You are on page 1of 20

PFI_12mmX178mm.

pdf + eps format

Journal of Child Psychology and Psychiatry **:* (2017), pp **–** doi:10.1111/jcpp.12812

Practitioner Review: Treatment of chronic insomnia in


children and adolescents with neurodevelopmental
disabilities
Oliviero Bruni,1 Marco Angriman,2 Fabrizio Calisti,3 Alessandro Comandini,3
Giovanna Esposito,3 Samuele Cortese,4,5,6 and Raffaele Ferri7
1
Department of Developmental and Social Psychology, Sapienza University, Rome; 2Department of Pediatrics, Child
Neurology and Neurorehabilitation Unit, Central Hospital of Bolzano, Bolzano; 3Angelini, Research Center,
S.Palomba, Rome, Italy; 4Academic Unit of Psychology, Developmental Brain-Behavior Laboratory, University of
Southampton, Southampton, UK; 5New York University Child Study Center, New York, NY, USA; 6Solent NHS Trust,
Southampton, UK; 7Sleep Research Centre; Department of Neurology I.C., Oasi Institute for Research on Mental
Retardation and Brain Aging (IRCCS), Troina, Italy

Background: Sleep disturbances, in particular insomnia, represent a common problem in children with neurode-
velopmental disabilities (NDDs). Currently, there are no approved medications for insomnia in children by the US
Food and Drug Administration or European Medicines Agency and therefore they are prescribed off-label. We
critically reviewed pediatric literature on drugs as well as nonpharmacological (behavioral) interventions used for
sleep disturbances in children with NDDs. Methods: PubMed, Ovid (including PsycINFO, Ovid MEDLINEâ, and
Embase), and Web of Knowledge databases were searched through February 12, 2017, with no language restrictions.
Two authors independently and blindly performed the screening. Results: Good sleep practices and behavioral
interventions, supported by moderate-to-low level evidence, are the first recommended treatments for pediatric
insomnia but they are often challenging to implement. Antihistamine agents, such as hydroxyzine or diphenhy-
dramine, are the most widely prescribed sedatives in the pediatric practice but evidence supporting their use is still
limited. An increasing body of evidence supports melatonin as the safest choice for children with NDDs.
Benzodiazepines are not recommended in children and should only be used for transient insomnia, especially if
daytime anxiety is present. Only few studies have been carried out in children’s and adolescents’ zolpidem, zaleplon,
and eszopiclone, with contrasting results. Limited evidence supports the use of alpha-agonists such as clonidine to
improve sleep onset latency, especially in attention deficit/hyperactivity disorder subjects. Tricyclic antidepressants,
used in adults with insomnia, are not recommended in children because of their safety profile. Trazodone and
mirtazapine hold promise but require further studies. Conclusions: Here, we provided a tentative guide for the use of
drugs for insomnia in children with NDDs. Well-controlled studies employing both objective polysomnography and
subjective sleep measures are needed to determine the efficacy, effectiveness, and safety of the currently prescribed
pediatric sleep medicines in children with NDDs. Keywords: Sleep disorders; insomnia; neurodevelopmental
disorders; drug effects.

epilepsy or gastroesophageal reflux), poor sleep


Introduction
hygiene, or behavioral insomnia of childhood that
Sleep disturbances in children with neurodevelop-
can be difficult to identify in children with NDDs, often
mental disabilities (NDDs), in particular difficulty in
exacerbated by their reduced communication skills.
falling asleep, night awakenings, and reduced sleep
Providers may have received insufficient training, or
duration, are among the most common parental
not have the time to implement behavioral interven-
complaints to health care professionals, with a
tions for sleep. Parent-based education and behavioral
reported prevalence up to 86% (Robinson-Shelton
interventions are the first line of treatment for insom-
& Malow, 2016). Sleep disturbances impact on
nia, unless symptom severity needs early pharma-
cognitive and emotional development, aggravating
cotherapy (Malow et al., 2016).
the functional impairment associated with NDDs
In some patients, problematic sleep is a pheno-
(Van de Wouw, Evenhuis, & Echteld, 2012). Sleep
typic characteristic of a particular disorder or genetic
disturbances also affect the entire family environ-
condition and the knowledge of the distinctive
ment, disrupting the siblings and marital relation-
features of sleep disorders in patients with NDDs is
ships and increasing the levels of stress (Goldman,
crucial for their effective treatment (Grigg-Damber-
Bichell, Surdyka, & Malow, 2012).
ger & Ralls, 2013). In other cases, sleep disorders
The causes of sleep problems in children with NDDs
represent a main comorbidity. For instance, a meta-
are often complex and multifactorial: sleep difficulties
analysis (Cortese, Faraone, Konofal, & Lecendreux,
may be related to co-occurring medical conditions (e.g.
2009) on sleep parameters found that in children
with attention deficit/hyperactivity disorder (ADHD),
scores of reported bedtime resistance, sleep onset
Conflict of interest statement: No conflicts declared.

© 2017 Association for Child and Adolescent Mental Health.


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

difficulties, night awakenings, difficulties with morn- and blindly performed the search and screening of papers
ing awakening, sleep breathing problems, and day- against eligibility criteria. Any disagreement between the two
authors was resolved by consensus. Figure 1 shows the
time sleepiness were significantly higher than in
Preferred Reporting Items for Systematic Reviews and Meta-
healthy controls. Moreover, sleep disorders might be Analyses flow diagram reporting the screening process.
aggravated by common issues linked to NDDs (such
as sensory and motor deficits, psychopathological
disturbances, respiratory disorders, epilepsy, and
Treatment of chronic insomnia in
mental retardation), all contributing to the develop-
neurodevelopmental disabilities
mental delay.
Current practice worldwide
Although for some NDDs specific sleep distur-
bances have been reported, (e.g. sleep apnea in Down In children and adolescents with NDDs, behavioral
or Prader–Willi syndromes), the most commonly techniques may be difficult to implement and may
described sleep complaints in children with NDDs take weeks or months to show benefits. Therefore,
are mainly represented by difficulty in settling at night sleep-promoting agents should be recommended as
(present in up to 51% of the patients) and nocturnal an option while continuing behavioral intervention.
awakenings (up to 67%; Quine, 1991). Fragmented Furthermore, children who do not respond to behav-
sleep throughout the day and night contributes to ioral interventions may be candidate for the pharma-
daytime sleepiness and irregular sleep schedule, cological treatment of insomnia (Angriman, Caravale,
which may lead to a non-24-hr sleep–wake rhythm Novelli, Ferri, & Bruni, 2015; Mindell, Kuhn, Lewin,
with a period longer than 24 hr or to a complete Meltzer, & Sadeh, 2006; Pelayo & Yuen, 2012).
reversal of the night–day cycle (Okawa, 1987). Due to the paucity of controlled studies, there are
Although behavioral interventions should be con- no sleep medications approved by the FDA or the
sidered the first-line option to address sleep distur- EMA for pediatric insomnia in typically developing
bances in children with ADHD, in a sizable portion of children, as well as in those with NDDs.
patients they need to be complemented by a phar- Drugs could be initially useful for relief of parents
macological treatment. Currently, there are no med- and children and in general it is better not to wait a long
ications licensed by the US Food and Drug time to treat insomnia, implementing an immediate
Administration (FDA) or by the European Medicines brief drug trial rather than acting later on a chronic
Agency (EMA) to treat insomnia in pediatric patients insomnia that has already lasted for several months or
including those with NDDs, and therefore all avail- years (Pelayo & Dubik, 2008). Especially in infants and
able drugs are prescribed off-label. children, chronic insomnia can lead to maternal
Here, based on a systematic search in a broad set depression, family dysfunction, and impaired social
of databases, we critically discuss available evidence functioning, but it can also affect the child physical
for the treatment of chronic insomnia in children and and mental health. Due to these serious implications
adolescents with NDDs. Although the review is for both individuals and society, it is essential to treat
focused mainly on the pharmacological treatments, insomnia either through education of parents, teach-
we include an initial section on nonpharmacological ers, and other caregivers but also with an effective drug
approaches to highlight their relevance as first-line treatment, when insomnia leads to a disruption of
option in the management of sleep disturbances in family life, or of the child and parent health.
children with NDDs. A recent Australian survey on pharmacological man-
agement of insomnia in children reported that the most
commonly prescribed medications for poor sleep initi-
Methods ation were melatonin (89.1%), clonidine (48%), and
Search strategy antihistamines (29%). Most pediatricians (82%)
reported also using behavioral strategies for sleep
To ensure a high level of methodological adequacy, as recom-
disturbances, most commonly anxiety relaxation tech-
mended by the Cochrane group (Higgins & Green, 2011), and
avoid the inevitable bias caused by dependence on investiga- niques (75%) for poor sleep initiation and graduated
tors agreeing to provide data from unpublished studies, we did extinction (i.e. “controlled crying”, 52%) for disrupted
not search for unpublished data. We excluded nonpeer- overnight sleep. They prescribed melatonin especially
reviewed references (e.g. conference proceedings) as we con- for children with autism (85.2%), developmental delay
sidered the peer-review process as essential to the quality of
(76.2%), ADHD (54.5%), behavioral disorders (42.6%),
the publication. We retained all types of study designs. We
searched the following electronic databases: PubMed, Ovid visual impairment (40.6%), and anxiety disorders
(including PsycINFO, Ovid MEDLINEâ, and Embase), and Web (25.7%). However, over half of the pediatricians
of Knowledge (Web of Science, Biological abstracts, BIOSIS, (54.5%) prescribed melatonin for typically developing
and FSTA). The specific search terms and syntax for each children as well (Heussler et al., 2013).
database are reported in the Supplemental Material. The
A survey by Owens et al. (Owens, Rosen, Mindell, &
search was finalized on February 12, 2017. No language
limitations were applied to avoid biases due to language Kirchner, 2010) showed that more than one third of
restrictions. The reference lists of pertinent reviews/systematic 1,273 child psychiatrists in the United States treated
reviews were screened to reduce the likelihood of missing any insomnia with medication at least half of the times in
relevant publication. Two authors (MA and SC) independently patients with ADHD, autism spectrum disorder

© 2017 Association for Child and Adolescent Mental Health.


Treatment of insomnia in NDD 3

Identification
Records identified throug Additional records identified
database searching through other sources
(n = 2709 ) (n = 0)

Records after duplicates removed


(n = 1519 )
Screening

Records screened Records excluded


(n = 1519 ) (n =1382)

Full-text articles assessed Full-text articles excluded,


Eligibility

for eligibility with reasons


(n = 137) (n =15)

Studies included in
qualitative synthesis
(n =112)
Included

Studies included in
quantitative synthesis
(meta-analysis)
not applicable

Figure 1 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMAa) flow diagram on treatment of chronic insomnia
in children and adolescents with neurodevelopmental disabilities

(ASD), and with mental retardation/developmental in a group of 257 school-aged Australian children
disabilities (MR/DD). Moreover, they reported to treat with ADHD and 57 of them (22%) were taking sleep
17% of preschoolers and at least one-quarter of medications (melatonin 14% and clonidine 9%). Sleep
school-aged and adolescent patients. Overall, 96% medication use was associated with combined-type
of psychiatrists recommended at least one medication ADHD and ADHD medication use. The presence of co-
in a typical month, and 88% recommended an over- occurring internalizing and externalizing comorbidi-
the-counter (OTC) medication. In general, psychia- ties was also associated with sleep medication use in
trists were more likely to use herbal preparations in ad-hoc analyses. A recent US study in ASD showed
children with anxiety or mood disorders than in that medications for sleep were prescribed in 46% of
children with NDDs or ADHD; melatonin was recom- 4- to 10-year-old children with autism and sleep
mended by more than one third of them (39%) difficulties. The most common medication used for
although it is unclear whether it was being used sleep was melatonin, followed by a-agonists, and a
primarily at bedtime for its mild hypnotic effects or as variety of other medications taken to improve sleep
a chronobiotic. For sleep disorders (mainly insomnia), (anticonvulsants, antidepressants, atypical antipsy-
physicians recommended nonprescription antihis- chotics, and benzodiazepines). Children taking med-
tamines in 69% of cases and a-agonists in 67% of ications for sleep had worse daytime behavior and
children with MR/DD. Trazodone was the second pediatric quality of life than children not taking sleep
most frequently prescribed medication for children medications (Malow et al., 2016).
with MR/DD (66%), while sedating antidepressants Another recent Canadian research (Bock, Roach-
were used in 75.5% of MR cases. Atypical antipsy- Fox, Seabrook, Rieder, & Matsui, 2016) found that
chotics were used by more than half of the psychiatrists the most common circumstances and sleep prob-
in children with MR/DD (52%) and benzodiazepines lems for which OTC or prescribed medications were
were used in 21.6% of cases of MR/DD. Tricyclic recommended were mood disorders, developmental
antidepressants were also used for children in these delay, and ADHD (56%, 40%, and 39%, respectively),
diagnostic groups (25.5%). and insomnia, bedtime struggles/delayed sleep
Although the above-mentioned data come mainly onset, and circadian rhythm disorders (52%, 48%,
from the United States, similar results have been and 28%, respectively). A total of 30% OTC or pre-
replicated also in other countries (Bruni et al., 2004; scribed medications were recommended to otherwise
Heussler et al., 2013). For example, Efron, Lycett, & healthy children with sleep problems. Melatonin
Sciberras (2014) described sleep medication use (73%), OTC antihistamines (41%), antidepressants

© 2017 Association for Child and Adolescent Mental Health.


