo disorders and hyperactivity. J Autism Dev Disord 39:
controlled crossover trial of liquid fluoxetine on repeti- 395–404.
tive behaviours in childhood and adolescent autism. Jaselskis CA, Cook EH, Fletcher KE et al. (1992). Clonidine
Neuropsychopharmacology 30: 582–589. treatment of hyperactive and impulsive children with
Hollander E, Wasserman S, Swanson EN et al. (2006a). autistic disorder. Clin Psychopharmacol 12: 322–327.
A double-blind placebo-controlled pilot study of olanza- Johnson ZV, Young LJ (2015). Neurobiological mechanisms
pine in childhood/adolescent pervasive developmental dis- of social attachment and pair bonding. Curr. Opin Behav
order. J Child Adolesc Psychopharmacol 16: 541–548. Sci 3: 38–44.
Hollander E, Soorya L, Wasserman S et al. (2006b). Johnson ZV, Walum H, Xiao Y et al. (2016). Oxytocin recep-
Divalproex sodium vs placebo in the treatment of repetitive tors modulate a social salience neural network in male prai-
behaviours in autism spectrum disorder. Int rie voles. Horm Behav 87: 16–24.
J Neuropsychopharmacol 9: 209–213. Kaluzna-Czaplinska J, Blaszczyk S (2012). The level of arabi-
Hollander E, Chaplin W, Soorya L et al. (2010). Divalproex nitol in autistic children after probiotic therapy. Nutrition
sodium vs placebo for the treatment of irritability in chil- 28: 124–126.
dren and adolescents with autism spectrum disorders. Kang DW, Adams JB, Gregory AC et al. (2017). Microbiota
Neuropsychopharmacology 35: 990–998. Transfer Therapy alters gut ecosystem and improves gas-
Hollander E, Soorya L, Chaplin W et al. (2012). A double- trointestinal and autism symptoms: an open-label study.
blind placebo-controlled trial of fluoxetine for repetitive Microbiome 5: 10.
behaviors and global severity in adult autism spectrum dis- Kathuria A, Sala C, Verpelli C et al. (2017). Modelling autistic
orders. Am J Psychiatry 169: 292–299. neurons with induced pluripotent stem cells. Adv Anat
Hsiao EY, McBride SW, Hsien S et al. (2013). Microbiota Embryol Cell Biol 224: 49–64.
modulate behavioral and physiological abnormalities asso- Kaufmann WE, Stallworth JL, Everman DB et al. (2016).
ciated with neurodevelopmental disorders. Cell 155: Neurobiologically-based treatments in Rett syndrome:
1451–1463. opportunities and challenges. Expert Opin Orphan Drugs
Humphreys P, Barrowman N (2016). The incidence and evo- 4: 1043–1055.
lution of parkinsonian rigidity in Rett syndrome: a pilot Keech B, Crowe S, Hocking DR (2017). Intranasal oxytocin,
study. Can J Neurol Sci 43: 567–573. social cognition and neurodevelopmental disorders: a
Huppke P, K€ohler K, Brockmann K et al. (2007). Treatment of meta-analysis. Psychoneuroendocrinology 87: 9–19.
epilepsy in Rett syndrome. Eur J Paediatr Neurol 11: 10–16. Kem DL, Posey DJ, McDougle CJ (2002). Priapism associated
Ichikawa H, Mikami K, Okada T et al. (2017). Aripiprazole in with trazodone in an adolescent with autism. J Am Acad
the treatment of irritability in children and adolescents with Child Adolesc 41: 758.
autism spectrum disorder in Japan: a randomized, double- Kemner C, Willemsen-Swinkels SH, de Jonge M et al. (2002).
blind, placebo-controlled study. Child Psychiatry Hum Open-label study of olanzapine in children with pervasive
Dev 48: 796–806. developmental disorder. J Clin Psychopharmacol 22:
Ichikawa H, Hiratani M, Yasuhara A et al. (2018). An open- 455–460.
label extension long-term study of the safety and efficacy Kempuraj D, Tagen M, Iliopoulou BP et al. (2008). Luteolin
of aripiprazole for irritability in children and adolescents inhibits myelin basic protein-induced human mast cell acti-
with autistic disorder in Japan. Psychiatry Clin Neurosci vation and mast cell dependent stimulation of Jurkat
72: 84–94. T cells. Br J Pharmacol 155: 1076–1084.
Insel TR (2010). The challenge of translation in social neuro- Kent J, Kushner S, Ning X et al. (2013a). Risperidone dosing in
science: a review of oxytocin, vasopressin, and affiliative children and adolescents with autistic disorder: a double-
behavior. Neuron 65: 768–779. blind, placebo-controlled study. J Autism Dev Disord 43:
Ishibashi T, Horisawa T, Tokuda K et al. (2010). 1773–1783.
Pharmacological profile of lurasidone, a novel antipsy- Kent JM, Hough D, Singh J et al. (2013b). An open-label
chotic agent with potent 5-hydroxytryptamine 7 (5-HT7) extension study of the safety and efficacy of risperidone
and 5-HT1A receptor activity. J Pharmacol Exp Ther in children and adolescents with autistic disorder. J Child
334: 171–181. Adolesc Psychopharmacol 23: 676–686.
410 A.M. PERSICO ET AL.
Kerbeshian J, Burd L, Fisher W (1987). Lithium carbonate in adolescents with autism spectrum disorders. Transl
the treatment of two patients with infantile autism and atyp- Psychiatry 7: e1124.
ical bipolar symptomatology. J Clin Psychopharmacol 7: Leoncini S, De Felice C, Signorini C et al. (2011). Oxidative
401–405. stress in Rett syndrome: natural history, genotype, and
Kern JK, Jones AM (2006). Evidence of toxicity, oxidative variants. Redox Rep 16: 145–153.
stress, and neuronal insult in autism. J Toxicol Environ Lipton SA (2006). Paradigm shift in neuroprotection by
Health B Crit Rev 9: 485–499. NMDA receptor blockade: memantine and beyond. Nat
Kim SJ, Shonka S, French WP et al. (2017). Dose response Rev Drug Discov 5: 160–170.
effects of long-acting liquid methylphenidate in children Loebel A, Brams M, Goldman RS et al. (2016). Lurasidone for
with attention deficit/hyperactivity disorder (ADHD) and the treatment of irritability associated with autistic disor-
autism spectrum disorder (ASD): a pilot study. J Autism der. J Autism Dev Disord 46: 1153–1163.
Dev Disord 47: 2307–2313. Lombardi LM, Baker SA, Zoghbi HY (2015). MECP2 disor-
King BH, Hollander E, Sikich L et al. (2009). Lack of efficacy ders: from the clinic to mice and back. J Clin Invest 125:
of citalopram in children with autism spectrum disorders 2914–2923.
and high levels of repetitive behavior. Arch Gen Loomes R, Hull L, Mandy WPL (2017). What is the male-to-
Psychiatry 66: 583–590. female ratio in autism spectrum disorder? A systematic
Kirby RS, Lane JB, Childers J et al. (2010). Longevity in Rett review and meta-analysis. J Am Acad Child Adolesc
syndrome: analysis of the North American Database. Psychiatry 56: 466–474.
J Pediatr 156: 135–138. Luby J, Mrakotsky C, Stalets MM et al. (2006). Risperidone in
Kirsch P, Esslinger C, Chen Q et al. (2005). Oxytocin modu- preschool children with autistic spectrum disorders: an
lates neural circuitry for social cognition and fear in investigation of safety and efficacy. J Child Adolesc
humans. J Neurosci 25: 11489–11493. Psychopharmacol 16: 575–587.
Knafo A, Israel S, Darvasi A et al. (2008). Individual differ- Lucht MJ, Barnow S, Sonnenfeld C et al. (2009). Associations
ences in allocation of funds in the dictator game associated between the oxytocin receptor gene (OXTR) and affect,
with length of the arginine vasopressin 1a receptor RS3 loneliness and intelligence in normal subjects. Prog
promoter region and correlation between RS3 length and Neuropsychopharmacol Biol Psychiatry 33: 860–866.
hippocampal mRNA. Genes Brain Behav 7: 266–275. Lynch BA, Lambeng N, Nocka K et al. (2004). The synaptic
Kornhuber J, Bormann J, Retz W et al. (1989). Memantine dis- vesicle protein SV2A is the binding site for the antiepileptic
places [3H]MK-801 at therapeutic concentrations in post- drug levetiracetam. Proc Natl Acad Sci USA 101:
mortem human frontal cortex. Eur J Pharmacol 166: 9861–9866.
589–590. Malone RP, Cater J, Sheikh RM et al. (2001). Olanzapine ver-
Kowalski JL, Wink LK, Blankenship K et al. (2011). sus haloperidol in children with autistic disorder: an open
Paliperidone palmitate in a child with autistic disorder. pilot study. J Am Acad Child Adolesc Psychiatry 40:
J Child Adolesc Psychopharmacol 21: 491–493. 887–894.