4 Oliviero Bruni et al.

(37%), and benzodiazepines (29%) were the most Clearly, the involvement of parents as agents for
commonly recommended prescribed medications, changing problematic sleep behaviors is fundamen-
respectively. tal; cognitive-behavioral techniques involving par-
ents have been shown to be effective in improving
significantly quality of life and well-being of patients
Nonpharmacological treatment
and caregivers (Grigg-Damberger & Ralls, 2013; Jan
Prevention is the best treatment for insomnia of et al., 2008). Training group of parents in behavioral
childhood but, unfortunately, most parents are approaches to manage sleep problems represents a
referred when the disorder has become chronic. novel behavioral approach: Stuttard et al. (Stuttard,
Although we will focus on drug treatment, we should Beresford, Clarke, Beecham, & Curtis, 2015)
emphasize that when there is a decision to start a reported the findings from a preliminary evaluation
pharmacologic intervention, behavioral interven- of a group-delivered intervention routinely delivered
tions should be always associated (Jan et al., by a Child and Adolescent Mental Health Service
2008); good sleep practices and behavioral interven- Learning Disability team in England: parents
tions are the first recommended treatments for (n = 23) of children with intellectual disabilities were
pediatric insomnia in either healthy or NDDs chil- recruited and the follow-up was of 6 months. No
dren (Honaker & Meltzer, 2014). parent dropped out and statistically significant
The first line of treatment is the promotion of better improvements in night wakings, parent-set goals,
sleep habits that need to be modified and adapted to and parents’ sense of efficacy were observed.
these children. This may be somewhat challenging to As for the treatment of sleep disturbances in
implement, and should be associated with other specific syndromes, Allen, Kuhn, DeHaai, & Wallace
behavioral interventions using a gradual approach (2013) evaluated the effectiveness of a behavioral
(such as gradual withdrawal, gradual extinction, and treatment package to reduce chronic sleep problems
fading), rather than an abrupt change (e.g. extinction in five children (2–11 years of age) with Angelman
techniques), that may be easier and more acceptable syndrome. The treatment package targeted sleep
for parents as well as being more appropriate for environment, sleep–wake schedule, and parent–
some children with special needs. The choice of a child interactions during sleep times. Changes in
specific form of behavioral intervention should be disruptive bedtime behaviors and in sleep onset were
guided by the parent preferences (Wiggs & France, found to be statistically significant.
2000) and, like in typically developing children, there Table 1 summarizes the key principles of non-
is no evidence that one approach is more effective pharmacological approaches to insomnia in children
than another (Angriman et al., 2015). with NDDs.
A systematic review of behavioral treatments for
insomnia in youth concluded that low-to-moderate
level of evidence supports behavioral interventions in Table 1 Behavioral strategies for insomnia in children with
adolescents and in children with NDDs (Meltzer & neurodevelopmental disabilities
Mindell, 2014). This review found only two studies
Adjust sleeping environment
involving behavioral interventions for sleep problems Dark, quiet, nonstimulating
for children with special needs that met the inclusion Turn off electronic devices
criteria. One study included children with ASDs Develop a constant bedtime routine
(Adkins et al., 2012), whereas the other focused on Promote self-soothing skills that allow the child to manage
children with Down syndrome (Stores & Stores, nocturnal awakenings
Put to bed and get them up at the same time every day
2004). An older study showed that bedtime fading In case of difficulties falling asleep:
was effective in advancing bedtime hour in three Avoid naps 4 hr before bedtime
patients with Rett’s syndrome. This treatment pro- Apply bedtime fading (delay bedtime closer to the child target
moted more regular sleep patterns by increasing bedtime of about 30 min; then move bedtime earlier)
appropriate nighttime sleep duration, reducing inap- Favor light exposure when the child gets up and reduce
photic stimulation at evening, to reinforce circadian
propriate daytime sleep and reducing problematic alignment
nighttime behaviors (e.g. nocturnal awakenings; Apply graduated extinction for the child disruptive bedtime
Piazza, Fisher, & Moser, 1991). There were no behaviors
significant effects for any of the four sleep outcome In case of frequent nocturnal awakenings:
measures (Meltzer & Mindell, 2014) but sleep Extinction with parental presence: The parent remains in the
room during extinction, acting as a reassurance for the
hygiene education was associated with improvement child but providing little interaction
in daytime behaviors, pediatric quality of life, and Graduate extinction: ignore negative behaviors (i.e. crying) for
sense of competence in parents. a given amount of time before checking on the child. The
Further studies are needed to identify factors that parent gradually increases the amount of time between
may predict treatment success and to tailor behav- crying and parental response. Parents provide reassurance
through their presence for short durations and with
ioral interventions for young children based on the minimal interaction
child (e.g. temperament, age), parental, and environ-
mental factors and on the underlying disease. (adapted from Grigg-Damberger & Ralls, 2013)

© 2017 Association for Child and Adolescent Mental Health.


Treatment of insomnia in NDD 5

A novel nonpharmacological approach relies on bed The following sections summarize the evidence
materials and accessories. Gringras et al. (2014) base for the drugs commonly used in chronic
assessed the effectiveness of a weighted blanket insomnia of children with NDDs.
intervention in treating severe sleep problems in
children with ASD with a randomized, placebo-
Melatonin. Melatonin (N-acetyl-5-methoxytrypta-
controlled crossover design. The use of a weighted
mine) is a chronobiotic and hypnotic agent involved
blanket was not superior, compared to the control
in the regulation of the sleep–wake cycle. In older
condition, to increase sleep duration or to reduce sleep
children and adults, its production and secretion
onset delay and night awakenings. More recently,
begin in the evening and peak during the night
Frazier et al. (2017) in a preliminary, randomized
between 2:00 and 4:00 AM and are inhibited by light
study investigated the efficacy and tolerability of a
(Kennaway, 2000).
novel mattress technology – the Sound-To-Sleep
A nationwide study showed that hypnotic drug use in
system – in the treatment of sleep problems in 45
0- to 17-year-old patients increased during the period
children with autism, aged 2.5–12.9 years. Satisfac-
2004–2011, from 8.9 to 12.3/1,000, mainly owing to
tory compliance to this technology was observed and
the doubling of melatonin use that was dispensed in the
parent-diary outcomes indicated improvements in
highest annual amount among all hypnotic drugs
falling asleep and reduced daytime challenging behav-
(Hartz, Furu, Bratlid, Handal, & Skurtveit, 2012).
ior. Actigraphy-derived sleep parameters indicated
Approximately 56% of pediatricians prescribed mela-
significant improvement of sleep duration and sleep
tonin for sleep onset insomnia (89.1%), delayed sleep
efficiency.
phase (66.3%), and nighttime awakenings (30.7%).
More specifically, it was prescribed in children with
Pharmacological treatment autism (85.2%), NDDs (76.2%), ADHD (54.5%), behav-
ioral disorders (42.6%), visual impairment (40.6%),
General considerations on prescribing medications
anxiety disorders (25.7%), and typically developing
for sleep disturbances in children. Before begin-
(54.5%;Heussler et al., 2013).
ning a drug treatment in children with NDDs,
Systematic reviews and meta-analyses of placebo-
different practical aspects should be taken into
controlled, randomized controlled trials (RCTs) in
account: (a) commonly these children are taking
children with NDDs, especially ASD, have demon-
other drugs (such as mainly sedatives or antiepilep-
strated that, compared to the control condition,
tics) and the eventual interactions should be con-
exogenous melatonin significantly improves sleep,
sidered cautiously; (b) the clinician should enquire
either by reducing the time to fall asleep (sleep onset
about previous sleep medications or homeopathic or
latency) or by increasing total sleep time, or both
OTC preparations to consider possible interactions
(Braam et al., 2009; Phillips & Appleton, 2004).
with other medications; (c) the age of the child and
In particular, a large clinical trial confirmed the
his/her clinical history should be taken into
efficacy of melatonin in the treatment of sleep
account; (d) a thorough sleep history/sleep diary
impairment in children with NDDs, using different
prior to any intervention being planned should be
doses, ranging from 0.5 to 12 mg; the main effects of
collected. Treatment goals must be established with
melatonin were reduced sleep latency (from 102 to
the parents and, if possible, with the patient and
55 min in 12 weeks) and increased total sleep time
should be realistic, clearly defined, and measurable.
(40 min;Appleton et al., 2012). Another placebo-
The immediate goal of treatment will usually be to
controlled study in 146 children (age 3–15 years)
alleviate or improve, rather than to completely elim-
with intellectual disability showed similar results
inate, sleep problems; (e) abrupt discontinuation of a
(Gringras et al., 2012). Another randomized, pla-
sleep medication should be avoided and the treat-
cebo-controlled, double-blind, crossover trial of con-
ment should be carefully monitored as there is a
trolled-release melatonin 5–15 mg 20–30 min before
natural inclination of the parents to give the lowest
bedtime in 50 children with NDDs and delayed sleep
dose (Pelayo & Dubik, 2008); (f) the choice of the
phase syndrome (DSPS) confirmed a significant
drug, in view of the lack of scientific evidence, should
efficacy in decreasing sleep latency (p < .01), albeit
be guided by an accurate evaluation of the main
with a nonsignificant improvement of total nighttime
complaint (i.e. difficulty falling asleep, night awak-
sleep by 31 min. (Wasdell et al., 2008).
enings, phase advance or delay, nocturnal hyperac-
Unlike traditional hypnotics such as chloral
tivity, midnight awakening, etc.) and also a correct
hydrate and the benzodiazepines, melatonin does
family history can be useful.
not affect sleep architecture (Bruni et al., 2015).
The three greatest unmet clinical needs in the
With regard to the efficacy/effectiveness of mela-
treatment of insomnia are residual daytime sedation,
tonin for specific NDDs, several studies have shown
tolerance/addiction potential, and improvement in
that melatonin (with doses of 1–10 mg given 30–
sleep maintenance.
60 min before bedtime) alone or in combination with
Table 2 reports the most commonly used pharma-
cognitive-behavioral therapy is effective in insomnia
cologic agents for insomnia in children with NDDs.
of children with ASD (Cortesi, Giannotti, Sebastiani,

© 2017 Association for Child and Adolescent Mental Health.


6
Table 2 Drugs/compounds commonly used to treat insomnia with special considerations for children with NDDs

Compound Dosage Mechanism of action Special considerations Effect on sleep structure Common adverse effects

Melatonin and melatonin agonists


Melatonin 1–3 mg at bedtime, ↑s Action on MT1 receptors ASD, Angelman, Rett, Smith– ↓ SOL; less efficacy in sleep Headache, nausea
to 6 mg maximum contribute to sleep-promoting Magenis syndrome, delayed maintenance, regulate
(no specific benefit effects; action on MT2 sleep phase disorder in ADHD circadian rhythms
at higher doses) receptors apparently mediate
effects on circadian rhythms
Oliviero Bruni et al.

Ramelteon 4–8 mg Potent, selective agonist of Sleep onset insomnia in ↓ SOL; main effect on sleep Somnolence, dizziness,
MT1 and MT2 receptors. intellectual disability or ASD onset, no effect on sleep nausea, fatigue, headache,
maintenance and insomnia
Antihistamines
Diphenhydramine 0.5–1 mg/kg Histamine H1-receptor Primary insomnia with ↓ SOL; ↓ arousal threshold Daytime drowsiness,
agonist, cross blood–brain frequent night awakenings gastrointestinal
barrier well accepted by parents (disturbances, vomiting,
constipation), paradoxical
excitation
Hydroxyzine 0.5–1 mg/kg Histamine H1-receptor Primary insomnia with ↓ SOL; ↓ arousal threshold Daytime drowsiness,
agonist, cross blood–brain frequent night awakenings gastrointestinal
barrier well accepted by parents (disturbances, vomiting,
constipation), paradoxical
excitation,
Niaprazine 1 mg/kg Phenylpiperazine; low or no Primary insomnia with ↓ SOL; ↓ arousal threshold, Daytime drowsiness,
binding affinity for the H and frequent night awakenings effect on sleep maintenance paradoxical excitation,
mACh receptors; potent and well accepted by parents convulsions
selective 5-HT and a-
adrenergic receptor
antagonist
Alpha2-adrenergic agonist
Clonidine 0.05–0.225 mg/day a Adrenergic agonist and Sleep onset insomnia in ADHD ↑ REM latency, stage 2, and Dizziness, orthostatic
or 0.1–0.3 mg imidazoline receptor agonist and ASD SWS hypotension, somnolence, dry
mouth, headache, fatigue,
hypotension
Gaba agonists
Clonazepam 0.25–0.5 mg; begin Potentiation of GABAergic Sleep onset insomnia, when ↓ SOL, suppresses SWS; ↓ Daytime sedation, rebound
with 0.01–0.03 mg/ system associated with RLS, arousal threshold insomnia, psychomotor/
kg/dose before bruxism, RMD cognitive impairment,
bedtime. Not exceed anterograde amnesia (dose-
0.2 mg/kg/dose or dependent); respiratory
1 mg depression
Zolpidem 5–10 mg Enhances the activity of the Insomnia, mainly used in ↓ SOL and WASO, ↑ TST, Clumsiness, confusion,
GABA, via selective agonism adolescence depression, headache,
at the benzodiazepine-1 (BZ1) anterograde amnesia, nausea
receptor;

(continued)

© 2017 Association for Child and Adolescent Mental Health.


Table 2 (continued)

Compound Dosage Mechanism of action Special considerations Effect on sleep structure Common adverse effects

Gabapentin 3–5 mg/kg before Structurally related to GABA RLS, epilepsy, resistant sleep ↑ SWS Sleepiness, dizziness,
bedtime but does not bind to GABA onset insomnia eosinophilia, behavioral
Titrate by 3–5 mg/ receptors, unclear disturbances, weight gain
kg every week to mechanism of action:
15 mg/kg interaction with voltage-gated
maximum calcium channels and NMDA
receptors
Antidepressants
Trimipramine 10–25 mg Antagonist of H, 5HT, and Sleep maintenance problems, ↓ sleep latency, ↑ REM latency Dry mouth, sedation, weight
alpha adrenergic receptors depression gain, dizziness, headache,
tachycardia, blurred vision,
constipation, urinary
retention
Imipramine 5–25 mg Blockade of 5HT, NE, and DA Sleep maintenance problems, ↓ sleep latency, REM Dry mouth, sedation, weight
reuptake depression suppression, ↑ REM latency gain, dizziness, headache,
lack of coordination, blurred
vision, constipation, urinary
retention, leukopenia
Amitryptiline 5–25 mg A serotonin-norepinephrine Sleep maintenance problems, ↓ sleep latency, REM Anticholinergic effects,
reuptake inhibitor, 5HT, depression suppression, ↑ REM latency sedation, weight gain,

© 2017 Association for Child and Adolescent Mental Health.


alpha adrenergic, H and dizziness, headache, blurred
mACh receprtors vision, constipation
Mirtazapine 7.5 mg-30 mg Presynaptic alpha2-adrenergic Sleep onset and sleep ↑ SWS; ↓ SOL Sedation, weight gain,
antagonist; potent antagonist maintenance problems, ASD, xerostomia
of 5-HT2 and 5-HT3 serotonin depression
and H1 histamine receptors
and moderate peripheral
alpha1-adrenergic and
muscarinic antagonist
Trazodone 1 mg/kg/dose; up to Low doses exploit trazodone’s Night awakenings; midnight ↓ SOL, improve sleep Dry mouth, nausea, vomiting,
25 mg for children potent actions as a 5HT2A and terminal insomnia; continuity, ↓ REM, ↑ SWS drowsiness, dizziness/light-
≥3 years; ↑ gradually antagonist, a1-adrenergic comorbidity with depression headedness, headache, and
until 50 mg in antagonist, and less relevant morning hangover. Less
adolescence in humans as an antagonist common or rare hypotension,
of H1-histaminic receptors tachycardia, serotonin
syndrome, and priapism.
Atypical antipsychotics
Risperidone 0.5–2 mg Blocks dopamine 2 receptors Insomnia with comorbid ↓ SOL and WASO, ↑ TST and Sedation, hypotension, weight
and 5HT 2A receptors; mild conditions like aggressive or SWS gain, hyperglycemia,
alpha 2 antagonist self-injurious behaviors dyslipidemia, tardive
dyskinesia, neuroleptic
malignant syndrome,
hyperprolactinemia,

(continued)
Treatment of insomnia in NDD
7
8
Table 2 (continued)

Compound Dosage Mechanism of action Special considerations Effect on sleep structure Common adverse effects

Aripiprazole 1–5 mg Dopamine partial agonist with Insomnia with comorbid ↓ SOL and WASO, ↑ TST and Sedation, dizziness, weight
action on dopamine 3 conditions like aggressive or SWS gain, headache
receptors; blockade of 5HT 2A self-injurious behaviors
receptors
Quetiapine 25 mg Blocks dopamine and 5HT 2A Insomnia with comorbid ↓ SOL and WASO, ↑ TST and Dizziness, headache, sedation,
receptors; modulating effect conditions like aggressive or SWS ortosthatic hypotension,
Oliviero Bruni et al.

on 5HT1A receptors; alpha self-injurious behaviors hyperglycemia, dyslipidemia,


adrenergic antagonist weight gain, tardive
dyskinesia
Orexin antagonists
Suvorexant 10 mg; maximum Blocks the binding of wake- Promising agent for use in ↓ SOL, ↓ WASO Drowsiness, headache,
20 mg promoting neuropeptides children and adolescents with abnormal dreams;
orexin A and orexin B to NDDs. antagonism of orexin
receptors OX1R and OX2R. receptors may also underlie
potential adverse effects such
as signs of narcolepsy/
cataplexy.
Supplements and various
Chloral hydrate 25–50 mg/kg Central nervous system Insomnia refractory to other ↓ SOL Sedation, impairment
depressant effects are due to pharmacological treatments respiratory function, nausea,
its active metabolite vomiting, cardiac arrhythmia,
trichloroethanol, mechanism hypotension, liver toxicity
unknown
Iron 2–6 mg/kg/day of Iron is a cofactor for tyrosine When poor sleep is reported in Supplemental iron was Constipation; nausea
elemental iron and hydroxylase (dopamine children with ADHD or NDDs, associated with ↑ TST
titrate to serum synthesis) and therefore affect serum ferritin levels should
ferritin level of ≥30– dopamine metabolism be monitored
50 ng/ml
5-OH-Tryptophan 2–5 mg/kg ↑ 5-HT synthesis in the central Effective in sleep maintenance ↑ SOL and ↓ WASO, ↑ or ↓ REM Gastrointestinal disturbances,
nervous system and latency and ↑ SWS nausea, vomiting, headache,
drowsiness, visual blurring,
muscle weakness
Vitamin D Infants and children: Vitamin D is related to Vitamin D and ferritin levels in Vitamin D supplementation Hypercalcemia, headache,
1,000–2,000 int. dopamine metabolism; children with NDDs and may improve sleep efficiency nausea, vomiting, confusion,
units/day for chronically low vitamin D is insomnia should be and quality abdominal pain, bone pain,
6 weeks followed by related to sleepiness and monitored polyuria, polydipsia,
maintenance dosing other symptoms of wake weakness, cardiac
of 400–800 int. impairment; may be a arrhythmias,
units/day cofactor for the development nephrocalcinosis
of OSA and OSA-associated
cardiovascular disease.

ADHD, attention deficit/hyperactivity disorder; ASD, autism spectrum disorder; GABA, c-aminobutyric acid; REM, rapid eye movements; SOL, sleep onset latency; WASO, wake after
sleep onset; SWS, slow-wave sleep; RLS, restless legs syndrome; RMD, rhythmic movement disorders; NDD, neurodevelopmental disability; NE, norepinephrine; DA, dopamine; 5HT,
serotonin; Ach, acetylcholine; OX, orexin.

© 2017 Association for Child and Adolescent Mental Health.


Treatment of insomnia in NDD 9

Panunzi, & Valente, 2012; Goldman et al., 2014) or melatonin. Previous reports of poor seizure control,
with Angelman syndrome (Andersen, Kaczmarska, poor asthma control, and adverse endocrinological
McGrew, & Malow, 2008; Braam, Didden, Smits, & problems during puberty have not been confirmed.
Curfs, 2008). Additionally, in nine girls with Rett Both systematic reviews and meta-analyses of RCTs
syndrome (melatonin 2.5–7.5 mg), actigraphy suggest that there are no significant adverse side
showed that sleep onset latency was significantly effects associated with the use of melatonin.
decreased while the number of nighttime awaken- Unanswered clinical questions on the use of
ings was not affected, and the mean total sleep time melatonin include whether slow-release prepara-
increased (McArthur & Budden, 1998). tions are superior to immediate-release in increasing
As for ADHD, a systematic review of the literature total sleep time, and whether a more rational and
concluded that melatonin given at doses 3–6 mg/ optimal prescription of melatonin might be achieved
night significantly reduced sleep onset latency and by measuring salivary melatonin before its use.
increased total sleep duration, but did not impact on Table 3 reports some clinical tips on the use of
daytime ADHD core symptoms (Cortese et al., 2013). melatonin, suggested during a recent expert consen-
Melatonin has also been found to be effective in sus conference (Bruni et al., 2015).
children with ADHD with DSPS and insomnia at a
dosage of about 5 mg (Smits et al., 2003; Van der Antihistamines. Histamine is a wake-promoting
Heijden, Smits, Van Someren, & Gunning, 2005; neurotransmitter, and its inactivation or suppres-
Van der Heijden, Smits, Van Someren, Ridderinkhof, sion in various animal models has led to sedation
& Gunning, 2007; Weiss, Wasdell, Bomben, Rea, & and disrupted wakefulness patterns (Mignot, Taheri,
Freeman, 2006). A recent randomized controlled trial & Nishino, 2002).
(RCT) suggested that melatonin (3–6 mg) may be First-generation antihistamines are lipid soluble
effective in reducing sleep latency in ADHD children and pass through the blood–brain barrier; they bind
with sleep onset delay caused by methylphenidate to H1 receptors in the central nervous system (CNS)
(Mohammadi et al., 2012). and have minimal effects on sleep architecture.
Finally, melatonin has been found to be effective Ethanolamines (such as diphenhydramine) have
also for sleep problems in children with Smith– potent sedative effects as do piperazine derivatives
Magenis syndrome, a rare syndrome characterized (such as hydroxyzine).
by an inverted circadian rhythm of melatonin. An Diphenhydramine is the most commonly used and
open-label study demonstrated that the most effica- is a competitive H1-histamine receptor blocker. Peak
cious treatment is a combination of acebutolol, a b1- blood and tissue levels are achieved within 2 hr of
adrenergic antagonist, given in the morning that ingestion. The recommended dosage for adults is 25–
decreases daytime plasma melatonin levels during 50 mg, whereas in children the effective dose is
the day and exogenous melatonin at night to replace
Table 3 Recommendations for prescribing melatonin for
normal peak endogenous melatonin (De Leersnyder insomnia in children with NDDs
et al., 2001).
From a practical standpoint, in some patients with Minimum No studies reported the minimum age for
age for administering melatonin. As it has been
NDDs, the loss of efficacy of melatonin treatment administration used in high dosage in infants to prevent
after an initial good response is a major problem. neuronal injuries without side effects,
This is possibly caused by slow metabolism because we can expect that melatonin
of decreased activity of the CYP1A2 enzyme in some administration after 6 months of age
individuals (Braam et al., 2013). This may result in could be quite safe
Time of If used as chronobiotic, administer
increased daily melatonin levels. Consequently, administration melatonin 3–4 hr before actual sleep
melatonin levels accumulate and after some time, in children onset time
the circadian melatonin rhythm is lost. This loss of If used as sleep inductor 30 min before
circadian rhythm might explain why exogenous Dosage If used as chronobiotic, start with a low
melatonin loses its effectiveness over time (Braam dose of 0.2–0.5 mg fast release
melatonin 3–4 hr before bedtime;
et al., 2010). As a consequence, there is now a increase by 0.2–0.5 mg every week as
greater understanding that low, rather than high, needed (maximum 3 mg; adolescents:
doses (0.5 mg) can be effective for some children, 5 mg) until effect
with diminishing benefit with doses exceeding 6 mg. When 1 mg is effective: try lower dose
A single study reported that melatonin for treat- Maximum dose: <40 kg: 3 mg; >40 kg:
5 mg
ment of chronic sleep onset insomnia in children is Treatment Treatment duration should be tailored to
effective in a dosage of 0.05 mg/kg given at least 1– duration the specific patient in relation to the
2 hr before desired bedtime (Van Geijlswijk, van der peculiar neurodevelopmental disabilities
Heijden, Egberts, Korzilius, & Smits, 2010). but in general should be not <1 month
Headaches, confusion, dizziness, cough, and Stop melatonin treatment once a year
during 1 week (preferably in summer)
rashes have been reported, but these common after a normal sleep cycle is established
symptoms are likely to be coincidental or caused by
impurities in the unregulated formulations of (adapted from Bruni et al., 2015).