Krajnc N (2015). Management of epilepsy in patients with Rett Malone RP, Maislin G, Choudhury MS et al. (2002).
syndrome: perspectives and considerations. Ther Clin Risk Risperidone treatment in children and adolescents with
Manag 11: 925–932. autism: short- and long-term safety and effectiveness.
Krajnc N, Zupancic N, Orazem J (2011). Epilepsy treatment in J Am Acad Child Adolesc Psychiatry 41: 140–147.
Rett syndrome. J Child Neurol 26: 1429–1433. Malone RP, Delaney MA, Hyman SB et al. (2007).
Kritas SK, Saggini A, Varvara G et al. (2013). Luteolin inhibits Ziprasidone in adolescents with autism: an open-label
mast cell-mediated allergic inflammation. J Biol Regul pilot study. J Child Adolesc Psychopharmacol 17:
Homeost Agents 27: 955–959. 779–790.
Kurtis LB (1966). Clinical study of the response to nortripty- Maloney A, Mick EO, Frazier J (2014). Aripiprazole decreases
line on autistic children. Int J Neuropsychiatry 2: 298–301. irritability in 12 out of 14 youth with autism spectrum
Lamberti M, Siracusano R, Italiano D et al. (2016). Head-to- disorders. J Child Adolesc Psychopharmacol 24: 357–359.
head comparison of aripiprazole and risperidone in the Mancini J, Dubus JC, Jouve E et al. (2017). Effect of desipra-
treatment of ADHD symptoms in children with autistic mine on patients with breathing disorders in RETT
spectrum disorder and ADHD: a pilot, open-label, random- syndrome. Ann Clin Transl Neurol 5: 118–127.
ized controlled study. Paediatr Drugs 18: 319–329. Mankoski R, Stockton G, Manos G et al. (2013). Aripiprazole
Lambrey S, Falissard B, Martin-Barrero M et al. (2010). treatment of irritability associated with autistic disorder
Effectiveness of clozapine for the treatment of aggression and the relationship between prior antipsychotic exposure,
in an adolescent with autistic disorder. J Child Adolesc adverse events, and weight change. J Child Adolesc
Psychopharmacol 20: 79–80. Psychopharmacol 23: 572–576.
Lemonnier E, Degrez C, Phelep M et al. (2012). A randomised Marcus RN, Owen R, Kamen L et al. (2009). A placebo-
controlled trial of bumetanide in the treatment of autism in controlled, fixed-dose study of aripiprazole in children
children. Transl Psychiatry 2: e202. and adolescents with irritability associated with autistic
Lemonnier E, Villeneuve N, Sonie S et al. (2017). Effects of disorder. J Am Acad Child Adolesc Psychiatry 48:
bumetanide on neurobehavioral function in children and 1110–1119.
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 411
Marcus RN, Owen R, Manos G et al. (2011). Aripiprazole in Ming X, Gordon E, Kang N et al. (2008). Use of clonidine in chil-
the treatment of irritability in pediatric patients (aged dren with autism spectrum disorders. Brain Dev 30: 454–460.
6-17 years) with autistic disorder: results from a Minshawi NF, Wink LK, Shaffer R et al. (2016).
52-week, open-label study. J Child Adolesc A randomized, placebo-controlled trial of D-cycloserine
Psychopharmacol 21: 229–236. for the enhancement of social skills training in autism
Marrosu F, Marrosu G, Rachel M et al. (1987). Paradoxical spectrum disorders. Mol Autism 7: 2.
reactions elicited by diazepam in children with classic Miral S, Gencer O, Inal-Emiroglu FN et al. (2008).
autism. Funct Neurol 2: 355–361. Risperidone versus haloperidol in children and adolescents
Martin A, Koenig K, Scahill L et al. (1999). Open-label quetia- with AD: a randomized, controlled, double-blind trial. Eur
pine in the treatment of children and adolescents with autistic Child Adolesc Psychiatry 17: 1–8.
disorder. J Child Adolesc Psychopharmacol 9: 99–107. Moog U, Smeets EEJ, van Roozendaal KEP et al. (2003).
Masi G, Cosenza A, Mucci M et al. (2001). Open trial of ris- Neurodevelopmental disorders in males related to the gene
peridone in 24 young children with pervasive developmen- causing Rett syndrome in females (MECP2). Eur J Paediatr
tal disorders. J Am Acad Child Adolesc Psychiatry 40: Neurol 7: 5–12.