© 2017 Association for Child and Adolescent Mental Health.


10 Oliviero Bruni et al.

0.5 mg/kg up to 25 mg (Russo, Gururaj, & Allen, duration of REM sleep. At medium-to-high doses
1976). Hydroxyzine is effective at 0.5–1 mg/kg in (0.1–0.3 mg/nocte), clonidine appears to have post-
children (Sezer & Alehan, 2013). Other antihis- synaptic activity on the a2-adrenergic receptors,
taminines have been used: trimeprazine was used which results in decrease of acetylcholine, increas-
in 22 children with night awakenings showing a ing REM latency, stage 2 sleep, and slow-wave sleep
moderate improvement (France, Blampied, & Wilkin- (Delbarre & Schmitt, 1971).
son, 1999); niaprazine (1 mg/kg in a single dose at The most commonly reported side effects of cloni-
bedtime) showed a decrease of sleep onset latency dine include drowsiness, transient sedation, head-
and an increase of sleep duration (Ottaviano, Gian- ache, dizziness, fatigue, somnolence, insomnia,
notti, & Cortesi, 1991) even if compared with benzo- hypotension, and bradycardia.
diazepines (Montanari, Schiaulini, Covre, Steffan, & In a retrospective chart review, Ingrassia and Turk
Furlanut, 1992). (2005) found clonidine to be an effective therapeutic
Although antihistamines are the most prescribed or intervention for alleviating sleep disturbances in six
obtained OTC agents for pediatric insomnia (Schnoes, children with NDDs, aged 6–14 years at a dose of
Kuhn, Workman, & Ellis, 2006), randomized con- 0.05–0.1 mg at bedtime. No relevant side effects
trolled data in children are lacking. A nonrandomized were reported.
trial showed a significant decrease in sleep latency In another retrospective review, 19 children with
time and number of awakenings (Russo et al., 1976), ASD were treated with oral clonidine at 50 lg with a
while another study in infants showed lack of supe- slow titration up to 100 lg, 30 min before bedtime,
riority compared to placebo (Merenstein, Diener- reporting reduced sleep latency and nocturnal awak-
West, Halbower, Krist, & Rubin, 2006). enings. The most common reported adverse effects
The most common adverse reaction to antihistami- included skin irritation with transdermal adminis-
nes at therapeutic doses is impaired consciousness. tration, and daytime drowsiness with administration
With overdose, adverse effects are predominately of tablets (Ming, Gordon, Kang, & Wagner, 2008).
anticholinergic, including fever, mydriasis, blurred Overall, in a comprehensive literature review of
vision, dry mouth, constipation, urinary retention, noncontrolled studies, Hollway and Aman (2011)
tachycardia, dystonia, and confusion. There are some showed that clonidine was effective on sleep distur-
reports on toxicity of diphenhydramine with catatonic bances in children with comorbid ASD and other
stupor, anxiety, visual hallucinations, and more NDDs at doses ranging from 0.05 to 0.225 mg/day.
rarely, respiratory insufficiency and seizures (Din- Of note, in children who are taking concomitant
ndorf, McCabe, & Frierdich, 1998) with fatal toxicity CNS-depressing agents and in individuals with
in five infants of 6–12 weeks old (Baker et al., 2003). hemodynamic instability or cardiac pathology, the
Hydroxyzine seems to be safer and no fatal cases have use of clonidine should be accurately monitored
been reported (Magera, Betlach, Sweatt, & Derrick, (Spiller et al., 2005).
1981).
It is important to highlight that tolerance to Benzodiazepines: Clonazepam. Benzodiazepines
antihistaminergic agents can develop quickly and bind to the benzodiazepine subunit of the c-amino
some children can experience dramatic and para- butyric acid (GABA) chloride receptor complex, facil-
doxical overarousal (Gringras, 2008). Due to the lack itating the action of the inhibitory neurotransmitter
of controlled trials and the relatively poor tolerability GABA. These hypnotics have long been the first-
profile, we deem that antihistamines should not be choice treatment for insomnia in adults, but their use
considered as a first-line option for the treatment of raises concerns about cognitive impairment, rebound
sleep disturbances in children with NDDs. insomnia, and the potential risk for dependence.
Only a very limited number of open studies testing
Clonidine. Clonidine is a central and peripheral a- the effectiveness and tolerability of benzodiazepines
adrenergic agonist that acts on presynaptic neurons in children have been published. In five children
and inhibits noradrenergic release and transmis- aged 1.5–10 years with Williams syndrome, clon-
sion, approved by the FDA for the treatment of azepam 0.25–0.75 mg at bedtime, determined an
hypertension, and, in its extended release form, for immediate improvement in nighttime awakenings
ADHD. Due to its sedative effects, clonidine is and daytime behaviors in four of the five patients
commonly prescribed as a sleep aid in children, that persisted at 3- to 6-month follow-up (Arens
but there are no well-controlled studies available et al., 1998). An effect on REM sleep behavior
(Nguyen, Tharani, Rahmani, & Shapiro, 2014). disorder in a child with autism has been reported
It is hypothesized that clonidine produces sedation (Thirumalai, Shubin, & Robinson, 2002).
decreasing norepinephrine via negative feedback by Concerns on their tolerability profile, along with
agonism of the a2-adrenergic receptors at the level of the limited availability of evidence-based data in the
the locus coeruleus, which would increase rapid eye pediatric population, contribute to limit their use in
movements (REM) sleep. Administration of low doses children (Owens, 2011). Short-term or as-needed
of clonidine (range 0.025–0.05 mg) has little effect on administration is the most frequent suggested treat-
sleep and can either increase or decrease the ment. Based on available data, clonazepam may

© 2017 Association for Child and Adolescent Mental Health.


Treatment of insomnia in NDD 11

represent a treatment option in children with arousal Side effects of tricyclics include anxiety and agita-
disorders (parasomnias) or periodic leg movement tion, anticholinergic effects (e.g. blurred vision, dry
disorder or restless legs syndrome (RLS), but future mouth, urinary retention, and orthostatic hypoten-
trials focused on objective sleep measures and safety sion), cardiotoxicity, and worsening of RLS symptoms.
issues are needed.
Mirtazapine: Mirtazapine is an a2-adrenergic, 5-
Non-benzodiazepine sedative-hypnotics (Z drugs). Z HT receptor agonist with a high degree of sedation at
drugs are non-benzodiazepine receptor agonists that low doses (Younus & Labellarte, 2002). It has been
bind preferentially to GABAA receptor complexes shown to decrease sleep onset latency, increase
containing a1 subunits; they have minimal effects sleep duration, and reduce wake after sleep onset
on sleep architecture with a slight increase slow- (WASO), with relatively little effect on REM sleep
wave sleep (Zisapel, 2012). (Wichniak, Wierzbicka, & Jernajczyk, 2012).
There are very few studies conducted in children. There is a single open-label study in autistic
One trial reported children with ADHD and insomnia children and children with other forms of pervasive
aged 6–11 years or 12–17 years receiving treatment developmental disorders, which suggests some effi-
with zolpidem at 0.25 mg/kg per day (max 10 mg/ cacy for insomnia; 9 out of 26 subjects (34.6%) were
day) versus placebo reported a mean change in judged to be responders, based on their improve-
latency to persistent sleep at week 4 that did not ment in a variety of symptoms, including aggression,
differ between zolpidem and placebo groups. No self-injury, irritability, hyperactivity, anxiety,
next-day residual effects of treatment and no depression, and insomnia (Posey, Guenin, Kohn,
rebound phenomena occurred after treatment dis- Swiezy, & McDougle, 2001).
continuation (Blumer, Findling, Shih, Soubrane, &
Reed, 2009). Eszopiclone failed to show a benefit Trazodone: Trazodone is one of the most sedating
over placebo in a group of 371 children aged 6– antidepressants available and the most widely stud-
17 years with ADHD and sleep disturbances; dose- ied of antidepressants in terms of sleep in adults. It
dependent adverse events were reported in 46–61% was the most commonly prescribed insomnia med-
of patients (Sangal, Blumer, Lankford, Grinnell, & ication for children with mood and anxiety disorders
Huang, 2014). The most frequent adverse events in a survey of child and adolescent psychiatrists in
(>5%) were dizziness and headache and also disin- the United States (Owens et al., 2010).
hibition and hallucinations have been reported Its action is mediated by the 5-HT2A/C antago-
(Liskow & Pikalov, 2004). nism and inhibition of postsynaptic binding of sero-
tonin and blocking of histamine receptors.
Antidepressants. Tricyclic and atypical antide- Recent empirical evidence suggests that trazodone
pressants (mirtazapine, nefazodone, and trazodone) may interact with the melatonin system. Giannaccini
are used in clinical practice to treat insomnia in et al. (2016) measured clinical and melatonin
adult and pediatric populations. Their effects on parameters before and after 1 month of therapy with
sleep are mediated by an action on different neuro- trazodone in insomniac mood-depressed patients.
transmitters, such as serotonin, histamine, and Patients with refractory depressed-mood and insom-
acetylcholine, involved in sleep regulation. nia improved after treatment and responsive
patients excreted more urinary 6-hydroxy-melatonin
Tricyclic antidepressants: Tricyclic antidepressants sulfate than before treatment, reflecting an
(amitriptyline, trimipramine, and doxepin) determine enhanced nighttime production of the pineal hor-
a reduction of REM and slow-wave sleep and have mone in these subjects and suggesting an interac-
been used to treat insomnia. Both amitriptyline and tion between trazodone and melatonin system.
trimipramine have been shown to have sedative Based on these data, the authors supported the role
effects in adults (Holmberg, 1988; Riemann et al., of melatonin as a biological indicator of prohypnotic
2002; Ware, Brown, Moorad, Pittard, & Cobert, and antidepressant benefits of trazodone.
1989). Trazodone suppresses REM sleep and increases
There are no data supporting the use of amitripty- slow-wave sleep, which may prove to have beneficial
line or trimipramine in children with NDDs; however, effects in memory function of children who are chal-
amitriptyline is commonly used in children with lenged intellectually and who struggle to generalize and
NDDs beginning with a very low dose (5 mg) followed maintain adaptive skills of daily living.
by gradual increase, up to 50 mg, until the desired Despite being commonly used to manage insomnia
effect is achieved. in the clinical practice, only very few studies have
been conducted in children and adolescents to test
Doxepin: In adults, doxepin (3–6 mg) has been the effectiveness and tolerability of trazodone as a
shown to improve sleep maintenance and early sleep medication.
morning awakenings; when used in children with Trazodone has been used for insomnia in 17
pruritus, it led to CNS depression (Zell-Kanter, children with opsoclonus–myoclonus syndrome
Toerne, Spiegel, & Negrusz, 2000). (Pranzatelli et al., 2005). The starting dose of

© 2017 Association for Child and Adolescent Mental Health.


12 Oliviero Bruni et al.

25 mg was increased to a maximum of 100 or treatment of neuropathic pain and RLS in addition
150 mg depending on age. The effects on sleep were to its original purpose as an anticonvulsant med-
not dose-dependent and low dosages were effective ication. However, its precise pharmacological mech-
for insomnia. Additionally, anecdotal reports show anism in humans remains unknown. In addition to
the efficacy of trazodone mainly in midnight and its demonstrated efficacy in these indications,
terminal insomnia. patient-reported sleep assessments among a variety
The most commonly reported side effects are dry of clinical conditions suggest that gabapentin has
mouth, nausea, vomiting, drowsiness, dizziness/ beneficial effects on sleep. The improvement of
light-headedness, headache, and morning hangover. sleep might be mediated by the increase in slow-
Less common or rare side effects are hypotension, wave sleep and the decrease of WASO (Rosenberg
tachycardia, serotonin syndrome, and priapism (Bos- et al., 2014).
sini et al., 2015). It is not commonly indicated in Rett In a recent case series, gabapentin was found to
syndrome for the risk of QT interval prolongation. It be a safe and a well-tolerated treatment for sleep
should be noted that, as the doses of trazodone for onset and sleep maintenance insomnia in 23
insomnia are lower than those used for depression, children, 87% of whom had NDDs. Following
the incidence of side effects is generally also lower. gabapentin treatment initiated at an average dose
of 5 mg/kg (range 3–7.5 mg/kg) 30–45 min before
Atypical antipsychotics. Atypical antipsychotics bedtime, with titration to a maximum dose of
such as risperidone, quetiapine, aripiprazole, and 15 mg/kg (range 6–15 mg/kg), 78% of children
olanzapine are typically used off-label in children showed improvement in sleep (as reported by
with psychiatric or developmental disorders to parents). Furthermore, this beneficial response
reduce disruptive behaviors (Capone, Goyal, Grados, was noted at doses of 5–15 mg/kg orally at
Smith, & Kammann, 2008; Masi et al., 2009). Ris- bedtime, much lower than the recommended dose
peridone and olanzapine have been prescribed also to treat epileptic seizures (40 mg/kg divided three
for sleep disturbances in children, but no studies are times daily). Side effects in a few cases included
available evaluating their safety and effectiveness agitated nighttime awakenings and difficulty falling
(Hollway & Aman, 2011; Meltzer, Mindell, Owens, & asleep (Robinson & Malow, 2013).
Byars, 2007; Schnoes et al., 2006).
Tolerability profile of these medications can be highly Ramelteon. Ramelteon is a synthetic melatonin
problematic: excessive weight gain may exacerbate any receptor agonist with high affinity for the MT1 and
sleep-disordered breathing present in the child; meta- MT2 receptors, and is approved by the FDA for use in
bolic effects like hyperglycemia or hyperprolactinemia adults. Only few case reports have been published in
may be unsafe in certain clinical conditions (i.e. Down children with NDDs reporting in general limited effi-
syndrome or Prader–Willi syndrome). cacy on night awakenings (Asano, Ishitobi, Kosaka,
Atypical antipsychotics (e.g. risperidone or arip- Hiratani, & Wada, 2014; Miyamoto et al., 2013;
iprazole) should be considered to treat a comorbid Stigler, Posey, & McDougle, 2006).
condition (i.e. aggressive, self-injurious behaviors in
children with ASD) and they might help to relief from Chloral hydrate. Chloral hydrate has been sug-
insomnia when dosed at bedtime. gested to be effective for nighttime sedation in
A small randomized and placebo-controlled study children as it was initially considered a safe agent
of quetiapine (25 mg/nocte, a relatively low dose) in in infants and young children at dosages of 25–
adults showed a statistically nonsignificant trend 100 mg/kg/day. However, respiratory depression
toward improvement in total sleep time and reduced and hepatotoxicity have been reported after admin-
sleep latency (Tassniyom, Paholpak, Tassniyom, & istration of chloral hydrate (Biban, Baraldi, Petten-
Kiewyoo, 2010). nazzo, Filippone, & Zacchello, 1993).
There is a single open-label trial involving 11 Therefore, chloral hydrate should be avoided or at
children with ASD which did demonstrate a signif- least carefully controlled in children with NDDs at
icant reduction in sleep disturbance (Golubchik, risk for obstructive sleep apnea (Sheldon, 2001).
Sever, & Weizman, 2011). Additionally, it may cause gastric distress, nausea,
Due to the paucity of data and, especially, to the vomiting, drowsiness, dizziness, malaise, and fati-
tolerability profile, the use of neuroleptics for insom- gue. Children may experience idiosyncratic reac-
nia in children, considering their tolerability profile, tions characterized by confusion, disorientation,
is generally not recommended. and paranoia. Used chronically, chloral hydrate is
habit forming and associated with tolerance (Glaze,
Gabapentin. Food and Drug Administration 2004).
approved gabapentin for the treatment of partial
seizures in 1993. It was originally designed as a Orexin antagonists. Recently, orexin neuropep-
precursor of GABA that easily enters the blood– tides have been identified as regulators of the sleep/
brain barrier, and increases brain synaptic GABA. wakefulness transition and shown to aid an initial
More recently, it has been approved for the transitory effect toward wakefulness by activating

© 2017 Association for Child and Adolescent Mental Health.


Treatment of insomnia in NDD 13

cholinergic/monoaminergic neural pathways of the serotonin. The effects of 5-HTP on sleep structure are
ascending arousal system (Chow & Cao, 2016). conflicting: increase or decrease of REM and increase
Orexins hold an important role in the wakefulness of slow-wave sleep (SWS; Meolie et al., 2005).
promoting ascending arousal system by having an The exact mechanism of action of the sedative
excitatory effect on almost every wake-promoting effects of 5-HTP is not completely clear and it is not
neuronal group of reticular ascending system and sure that it is mediated only by conversion in sero-
represent the critical modulators of the sleep–wake tonin. Serotonin (5-HT) should not be considered
cycle homeostasis. either wake-promoting or SWS-promoting, the effect
Orexin receptor antagonists can be classified on the of 5-HT on sleep–wake behavior would depend upon
basis of receptor-binding affinities, either as selective the activation of the serotonergic system (systemic
orexin receptor antagonists (i.e. selective for OX1 or administration of low vs. high doses of the precursor
OX2 receptors) or dual orexin receptor antagonists 5-HTP), and the time at which the activation occurs
(i.e. compound with spread binding capacity to OX1 (light vs. dark period of the light–dark cycle; Imeri,
and OX2;Kumar, Chanana, & Choudhary, 2016). It Mancia, Bianchi, & Opp, 2000). It has been hypoth-
has been hypothesized that antagonizing both orexin esized that the 5-hydroxytryptophan-related increase
receptors would elicit the most powerful sleep-pro- of SWS during the dark period depends upon the
moting effects (Morairty et al., 2012). synthesis or release of as yet to be identified sleep-
Dual orexin receptor antagonists molecules (al- promoting factors (Monti, 2011).
morexant and suvorexant) administered to healthy In terms of available evidence, one study with LT at
volunteers and patients with insomnia have been different dosages (250 mg, 1 g, and 3 g) in adults
shown to effectively reduce the number of awaken- showed a significant improvement in sleep latency,
ings and sleep latency while increasing total sleep along with a reduction in WASO, with a moderate
time (Kumar et al., 2016). effect on quality of sleep. None of the published
The most frequent dose-dependent adverse effects studies reported systematic information regarding
are mild somnolence, headaches, dizziness, and adverse effects associated with tryptophan (Hart-
abnormal dreams (Bennett, Bray, & Neville, 2014). mann, Lindsley, & Spinweber, 1983; Hudson, Hud-
The major route of metabolism for almorexant and son, Hecht, & MacKenzie, 2005; Spinweber, 1986).
suvorexant is CYP3A. Suvorexant is the most widely No data are available on the effects of 5-HTP on
studied and concomitant use with CYP3A Inhibitors is insomnia symptoms in children with NDDs.
not recommended as the effects could be increased
about two- to threefolds (e.g. ketoconazole, itracona- Iron. Iron is a cofactor for tyrosine hydroxylase, the
zole, clarithromycin, nefazodone, ritonavir, saquinavir, enzyme responsible for catalyzing the conversion of
nelfinavir, indinavir, boceprevir, telaprevir, ciproflox- the amino acid L-tyrosine to dopamine.
acin, diltiazem, erythromycin fluconazole, grapefruit Iron deficiency anemia in infancy has been reported
juice, and verapamil). On the other hand, strong CYP3A to be associated with higher motor activity during
inducers can substantially decrease almorexant and sleep, shorter night sleep duration, and higher fre-
suvorexant exposure (e.g. rifampin, carbamazepine, quency of night waking (Kordas et al., 2008), and
and phenytoin;Kishi, Matsunaga, & Iwata, 2015). supplemental iron was associated with longer sleep
This category of compounds might be promising duration (Kordas et al., 2009).
for children with NDDs as they act on a different Because iron deficiency is common in children,
neurotransmitter system with less interactions with measuring the ferritin level is reasonable. Different
other drugs commonly used in these children. RCTs reports showed a relation between low serum ferritin
are needed to assess both the short- and long-term levels and insomnia associated with sleep hyperki-
effects of these medications, as well as their efficacy nesia [i.e. RLS or periodic limb movements of sleep
in comorbid diseases that affect sleep architecture. (PLMs)] in children with ADHD or ASD (Abou-
Khadra, Amin, Shaker, & Rabah, 2013; Cortese,
Tryptophan/5-hydroxytryptophan (5-HTP). Tryp- Konofal, Dalla Bernardina, Mouren, & Lecendreux,
tophan is a precursor of serotonin and melatonin, 2009; Dosman et al., 2007).
widely used in the 1980s for the treatment of sleep A recent review showed an increased incidence of
disorders and headache prophylaxis. It does not PLMs (42%) compared with controls (8%) in 53
have opioid-like effects and does not impact on pediatric patients with ASD with low serum ferritin
cognitive performance or inhibit arousal from sleep level (below 35 ng/ml); sleep fragmentation and poor
(Lieberman, Kane, & Reife, 1985). Several positive sleep efficiency were associated with lower median
effects on sleep have been reported: improvement of ferritin level (Youssef, Singh, Huntington, Becker, &
sleep latency and decrease of arousals (Hartmann & Kothare, 2013).
Spinweber, 1979; Schneider-Helmert & Spinweber, In 102 children (68 with ASD, 18 typically devel-
1986). 5-HTP is the intermediate metabolite of the oping, and 16 with developmental delay), an increase
essential amino acid L-tryptophan (LT) in the biosyn- of PLMs has been found (Lane et al., 2015), but
thesis of serotonin. It easily crosses the blood–brain serum ferritin level was not significantly correlated
barrier and effectively increases CNS synthesis of to any sleep parameter.

© 2017 Association for Child and Adolescent Mental Health.