1206–1214. Motil KJ, Caeg E, Barrish JO et al. (2012). Gastrointestinal and
Mazzone L, Ruta L (2006). Topiramate in children with autis- nutritional problems occur frequently throughout life
tic spectrum disorders. Brain Dev 28: 668. in girls and women with Rett syndrome. J Pediatr
McArthur AJ, Budden SS (1998). Sleep dysfunction in Rett Gastroenterol Nutr 55: 292–298.
syndrome: a trial of exogenous melatonin treatment. Dev Mouti A, Reddihough D, Marraffa C et al. (2014). Fluoxetine
Med Child Neurol 40: 186–192. for autistic behaviors (FAB trial): study protocol for a ran-
McCracken JT, McGough J, Shah B et al. (2002). Risperidone domized controlled trial in children and adolescents with
in children with autism and serious behavioral problems. autism. Trials 15: 230.
N Engl J Med 347: 314–321. Nagaraj R, Singhi P, Malhi P (2006). Risperidone in children
McCracken J, Badashova KK, Posey DJ et al. (2014). Positive with autism: randomized, placebo-controlled, double-blind
effects of methylphenidate on hyperactivity are moderated study. J Child Neurol 21: 450–455.
by monoaminergic gene variants in children with autism Neul JL, Fang P, Barrish J et al. (2008). Specific mutations in
spectrum disorders. Pharmacogenomics J 14: 295–302. methyl-CpG-binding protein 2 confer different severity in
McDougle CJ, Naylor S, Cohen D et al. (1996). A double- Rett syndrome. Neurology 70: 1313–1321.
blind, placebo-controlled study of fluvoxamine in adults Neul JL, Kaufmann WE, Glaze DG et al. (2010). Rett syn-
with autistic disorder. Arch Gen Psychiatry 53: 1001–1008. drome: revised diagnostic criteria and nomenclature. Ann
McDougle CJ, Holmes JP, Bronson MR et al. (1997). Neurol 68: 944–950.
Risperidone treatment of children and adolescents with per- Nguyen M, Murphy T (2001). Mirtazapine for excessive mas-
vasive developmental disorders: a prospective open-label turbation in an adolescent with autism. J Am Acad Child
study. J Am Acad Child Adolesc Psychiatry 36: 685–693. Adolesc Psychiatry 40: 868–869.
McDougle CJ, Brodkin ES, Naylor ST et al. (1998). Nicolson R, Awad G, Sloman L (1998). An open trial of risper-
Sertraline in adults with pervasive developmental disor- idone in young autistic children. J Am Acad Child Adolesc
ders: a prospective open-label investigation. J Clin Psychiatry 37: 372–376.
Psychopharmacol 18: 62–66. Niederhofer H, Staffen W, Mair A (2003). Tianeptine: a
McDougle CJ, Kresch LE, Posey DJ (2000). Repetitive novel strategy of psychopharmacological treatment of chil-
thoughts and behavior in pervasive developmental disor- dren with autistic disorder. Hum Psychopharmacol 18:
ders: treatment with serotonin reuptake inhibitors. 389–393.
J Autism Dev Disord 30: 427–435. Nishimura M, Hashimoto T, Miyazaki M et al. (2003). An
McDougle CJ, Kem DL, Posey DJ (2002). Case series: use of open trial of risperidone in autistic children. No To
ziprasidone for maladaptive symptoms in youths with Hattatsu 35: 473–477.
autism. J Am Acad Child Adolesc Psychiatry 41: 921–927. Nissenkorn A, Levy-Drummer RS, Bondi O et al. (2015).
McDougle CJ, Scahill L, Aman MG et al. (2005). Risperidone Epilepsy in Rett syndrome—lessons from the Rett net-
for the core symptom domains of autism: results from the worked database. Epilepsia 56: 569–576.
study by the autism network of the research units on pedi- Ohno K, Saito Y, Ueda R et al. (2016). Effect of serotonin 1A
atric psychopharmacology. Am J Psychiatry 162: agonists and selective serotonin reuptake inhibitors on
1142–1148. behavioral and nighttime respiratory symptoms in Rett
Meyer-Lindenberg A, Domes G, Kirsch P et al. (2011). syndrome. Pediatr Neurol 60: 54–59.
Oxytocin and vasopressin in the human brain: social neu- Ooi YP, Weng SJ, Kossowsky J et al. (2017). Oxytocin
ropeptides for translational medicine. Nat Rev Neurosci and autism spectrum disorders: a systematic review
12: 524–538. and meta-analysis of randomized controlled trials.