14 Oliviero Bruni et al.

Iron supplementation (6 mg/kg/day of elemental Vitamin D is related to dopamine metabolism. It


iron) in 33 children with ASD showed an improve- could be useful to investigate vitamin levels in
ment of restless sleep score but no relation was association with iron parameters in children with
found with serum ferritin concentrations (Dosman NDDs and insomnia associated with motor hyperac-
et al., 2007). tivity during sleep.
When poor sleep is reported in children with ADHD
or NDDs, serum ferritin levels should be monitored
and questions on restless sleep should be asked. If Conclusions
ferritin levels are <50 lg/L, 1–2 mg/kg/day of ele- Insomnia in children with NDDs, along with the other
mental iron (up to 6 mg/kg/day of elemental iron) neurobehavioral comorbidities, affects the quality of
should be administered (Abou-Khadra et al., 2013; life of both children and families, is associated with
Cortese, Konofal, et al., 2009). Parents should be poorer developmental outcome, and contributes to
asked for a personal and family history of hemochro- worsen behavioral disturbances.
matosis. While behavioral interventions should be the first-
line approach, in a sizable portion of cases, a pharma-
Vitamin D. Clinical research on the relation between cological approach needs to be considered. Despite the
vitamin D and sleep is ongoing, and a few studies have widespread use of pharmacological treatment, the lack
been published on the role of vitamin D metabolism of well-designed, controlled studies on the efficacy/
and sleep disorders. Preliminary data suggest that effectiveness, tolerability, dosage, and safety profile of
altered vitamin D metabolism could play an important hypnotic medications in children raise the need of
role in the presentation and severity of sleep disorders. further research in this field of sleep medicine.
Low vitamin D concentrations may impact sleep qual- The lack of research in this area is detrimental for
ity via increased pain, myopathy, immune dysregula- children and their families and well-conducted trials
tion, and cardiovascular disease (McCarty, Chesson, should be performed, based on the physiopathology
Jain, & Marino, 2014). of the disorders, evaluating also the presence of
The few published studies suggest vitamin D sup- other comorbid sleep disorders and addressing the
plementation may improve sleep quality; especially in correct choice of drugs, not based only on their
patients with neurologic complaints who also have sedative or anxiolytic effects.
evidence of abnormal sleep. Most patients have With the support of expert recommendations and
improvement in neurologic symptoms and sleep but empirical evidence, we propose two algorithms for the
only by maintaining a narrow range of 25(OH) vitamin management of chronic insomnia in children with
D3 blood levels of 60–80 ng/ml (Gominak & Stumpf, NDDs. Figure 2 shows the management algorithm for
2012). chronic insomnia based on its main clinical

CHRONIC INSOMNIA IN NDDs

Psychological and
Drug treatment
behavioral tratment

Irregularity and Motor hyperactivity


Middle of the night Frequent nocturnal Abnormal pattern of
Sleep Hygiene difficulty in sleep with nocturnal NREM parasomnias
awakening awakenings sleep-wake timing
onset awakenings

Cognitive-behavioral Sedating Atypical Circadian rhythm


Antihistaminics Antihistaminics Consider RLS, PLMD Clonazepam
therapy antidepressants antipsychotics disorder

Iron, vitamin D,
Other behavioral 5-hydroxy-
Melatonin Trazodone Benzodiazepines gabapentin, Melatonin
treatment tryptophan
dopamine agonists

Clonidine Mirtazapine Z drugs Antihistaminics

Atypical
Clonazepam
antipsychotics

Z drugs

Figure 2 Management algorithm for chronic insomnia in patients with neurodevelopmental disabilities based on our analysis of the
retrospective data and expert recommendations. Note: The drugs indicated in the flowchart are represented as a hierarchy of choice
based on expert clinical experience and should be considered as a generic indication. The choice, the combinations of the drugs, and the
switch between the drugs should be adapted in the single case

© 2017 Association for Child and Adolescent Mental Health.


Treatment of insomnia in NDD 15

Insomnia in impairments, involving not only the child but also


specific NDDs the entire family, are expected. A better quality of
sleep may ameliorate daytime behavior and even
If comorbid with cognitive development and, for sure, might lead to an
Autism spectrum Intellectual
ADHD anxiety or
disorders disability
depression improvement of sleep-related psychological distress
in the family.
CBT, sleep CBT, sleep CBT, sleep
Benzodiazepines The assessment should consider medical and
hygiene hygiene hygiene
psychiatric contributing factors, primary sleep dis-
orders, and maladaptive behaviors related to sleep.
Iron, vitamin D, Sedating The correct treatment should follow a specific pat-
Melatonin Melatonin
gabapentin antidepressants
tern: a thorough sleep history/sleep diary prior to
any intervention being planned, behavioral treat-
Sedating
Melatonin
antidepressants
Z drugs ment strategies through the parents, circadian
rhythm regulation, and pharmacological treatment.
The use of medications for pediatric insomnia
Atypical
Clonidine
antipsychotics
Benzodiazepines should be diagnostically driven, and should be
implemented in conjunction with empirically based
Sedating
behavioral treatment strategies and adequate sleep
Z drugs Gabapentin
antidepressants hygiene.
Future studies should address a number of
Atypical
shortcomings identified in our review. First, it is
Benzodiazepines
antipsychotics imperative to carry out new studies to identify
objective and specific outcome indicators that can
Figure 3 Management algorithm for chronic insomnia in patients give measures of wake time after sleep onset, sleep
with specific neurodevelopmental disabilities based on our onset latency, number of awakenings, sleep time,
analysis of the retrospective data and expert recommendations.
or sleep efficiency. The use of actigraphy should be
Note: The drugs indicated in the flowchart are represented as a
hierarchy of choice based on expert clinical experience and an integrative part of studies in this field and also
should be considered as a generic indication. The choice, the integrate information from multiple informants (e.g.
combinations of the drugs, and the switch between the drugs parents, teachers, therapists, etc.). Second, a better
should be adapted in the single case understanding of the specific effects of the available
treatments, in relation to specific clinical popula-
manifestations. Figure 3 shows the management tions, is needed (e.g. RLS in children with ADHD or
algorithm for chronic insomnia in patients with some respiratory disturbances in Rett’s syndrome). Third,
specific NDDs. Importantly, given the lack of head-to- well-designed, double-blind RCTs on large samples
head trials and network meta-analyses, it is not of children with NDDs comparing the different
possible to provide an evidence-based hierarchy of drugs against placebo should be carried out.
available medications in terms of efficacy/effective- Importantly, head-to-head and pragmatic trials
ness and tolerability, although, given their tolerability would be important to understand how different
profile, we discourage the use antipsychotics and drugs compare and rank in terms of effectiveness
benzodiazepines as first-line treatment for sleep dis- and tolerability.
turbances in children with NDDs. The drugs indi- Fourth, studies should be devoted to identifying
cated in the flowchart are represented as a hierarchy the specific doses of all agents that may be required
of choice based on expert clinical experience and in younger children, and their potential for side
should be considered as a generic indication. The effects and drug–drug interactions.
choice, the combinations of the drugs, and the switch Fifth, an important step of future studies would be
between them should be adapted to each single case. to analyze if drugs are able to improve cognitive
Future research will hopefully lead to a develop- functions in specific populations by improving sleep,
ment of a drug with proved efficacy and suitable also together with cognitive- behavioral therapy.
safety profile that will allow a better health and Finally, there is a need to investigate the long-term
quality of life of children and adolescents with NDDs effectiveness of the different drugs by analyzing the
and their families. possible tolerance and their eventual lack of efficacy
over time (i.e. benzodiazepines).

Recommendations for future research


The treatment of insomnia in special populations Supporting information
such as children with NDDs is a field that needs to be Additional Supporting Information may be found in the
better explored because it is extremely important to online version of this article:
improve sleep quality and quantity in children with Appendix S1. Search strategy and results from each
NDDs. Improvement in insomnia-related daytime database.

© 2017 Association for Child and Adolescent Mental Health.


16 Oliviero Bruni et al.

organization; R.F. declares no financial disclosure. The


Acknowledgements authors have declared that they have no competing or
O.B. served as a consultant and provided expert testi- potential conflicts of interest.
mony for Angelini; M.A. declares no financial disclosure;
F.C., A.C., and G.E. were employees of Angelini during
the conduct of the study; S.C. has received grant or
research support from the Solent National Health Service Correspondence
(NHS) Trust, UK. He has received honorarium and travel Oliviero Bruni, Department of Developmental and Social
expenses from the Association for Child and Adolescent Psychology, Sapienza University Via dei Marsi 78 –
Mental Health (ACAMH), United Kingdom, a nonprofit 00185, Rome, Italy; Email: oliviero.bruni@uniroma1.it

Key points
• Insomnia represents a common problem in children with NDDs, affects the quality of life of both children and
families, is associated with poor developmental outcome, and contributes to worsen behavioral disturbances.
• Good sleep practices and behavioral interventions are the first recommended treatments for pediatric
insomnia in both healthy and NDD children; when a pharmacologic intervention is considered, behavioral
interventions should always be associated.
• There are no approved sleep medications by the United States FDA or EMA for pediatric insomnia so that
medications for sleep disturbances in children with NDDs are prescribed off-label. Based on available evidence,
melatonin appears to be the safest choice for children with NDDs.
• Benzodiazepines are not recommended in children and should only be used for transient insomnia; clonidine is
used to improve sleep onset latency in ADHD subjects; trazodone and mirtazapine hold promise but require
further studies.
• Orexin antagonists might be promising for children with NDDs as they act on a different neurotransmitter
system with fewer interactions with other drugs commonly used in these children; however, no studies are
available in children with NDDs.
• While no comprehensive evidence-based information is available to rank the available drugs in terms of
efficacy and tolerability, we present our recommendations for the management of chronic insomnia in
children with NDDs, based on available empirical evidence or expert consensus.

Areas for future research


• A standardized assessment of insomnia in children with NDDs should consider medical and psychiatric
contributing factors, primary sleep disorders, and maladaptive behaviors related to sleep.
• Research on treatment of insomnia in this special population should focus on a multimodal and individualized
approach: behavioral treatment strategies through the parents, circadian rhythm regulation, and pharma-
cological treatment. This will hopefully allow better health and quality of life of children and adolescents with
NDDs and their families.
• Despite the widespread use of pharmacological treatment, the lack of well-designed, controlled studies
concerning the efficacy, tolerability, dosage, and safety profile of hypnotic medications in children raise the
need of further research.
• Well-controlled studies employing both objective polysomnography and subjective sleep measures are needed
to determine the efficacy and safety of the currently prescribed pediatric sleep drugs.
• Mirtazapine, trazodone, and orexin antagonists are promising in the treatment of insomnia, but RCTs are
needed to assess both the short- and long-term effects of these medications, as well as their efficacy in
comorbid diseases that affect sleep architecture.
• Future, rigorous protocols are needed to guide clinical recommendations useful for clinical practitioners in
their everyday practice.

Allen, K.D., Kuhn, B.R., DeHaai, K.A., & Wallace, D.P. (2013).
References Evaluation of a behavioral treatment package to reduce
Abou-Khadra, M.K., Amin, O.R., Shaker, O.G., & Rabah, sleep problems in children with Angelman Syndrome.
T.M. (2013). Parent-reported sleep problems, symptom Research in Developmental Disabilities, 34, 676–686.
ratings, and serum ferritin levels in children with atten- Andersen, I.M., Kaczmarska, J., McGrew, S.G., & Malow, B.A.
tion-deficit/hyperactivity disorder: A case control study. (2008). Melatonin for insomnia in children with autism
BMC Pediatrics, 13, 217. spectrum disorders. Journal of Child Neurology, 23, 482–485.
Adkins, K.W., Molloy, C., Weiss, S.K., Reynolds, A., Goldman, Angriman, M., Caravale, B., Novelli, L., Ferri, R., & Bruni, O.
S.E., Burnette, C., & Malow, B.A. (2012). Effects of a (2015). Sleep in children with neurodevelopmental disabil-
standardized pamphlet on insomnia in children with ities. Neuropediatrics, 46, 199–210.
autism spectrum disorders. Pediatrics, 130(Suppl 2), Appleton, R.E., Jones, A.P., Gamble, C., Williamson, P.R.,
S139–S144. Wiggs, L., Montgomery, P., . . . & Gringras, P. (2012). The use

© 2017 Association for Child and Adolescent Mental Health.