Millard PH, McLaren JL, Coffey DB (2014). Lurasidone treat- Pharmacopsychiatry 50: 5–13.
ment in a child with autism spectrum disorder with irrita- Owen R, Sikich L, Marcus RN et al. (2009). Aripiprazole in the
bility and aggression. J Child Adolesc Psychopharmacol treatment of irritability in children and adolescents with
24: 354–356. autistic disorder. Pediatrics 124: 1533–1540.
412 A.M. PERSICO ET AL.
Owley T, Walton L, Salt J et al. (2005). An open-label trial of Posey DJ, Aman MG, McCracken JT et al. (2007). Positive
escitalopram in pervasive developmental disorders. J Am effects of methylphenidate on inattention and hyperactivity
Acad Child Adolesc Psychiatry 44: 343–348. in pervasive developmental disorders: an analysis of sec-
Pandina GJ, Bossie CA, Youssef E et al. (2007). Risperidone ondary measures. Biol Psychiatry 61: 538–544.
improves behavioral symptoms in children with autism in a Potenza MN, Holmes JP, Kanes SJ et al. (1999). Olanzapine
randomized, double-blind, placebo-controlled trial. treatment of children, adolescents, and adults with perva-
J Autism Dev Disord 37: 367–373. sive developmental disorders: an open-label pilot study.
Pardo CA, Buckley A, Thurm A et al. (2013). A pilot open- J Clin Psychopharmacol 19: 37–44.
label trial of minocycline in patients with autism and Quincozes-Santos A, Bobermin LD, Kleinkauf-Rocha J et al.
regressive features. J Neurodev Disord 5: 9. (2009). Atypical neuroleptic risperidone modulates glial func-
Parker RA, Hartman EE (2002). An 8-year-old boy with tions in C6 astroglial cells. Prog Neuropsychopharmacol Biol
autism, 1 year later. JAMA 287: 504. Psychiatry 33: 11–15.
Payne JA, Rivera C, Voipio J et al. (2003). Cation-chloride Quintana H, Birmaher B, Stedge D et al. (1995). Use of meth-
co-transporters in neuronal communication, development ylphenidate in the treatment of children with autistic disor-
and trauma. Trends Neurosci 26: 199–206. der. J Autism Dev Disord 25: 283–294.
Pearson D, Santos C, Aman M et al. (2013). Effects of Rajapakse T, Pringsheim T (2010). Pharmacotherapeutics of
extended release methylphenidate treatment on ratings of Tourette syndrome and stereotypies in autism. Semin
attention-deficit/hyperactivity disorder (ADHD) and asso- Pediatr Neurol 17: 254–260.
ciated behavior in children with autism spectrum disorders Ramaekers VT, Quadros EV, Sequeira JM (2013). Role of
and ADHD symptoms. J Child Adolesc Psychopharmacol folate receptor autoantibodies in infantile autism. Mol
23: 337–351. Psychiatry 18: 270–271.
Percy AK, Lane JB, Childers J et al. (2007). Rett syndrome: Remington G, Sloman L, Konstantareas M et al. (2001).
North American database. J Child Neurol 22: 1338–1341. Clomipramine versus haloperidol in the treatment of autis-
Persico AM, Napolioni V (2013). Autism genetics. Behav tic disorder: a double-blind, placebo-controlled, crossover
Brain Res 251: 95–112. study. J Clin Psychopharmacol 21: 440–444.
Persico AM, Arango C, Buitelaar JK et al. (2015). Unmet Rett A (1966). Uber € ein zerebral-atrophisches Syndrom
needs in paediatric psychopharmacology: present scenario bei Hyperammon€amie. Wien Med Wochenschr 116:
and future perspectives. Eur Neuropsychopharmacol 25: 723–726.
1513–1531. Rezaei V, Mohammadi MR, Ghanizadeh A et al. (2010).
Piazza CC, Fisher W, Kiesewetter K et al. (1990). Aberrant Double-blind, placebo-controlled trial of risperidone
sleep patterns in children with the Rett syndrome. Brain plus topiramate in children with autistic disorder.
Dev 12: 488–493. Prog Neuropsychopharmacol Biol Psychiatry 34:
Pini G, Congiu L, Benincasa A et al. (2016). Illness severity, 1269–1272.
social and cognitive ability, and EEG analysis of ten Robb AS, Andersson C, Bellocchio EE et al. (2011). Safety
patients with Rett syndrome treated with mecasermin and tolerability of aripiprazole in the treatment of irritabil-
(recombinant Human IGF-1). Autism Res Treat 2016: ity associated with autistic disorder in pediatric subjects
5073078. (6-17 years old): results from a pooled analysis of 2 studies.
Pintaudi M, Calevo MG, Vignoli A et al. (2015). Antiepilptic Prim Care Companion CNS Disord 13(1).
drugs in Rett syndrome. Eur J Paediatr Neurol 19: 446–452. Rodger J, Li C, Tan X et al. (2018). Environmental enrichment
Piras IS, Haapanen L, Napolioni V et al. (2014). Anti-brain intervention for Rett syndrome: an individually rando-
antibodies are associated with more severe cognitive and mised stepped wedge trial. Orphanet J Rare Dis 13: 3.
behavioral profiles in Italian children with autism spectrum Rossignol DA, Frye RE (2014a). Melatonin in autism
disorder. Brain Behav Immun 38: 91–99. spectrum disorders. Curr Clin Pharmacol 9: 326–334.
Plochl E, Sperl W, Wermuth B et al. (1996). Carnitine defi- Rossignol DA, Frye RE (2014b). The use of medications
ciency and carnitine therapy in a patient with Rett syn- approved for Alzheimer’s disease in autism spectrum
drome. Klin Padiatr 208: 129–134. disorder: a systematic review. Front Pediatr 2: 87.
Posey DI, Litwiller M, Koburn A et al. (1999). Paroxetine in Rugino TA, Samsock TC (2002). Levetiracetam in autistic
autism. J Am Acad Child Adolesc Psychiatry 38: 111–112. children: an open-label study. J Dev Behav Pediatr 23:
Posey DJ, Guenin KD, Kohn AE et al. (2001). A naturalistic 225–230.
open-label study of mirtazapine in autistic and other perva- RUPPAN: Research Units on Pediatric Psychopharmacology
sive developmental disorders. J Child Adolesc Autism Network (2005a). Risperidone treatment of
Psychopharmacol 11: 267–277. autistic disorder: longer-term benefits and blinded
Posey DJ, Jl P, Sasher TM et al. (2004a). Guanfacine treatment discontinuation after 6 months. Am J Psychiatry 162:
of hyperactivity and inattention in pervasive developmen- 1361–1369.
tal disorder: a retrospective analysis of 80 cases. J Child RUPPAN: Research Units on Pediatric Psychopharmacology
Adolesc Psychopharmacol 14: 233–241. Autism Network (2005b). Randomized, controlled, cross-
Posey DJ, Kem DL, Swiezy NB et al. (2004b). A pilot study of over trial of methylphenidate in pervasive developmental
D-cycloserine in subjects with autistic disorder. Am disorders with hyperactivity. Arch Gen Psychiatry 62:
J Psychiatry 161: 2115–2117. 1266–1274.
PSYCHOPHARMACOLOGY OF ASD AND RETT SYNDROME 413
Sahoo S, Padhy SK, Singla N et al. (2017). Effectiveness of clo- Steingard RJ, Zimnitzky B, DeMaso DR et al. (1997).
zapine for the treatment of psychosis and disruptive behav- Sertraline treatment of transition-associated anxiety and
iour in a child with atypical autism: a case report and a brief agitation in children with autistic disorder. J Child
review of the evidence. Asian J Psychiatr 29: 194–195. Adolesc Psychopharmacol 7: 9–15.
Sanchez LE, Campbell M, Small AM et al. (1996). A pilot Stigler KA, Diener JT, Kohn AE et al. (2009). Aripiprazole
study of clomipramine in young autistic children. J Am in pervasive developmental disorder not otherwise speci-
Acad Child Adolesc Psychiatry 35: 537–544. fied and Asperger’s disorder: a 14-week, prospective,
Scahill L, Aman MG, McDougle CJ et al. (2006). open-label study. J Child Adolesc Psychopharmacol 19:
A prospective open trial of guanfacine in children with per- 265–274.
vasive developmental disorder. J Child Adolesc Stigler KA, Erickson CA, Mullett JE et al. (2010).
Psychopharmacol 16: 589–598. Paliperidone for irritability in autistic disorder. J Child
Scahill L, McCracker JT, King BH, et al.RUPPAN (2015). Adolesc Psychopharmacol 20(1): 75–78.