Treatment of insomnia in NDD 17

of MElatonin in children with neurodevelopmental disorders spectrum disorders: A naturalistic study. Journal of Devel-
and impaired sleep: A randomised, double-blind, placebo- opmental and Behavioral Pediatrics: JDBP, 29, 106–116.
controlled, parallel study (MENDS). Health Technology Chow, M., & Cao, M. (2016). The hypocretin/orexin system in
Assessment (Winchester, England), 16, i–239. sleep disorders: Preclinical insights and clinical progress.
Arens, R., Wright, B., Elliott, J., Zhao, H., Wang, P.P., Brown, Nature and Science of Sleep, 8, 81–86.
L.W., . . . & Kaplan, P. (1998). Periodic limb movement in Cortese, S., Brown, T.E., Corkum, P., Gruber, R., O’Brien,
sleep in children with Williams syndrome. The Journal of L.M., Stein, M., . . . & Owens, J. (2013). Assessment and
Pediatrics, 133, 670–674. management of sleep problems in youths with attention-
Asano, M., Ishitobi, M., Kosaka, H., Hiratani, M., & Wada, Y. deficit/hyperactivity disorder. Journal of the American
(2014). Ramelteon monotherapy for insomnia and impulsive Academy of Child and Adolescent Psychiatry, 52, 784–796.
behavior in high-functioning autistic disorder. Journal of Cortese, S., Faraone, S.V., Konofal, E., & Lecendreux, M.
Clinical Psychopharmacology, 34, 402–403. (2009). Sleep in children with attention-deficit/hyperactivity
Baker, A.M., Johnson, D.G., Levisky, J.A., Hearn, W.L., Moore, disorder: Meta-analysis of subjective and objective studies.
K.A., Levine, B., & Nelson, S.J. (2003). Fatal diphenhy- Journal of the American Academy of Child and Adolescent
dramine intoxication in infants. Journal of Forensic Psychiatry, 48, 894–908.
Sciences, 48, 425–428. Cortese, S., Konofal, E., Dalla Bernardina, B., Mouren, M.-C.,
Bennett, T., Bray, D., & Neville, M.W. (2014). Suvorexant, a & Lecendreux, M. (2009). Sleep disturbances and serum
dual orexin receptor antagonist for the management of ferritin levels in children with attention-deficit/hyperactivity
insomnia. P & T: A Peer-Reviewed Journal for Formulary disorder. European Child & Adolescent Psychiatry, 18, 393–
Management, 39, 264–266. 399.
Biban, P., Baraldi, E., Pettennazzo, A., Filippone, M., & Cortesi, F., Giannotti, F., Sebastiani, T., Panunzi, S., & Valente,
Zacchello, F. (1993). Adverse effect of chloral hydrate in D. (2012). Controlled-release melatonin, singly and combined
two young children with obstructive sleep apnea. Pediatrics, with cognitive behavioural therapy, for persistent insomnia in
92, 461–463. children with autism spectrum disorders: A randomized
Blumer, J.L., Findling, R.L., Shih, W.J., Soubrane, C., & Reed, placebo-controlled trial. Journal of Sleep Research, 21, 700–
M.D. (2009). Controlled clinical trial of zolpidem for the 709.
treatment of insomnia associated with attention-deficit/ De Leersnyder, H., de Blois, M.C., Vekemans, M., Sidi, D.,
hyperactivity disorder in children 6 to 17 years of age. Villain, E., Kindermans, C., & Munnich, A. (2001). beta(1)-
Pediatrics, 123, e770–e776. adrenergic antagonists improve sleep and behavioural dis-
Bock, D.E., Roach-Fox, E., Seabrook, J.A., Rieder, M.J., & turbances in a circadian disorder. Smith-Magenis syndrome.
Matsui, D. (2016). Sleep-promoting medications in children: Journal of Medical Genetics, 38, 586–590.
Physician prescribing habits in Southwestern Ontario, Delbarre, B., & Schmitt, H. (1971). Sedative effects of alpha-
Canada. Sleep Medicine, 17, 52–56. sympathomimetic drugs and their antagonism by adrenergic
Bossini, L., Coluccia, A., Casolaro, I., Benbow, J., Amodeo, G., and cholinergic blocking drugs. European Journal of Phar-
De Giorgi, R., & Fagiolini, A. (2015). Off-label trazodone macology, 13, 356–363.
prescription: Evidence. Benefits and Risks. Current Pharma- Dinndorf, P.A., McCabe, M.A., & Frierdich, S. (1998). Risk of
ceutical Design, 21, 3343–3351. abuse of diphenhydramine in children and adolescents
Braam, W., Didden, R., Smits, M.G., & Curfs, L.M.G. (2008). with chronic illnesses. The Journal of Pediatrics, 133, 293–
Melatonin for chronic insomnia in Angelman syndrome: A 295.
randomized placebo-controlled trial. Journal of Child Neu- Dosman, C.F., Brian, J.A., Drmic, I.E., Senthilselvan, A.,
rology, 23, 649–654. Harford, M.M., Smith, R.W., . . . & Roberts, S.W. (2007).
Braam, W., Keijzer, H., Struijker Boudier, H., Didden, R., Children with autism: Effect of iron supplementation on
Smits, M., & Curfs, L. (2013). CYP1A2 polymorphisms in sleep and ferritin. Pediatric Neurology, 36, 152–158.
slow melatonin metabolisers: A possible relationship with Efron, D., Lycett, K., & Sciberras, E. (2014). Use of sleep
autism spectrum disorder? Journal of Intellectual Disability medication in children with ADHD. Sleep Medicine, 15, 472–
Research: JIDR, 57, 993–1000. 475.
Braam, W., Smits, M.G., Didden, R., Korzilius, H., Van France, K.G., Blampied, N.M., & Wilkinson, P. (1999). A
Geijlswijk, I.M., & Curfs, L.M.G. (2009). Exogenous mela- multiple-baseline, double-blind evaluation of the effects
tonin for sleep problems in individuals with intellectual of trimeprazine tartrate on infant sleep disturbance.
disability: A meta-analysis. Developmental Medicine and Experimental and Clinical Psychopharmacology, 7, 502–
Child Neurology, 51, 340–349. 513.
Braam, W., van Geijlswijk, I., Keijzer, H., Smits, M.G., Didden, Frazier, T.W., Krishna, J., Klingemier, E., Beukemann, M.,
R., & Curfs, L.M.G. (2010). Loss of response to melatonin Nawabit, R., & Ibrahim, S. (2017). A randomized, crossover
treatment is associated with slow melatonin metabolism. trial of a novel sound-to-sleep mattress technology in
Journal of Intellectual Disability Research: JIDR, 54, 547– children with autism and sleep difficulties. Journal of
555. Clinical Sleep Medicine: JCSM: Official Publication of the
Bruni, O., Alonso-Alconada, D., Besag, F., Biran, V., Braam, American Academy of Sleep Medicine, 13, 95–104.
W., Cortese, S., . . . & Curatolo, P. (2015). Current role of Giannaccini, G., Masala, I., Palego, L., Betti, L., Pacciardi, B.,
melatonin in pediatric neurology: Clinical recommenda- Palagini, L., . . . & Mauri, M. (2016). Melatonin and pro-
tions. European Journal of Paediatric Neurology: EJPN: hypnotic effectiveness of the antidepressant Trazodone: A
Official Journal of the European Paediatric Neurology Society. preliminary evaluation in insomniac mood-disorder
19, 122–133. patients. Clinical Biochemistry, 49, 1152–1158.
Bruni, O., Violani, C., Luchetti, A., Miano, S., Verrillo, E., & Di Glaze, D.G. (2004). Childhood insomnia: Why Chris can’t
Donatella Valente, C. (2004). The sleep knowledge of pedi- sleep. Pediatric Clinics of North America, 51, 33–50.
atricians and child neuropsychiatrists. Sleep and Hypnosis: Goldman, S.E., Adkins, K.W., Calcutt, M.W., Carter, M.D.,
A Journal of Clinical Neuroscience and Psychopathology, 6, Goodpaster, R.L., Wang, L., . . . & Malow, B.A. (2014).
130–138. Melatonin in children with autism spectrum disorders:
Capone, G.T., Goyal, P., Grados, M., Smith, B., & Kammann, Endogenous and pharmacokinetic profiles in relation to
H. (2008). Risperidone use in children with Down syndrome, sleep. Journal of Autism and Developmental Disorders, 44,
severe intellectual disability, and comorbid autistic 2525–2535.

© 2017 Association for Child and Adolescent Mental Health.