Extended-release guanfacine for hyperactivity in the chil- Stigler KA, Mullett JE, Erickson CA et al. (2012).
dren with autism spectrum disorders. Am J Psychiatry Paliperidone for irritability in adolescents and young adults
172: 1197–1206. with autistic disorder. Psychopharmacology (Berl) 223:
Serret S, Thummler S, Dor E et al. (2015). Lithium as a rescue 237–245.
therapy for regression and catatonia in two SHANK3 Sugie Y, Sugie H, Kukuda T et al. (2005). Clinical efficacy of
patients with autism spectrum disorder: case reports. fluvoxamine and functional polymorphism in a serotonin
BMC Psychiatry 15: 107. transporter gene on childhood autism. J Autism Dev
Shaaban SY, El Gendy YG, Mehanna NS et al. (2017). The Disord 35: 377–385.
role of probiotics in children with autism spectrum disor- Taliou A, Zintzaras E, Lykouras L et al. (2013). An open-label
der: a prospective, open-label study. Nutr Neurosci 7: 1–6. pilot study of a formulation containing the anti-
Shapiro DA, Renock S, Arrington E et al. (2003). Aripiprazole, inflammatory flavonoid luteolin and its effects on behavior
a novel atypical antipsychotic drug with a unique and in children with autism spectrum disorders. Clin Ther 35:
robust pharmacology. Neuropsychopharmacology 28: 592–602.
1400–1411. Tarquinio DC, Hou W, Neul JL et al. (2015). The changing
Shea S, Turgay A, Carroll A et al. (2004). Risperidone in the face of survival in Rett syndrome and MECP2-related dis-
treatment of disruptive behavioral symptoms in children orders. Pediatr Neurol 53: 402–411.
with autistic and other pervasive developmental disorders. Theoharides TC, Asadi S, Panagiotidou S (2012). A case
Pediatrics 114: e634–e641. series of a luteolin formulation (NeuroProtek®) in children
Siegel M, Beresford CA, Bunker M et al. (2014). Preliminary with autism spectrum disorders. Int J Immunopathol
investigation of lithium for mood disorder symptoms in Pharmacol 25: 317–323.
children and adolescents with autism spectrum disorder. Tomova A, Husarova V, Lakatosova S et al. (2015).
J Child Adolesc Psychopharmacol 24: 399–402. Gastrointestinal microbiota in children with autism in
Silverman JL, Pride MC, Hayes JE et al. (2015). GABAB Slovakia. Physiol Behav 138: 179–187.
receptor agonist R-baclofen reverses social deficits and Tonge BJ, Bull K, Brereton A et al. (2014). A review of
reduces repetitive behavior in two mouse models of autism. evidence-based early intervention for behavioural prob-
Neuropsychopharmacology 40: 2228–2239. lems in children with autism spectrum disorder: the core
Simonoff E, Pickles A, Charman T et al. (2008). Psychiatric components of effective programs, child-focused interven-
disorders in children with autism spectrum disorders: prev- tions and comprehensive treatment models. Curr Opin
alence, comorbidity, and associated factors in a population- Psychiatry 27: 158–165.
derived sample. J Am Acad Child Adolesc Psychiatry 47: Toward MA, Abdala AP, Knopp SJ et al. (2013). Increasing
921–929. brain serotonin corrects CO2 chemosensitivity in methyl-
Simonoff E, Taylor E, Baird G et al. (2013). Randomized con- CpG-binding protein 2 (Mecp2)-deficient mice. Exp
trolled double-blind trial of optimal dose methylphenidate Physiol 98: 842–849.
in children and adolescents with severe attention deficit Townend GS, Marschik PB, Smeets E et al. (2016). Eye gaze
hyperactivity disorder and intellectual disability. J Child technology as a form of augmentative and alternative com-
Psychol Psychiatry 54: 527–535. munication for individuals with Rett syndrome: experi-
Singh K, Connors SL, Macklin EA et al. (2014). Sulforaphane ences of families in the Netherlands. J Dev Phys Disabil
treatment of autism spectrum disorder (ASD). Proc Natl 28: 101–112.
Acad Sci USA 111: 15550–15555. Troost PW, Lahuis BE, Steenhuis MP et al. (2005).
Smith T, Aman MG, Arnold LE et al. (2016). Atomoxetine and Long-term effects of risperidone in children with au-
parent training for children with autism and attention- tism spectrum disorders: a placebo discontinuation
deficit/hyperactivity disorder: a 24-week extension study. study. J Am Acad Child Adolesc Psychiatry 44:
J Am Acad Child Adolesc Psychiatry 55: 868–876. 1137–1144.
Steingard R, Biederman J (1987). Lithium responsive manic- Tsilioni I, Taliou A, Francis K et al. (2015). Children with
like symptoms in two individuals with autism and mental autism spectrum disorders, who improved with a
retardation. J Am Acad Child Adolesc Psychiatry 26: luteolin-containing dietary formulation, show reduced
932–935. serum levels of TNF and IL-6. Transl Psychiatry 5e647.
414 A.M. PERSICO ET AL.
Tumuluru RV, Corbett-Dick P, Aman MG et al. (2017). Watson P, Black G, Ramsden S et al. (2001). Angelman syn-
Adverse events of atomoxetine in a double-blind drome phenotype associated with mutations in MECP2, a
placebo-controlled study in children with autism. J Child gene encoding a methyl CpG binding protein. J Med
Adolesc Psychopharmacol 27: 708–714. Genet 38: 224–228.
Urbano M, Okwara L, Manser P et al. (2014). A trial of Werling DM (2016). The role of sex-differential biology in
D-cycloserine to treat stereotypies in older adolescents risk for autism spectrum disorder. Biol Sex Differ 7: 58.
and young adults with autism spectrum disorder. Clin Wigton R, Radua J, Allen P et al. (2015). Neurophysiological
Neuropharmacol 37: 69–72. effects of acute oxytocin administration: systematic review
Urbano M, Okwara L, Manser P et al. (2015). A trial of and meta-analysis of placebo-controlled imaging studies.
d-cycloserine to treat the social deficit in older adolescents J Psychiatry Neurosci 40: E1–22.
and young adults with autism spectrum disorders. Williams K, Brignell A, Randall M et al. (2013). Selective
J Neuropsychiatry Clin Neurosci 27: 133–138. serotonin reuptake inhibitors (SSRIs) for autism
Vargas DL, Nascimbene C, Krishnan C et al. (2005). spectrum disorders (ASD). Cochrane Database Syst Rev
Neuroglial activation and neuroinflammation in the brain 8: CD004677.
of patients with autism. Ann Neurol 57: 67–81. Wink LK, Minshawi NF, Shaffer RC et al. (2017).
Veenstra-VanderWeele J, Cook EH, King BH et al. (2017). D-Cycloserine enhances durability of social skills training
Arbaclofen in children and adolescents with autism spec- in autism spectrum disorder. Mol Autism 8: 2.
trum disorder: a randomized, controlled, phase 2 trial. Wong C, Odom SL, Hume KA et al. (2015). Evidence-based
Neuropsychopharmacology 42: 1390–1398. practices for children, youth, and young adults with autism
Vignoli A, Savini MN, Nowbut MS et al. (2017). Effectiveness spectrum disorder: a comprehensive review. J Autism Dev
and tolerability of antiepileptic drugs in 104 girls with Rett Disord 45: 1951–1966.
syndrome. Epilepsy Behav 66: 27–33. Yamasue H, Kuwabara H, Kawakubo Y et al. (2009).
Vogl C, Mochida S, Wolff C et al. (2012). The synaptic vesicle Oxytocin, sexually dimorphic features of the social brain,
glycoprotein 2A ligand levetiracetam inhibits presynaptic and autism. Psychiatry Clin Neurosci 63: 129–140.
Ca2 + channels through an intracellular pathway. Mol Zappella M, Genazzani A, Facchinetti F et al. (1990).
Pharmacol 82: 199–208. Bromocriptine in the Rett syndrome. Brain Dev 12:
Wang M, Jiang L, Tang X (2017). Levetiracetam is associated 221–225.
with decrease in subclinical epileptiform discharges and Zappella M, Meloni I, Longo I et al. (2001). Preserved speech
improved cognitive functions in pediatric patients with variants of the Rett syndrome: molecular and clinical
autism spectrum disorder. Neuropsychiatr Dis Treat 13: analysis. Am J Med Genet 104: 14–22.
2321–2326. Zhang R, Zhang HF, Han JS et al. (2017). Genes related to
Wasserman S, Iyengar R, Chapllin WF et al. (2006). oxytocin and arginine-vasopressin pathways: associations
Levetiracetam versus placebo in childhood and adolescent with autism spectrum disorders. Neurosci Bull 33: 238–246.
autism: a double-blind placebo-controlled study. Int Clin Zuddas A, Ledda MG, Fratta A et al. (1996). Clinical effects of
Psychopharmacol 21: 363–367. clozapine on autistic disorder. Am J Psychiatry 153: 738.

You might also like