18 Oliviero Bruni et al.

Goldman, S.E., Bichell, T.J., Surdyka, K., & Malow, B.A. Kennaway, D.J. (2000). Melatonin and development: Physiol-
(2012). Sleep in children and adolescents with Angelman ogy and pharmacology. Seminars in Perinatology, 24, 258–
syndrome: Association with parent sleep and stress. Journal 266.
of Intellectual Disability Research: JIDR, 56, 600–608. Kishi, T., Matsunaga, S., & Iwata, N. (2015). Suvorexant for
Golubchik, P., Sever, J., & Weizman, A. (2011). Low-dose primary insomnia: A systematic review and meta-analysis of
quetiapine for adolescents with autistic spectrum disorder randomized placebo-controlled trials. PLoS ONE, 10,
and aggressive behavior: Open-label trial. Clinical Neu- e0136910.
ropharmacology, 34, 216–219. Kordas, K., Siegel, E.H., Olney, D.K., Katz, J., Tielsch, J.M.,
Gominak, S.C., & Stumpf, W.E. (2012). The world epidemic of Chwaya, H.M., & Stoltzfus, R.J. (2008). Maternal reports of
sleep disorders is linked to vitamin D deficiency. Medical sleep in 6-18 month-old infants from Nepal and Zanzibar:
Hypotheses, 79, 132–135. Association with iron deficiency anemia and stunting. Early
Grigg-Damberger, M., & Ralls, F. (2013). Treatment strategies Human Development, 84, 389–398.
for complex behavioral insomnia in children with neurode- Kordas, K., Siegel, E.H., Olney, D.K., Katz, J., Tielsch, J.M.,
velopmental disorders. Current Opinion in Pulmonary Med- Kariger, P.K., & Stoltzfus, R.J. (2009). The effects of
icine, 19, 616–625. iron and/or zinc supplementation on maternal reports of
Gringras, P. (2008). When to use drugs to help sleep. Archives sleep in infants from Nepal and Zanzibar. Journal of Devel-
of Disease in Childhood, 93, 976–981. opmental and Behavioral Pediatrics: JDBP, 30, 131–139.
Gringras, P., Gamble, C., Jones, A. P., Wiggs, L., Williamson, P. Kumar, A., Chanana, P., & Choudhary, S. (2016). Emerging
R., Sutcliffe, A., . . . & MENDS Study Group. (2012). Mela- role of orexin antagonists in insomnia therapeutics: An
tonin for sleep problems in children with neurodevelopmen- update on SORAs and DORAs. Pharmacological Reports: PR,
tal disorders: Randomised double masked placebo 68, 231–242.
controlled trial. BMJ (Clinical Research Ed.), 345, e6664. Lane, R., Kessler, R., Buckley, A.W., Rodriguez, A., Farmer, C.,
Gringras, P., Green, D., Wright, B., Rush, C., Sparrowhawk, Thurm, A., . . . & Felt, B. (2015). Evaluation of periodic limb
M., Pratt, K., & Wiggs, L. (2014). Weighted blankets and movements in sleep and iron status in children with autism.
sleep in autistic children–a randomized controlled trial. Pediatric Neurology, 53, 343–349.
Pediatrics, 134, 298–306. Lieberman, J.A., Kane, J.M., & Reife, R. (1985). Neuromuscu-
Hartmann, E., Lindsley, J.G., & Spinweber, C. (1983). Chronic lar effects of monoamine oxidase inhibitors. Journal of
insomnia: Effects of tryptophan, flurazepam, secobarbital, Clinical Psychopharmacology, 5, 221–228.
and placebo. Psychopharmacology (Berl), 80, 138–142. Liskow, B., & Pikalov, A. (2004). Zaleplon overdose associated
Hartmann, E., & Spinweber, C.L. (1979). Sleep induced by L- with sleepwalking and complex behavior. Journal of the
tryptophan. Effect of dosages within the normal dietary American Academy of Child and Adolescent Psychiatry, 43,
intake. The Journal of Nervous and Mental Disease, 167, 927–928.
497–499. Magera, B.E., Betlach, C.J., Sweatt, A.P., & Derrick, C.W.
Hartz, I., Furu, K., Bratlid, T., Handal, M., & Skurtveit, S. (1981). Hydroxyzine intoxication in a 13-month-old child.
(2012). Hypnotic drug use among 0-17 year olds during Pediatrics, 67, 280–283.
2004-2011: A nationwide prescription database study. Malow, B.A., Katz, T., Reynolds, A.M., Shui, A., Carno, M.,
Scandinavian Journal of Public Health, 40, 704–711. Connolly, H.V., . . . & Bennett, A.E. (2016). Sleep difficul-
Heussler, H., Chan, P., Price, A.M.H., Waters, K., Davey, M.J., ties and medications in children with autism spectrum
& Hiscock, H. (2013). Pharmacological and non-pharmaco- disorders: A registry study. Pediatrics, 137(Suppl 2), S98–
logical management of sleep disturbance in children: An S104.
Australian Paediatric Research Network survey. Sleep Med- Masi, G., Cosenza, A., Millepiedi, S., Muratori, F., Pari, C., &
icine, 14, 189–194. Salvadori, F. (2009). Aripiprazole monotherapy in chil-
Higgins, J.P.T., & Green, S. (2011). Cochrane Handbook for dren and young adolescents with pervasive developmen-
Systematic Reviews of Interventions Version 5.1.0. The tal disorders: A retrospective study. CNS Drugs, 23, 511–
Cochrane Collaboration. 521.
Hollway, J.A., & Aman, M.G. (2011). Pharmacological treat- McArthur, A.J., & Budden, S.S. (1998). Sleep dysfunction
ment of sleep disturbance in developmental disabilities: A in Rett syndrome: A trial of exogenous melatonin treat-
review of the literature. Research in Developmental Disabil- ment. Developmental Medicine and Child Neurology, 40,
ities, 32, 939–962. 186–192.
Holmberg, G. (1988). Sedative effects of maprotiline and McCarty, D.E., Chesson, A.L.J., Jain, S.K., & Marino, A.A.
amitriptyline. Acta Psychiatrica Scandinavica, 77, 584–586. (2014). The link between vitamin D metabolism and sleep
Honaker, S.M., & Meltzer, L.J. (2014). Bedtime problems and medicine. Sleep Medicine Reviews, 18, 311–319.
night wakings in young children: An update of the evidence. Meltzer, L.J., & Mindell, J.A. (2014). Systematic review and meta-
Paediatric Respiratory Reviews, 15, 333–339. analysis of behavioral interventions for pediatric insomnia.
Hudson, C., Hudson, S.P., Hecht, T., & MacKenzie, J. (2005). Journal of Pediatric Psychology, 39, 932–948.
Protein source tryptophan versus pharmaceutical grade Meltzer, L.J., Mindell, J.A., Owens, J.A., & Byars, K.C. (2007).
tryptophan as an efficacious treatment for chronic insom- Use of sleep medications in hospitalized pediatric patients.
nia. Nutritional Neuroscience, 8, 121–127. Pediatrics, 119, 1047–1055.
Imeri, L., Mancia, M., Bianchi, S., & Opp, M.R. (2000). 5- Meolie, A.L., Rosen, C., Kristo, D., Kohrman, M., Gooneratne,
Hydroxytryptophan, but not L-tryptophan, alters sleep and N., Aguillard, R.N., . . . & Mahowald, M. (2005). Oral nonpre-
brain temperature in rats. Neuroscience, 95, 445–452. scription treatment for insomnia: An evaluation of products
Ingrassia, A., & Turk, J. (2005). The use of clonidine for severe with limited evidence. Journal of Clinical Sleep Medicine:
and intractable sleep problems in children with neurodevel- JCSM: Official Publication of the American Academy of Sleep
opmental disorders–a case series. European Child & Adoles- Medicine, 1, 173–187.
cent Psychiatry, 14, 34–40. Merenstein, D., Diener-West, M., Halbower, A.C., Krist, A., &
Jan, J.E., Owens, J.A., Weiss, M.D., Johnson, K.P., Wasdell, Rubin, H.R. (2006). The trial of infant response to diphen-
M.B., Freeman, R.D., & Ipsiroglu, O.S. (2008). Sleep hygiene hydramine: The TIRED study–a randomized, controlled,
for children with neurodevelopmental disabilities. Pedi- patient-oriented trial. Archives of Pediatrics & Adolescent
atrics, 122, 1343–1350. Medicine, 160, 707–712.

© 2017 Association for Child and Adolescent Mental Health.


Mignot, E., Taheri, S., & Nishino, S. (2002). Sleeping with the Quine, L. (1991). Sleep problems in children with mental
hypothalamus: Emerging therapeutic targets for sleep dis- handicap. Journal of Mental Deficiency Research, 35(Pt 4),
orders. Nature Neuroscience, 5(Suppl), 1071–1075. 269–290.
Mindell, J.A., Kuhn, B., Lewin, D.S., Meltzer, L.J., & Sadeh, A. Riemann, D., Voderholzer, U., Cohrs, S., Rodenbeck, A.,
(2006). Behavioral treatment of bedtime problems and night Hajak, G., R€ uther, E., . . . & Berger, M. (2002). Trimipramine
wakings in infants and young children. Sleep, 29, 1263– in primary insomnia: Results of a polysomnographic double-
1276. blind controlled study. Pharmacopsychiatry, 35, 165–174.
Ming, X., Gordon, E., Kang, N., & Wagner, G.C. (2008). Use of Robinson, A.A., & Malow, B.A. (2013). Gabapentin shows
clonidine in children with autism spectrum disorders. Brain promise in treating refractory insomnia in children. Journal
& Development, 30, 454–460. of Child Neurology, 28, 1618–1621.
Miyamoto, A., Fukuda, I., Tanaka, H., Oka, R., Araki, A., & Robinson-Shelton, A., & Malow, B.A. (2016). Sleep Distur-
Cho, K. (2013). Treatment with ramelteon for sleep distur- bances in Neurodevelopmental Disorders. Current Psychia-
bance in severely disabled children and young adults. No to try Reports, 18, 6.
hattatsu. Brain and development, 45, 440–444. Rosenberg, R.P., Hull, S.G., Lankford, D.A., Mayleben, D.W.,
Mohammadi, M.R., Mostafavi, S.A., Keshavarz, S.A., Eshra- Seiden, D.J., Furey, S.A., . . . & Roth, T. (2014).
ghian, M.R., Hosseinzadeh, P., Hosseinzadeh-Attar, M.J., . . . A randomized, double-blind, single-dose, placebo-con-
& Akhondzadeh, S. (2012). Melatonin effects in methylphe- trolled, multicenter, polysomnographic study of gabapentin
nidate treated children with attention deficit hyperactivity in transient insomnia induced by sleep phase advance.
disorder: A randomized double blind clinical trial. Iranian Journal of Clinical Sleep Medicine : JCSM : Official Publica-
Journal of Psychiatry, 7, 87–92. tion of the American Academy of Sleep Medicine, 10, 1093–
Montanari, G., Schiaulini, P., Covre, A., Steffan, A., & Furla- 1100.
nut, M. (1992). Niaprazine vs chlordesmethyldiazepam in Russo, R.M., Gururaj, V.J., & Allen, J.E. (1976). The effective-
sleep disturbances in pediatric outpatients. Pharmacological ness of diphenhydramine HCI in pediatric sleep disorders.
Research, 25(Suppl 1), 83–84. Journal of Clinical Pharmacology, 16, 284–288.
Monti, J.M. (2011). Serotonin control of sleep-wake behavior. Sangal, R.B., Blumer, J.L., Lankford, D.A., Grinnell, T.A., &
Sleep Medicine Reviews, 15, 269–281. Huang, H. (2014). Eszopiclone for insomnia associated with
Morairty, S.R., Revel, F.G., Malherbe, P., Moreau, J.-L., attention-deficit/hyperactivity disorder. Pediatrics, 134,
Valladao, D., Wettstein, J.G., . . . & Borroni, E. (2012). Dual e1095–e1103.
hypocretin receptor antagonism is more effective for sleep Schneider-Helmert, D., & Spinweber, C.L. (1986). Evaluation
promotion than antagonism of either receptor alone. PLoS of L-tryptophan for treatment of insomnia: A review. Psy-
ONE, 7, e39131. chopharmacology (Berl), 89, 1–7.
Nguyen, M., Tharani, S., Rahmani, M., & Shapiro, M. (2014). A Schnoes, C.J., Kuhn, B.R., Workman, E.F., & Ellis, C.R.
review of the use of clonidine as a sleep aid in the child and (2006). Pediatric prescribing practices for clonidine and
adolescent population. Clinical Pediatrics, 53, 211–216. other pharmacologic agents for children with sleep distur-
Okawa, M.S.H. (1987). Sleep disorders in mentally retarded bance. Clinical Pediatrics, 45, 229–238.
and brain- impaired children. In C. Guilleminault (Ed.), Sezer, T., & Alehan, F. (2013). Chloral hydrate versus hydrox-
Sleep and its disorders in children (pp. 269–290). New yzine HCL for sedation prior to pediatric sleep EEG record-
York: Raven Press. ing. The International Journal of Neuroscience, 123, 719–
Ottaviano, S., Giannotti, F., & Cortesi, F. (1991). The effect of 723.
niaprazine on some common sleep disorders in children. A Sheldon, S.H. (2001). Insomnia in children. Current Treatment
double-blind clinical trial by means of continuous home- Options in Neurology, 3, 37–50.
videorecorded sleep. Child’s Nervous System: ChNS: Official Smits, M.G., van Stel, H.F., van der Heijden, K., Meijer, A.M.,
Journal of the International Society for Pediatric Neuro- Coenen, A.M.L., & Kerkhof, G.A. (2003). Melatonin improves
surgery, 7, 332–335. health status and sleep in children with idiopathic chronic
Owens, J.A. (2011). Update in pediatric sleep medicine. sleep-onset insomnia: A randomized placebo-controlled
Current Opinion in Pulmonary Medicine, 17, 425–430. trial. Journal of the American Academy of Child and Adoles-
Owens, J.A., Rosen, C.L., Mindell, J.A., & Kirchner, H.L. cent Psychiatry, 42, 1286–1293.
(2010). Use of pharmacotherapy for insomnia in child Spiller, H.A., Klein-Schwartz, W., Colvin, J.M., Villalobos, D.,
psychiatry practice: A national survey. Sleep Medicine, 11, Johnson, P.B., & Anderson, D.L. (2005). Toxic clonidine
692–700. ingestion in children. The Journal of Pediatrics, 146, 263–
Pelayo, R., & Dubik, M. (2008). Pediatric sleep pharmacology. 266.
Seminars in Pediatric Neurology, 15, 79–90. Spinweber, C.L. (1986). L-tryptophan administered to chronic
Pelayo, R., & Yuen, K. (2012). Pediatric sleep pharmacology. sleep-onset insomniacs: Late-appearing reduction of sleep
Child and Adolescent Psychiatric Clinics of North America, latency. Psychopharmacology (Berl), 90, 151–155.
21, 861–883. Stigler, K.A., Posey, D.J., & McDougle, C.J. (2006). Ramelteon for
Phillips, L., & Appleton, R.E. (2004). Systematic review of insomnia in two youths with autistic disorder. Journal of Child
melatonin treatment in children with neurodevelopmental and Adolescent Psychopharmacology, 16, 631–636.
disabilities and sleep impairment. Developmental Medicine Stores, R., & Stores, G. (2004). Evaluation of brief group-
and Child Neurology, 46, 771–775. administered instruction for parents to prevent or minimize
Piazza, C.C., Fisher, W., & Moser, H. (1991). Behavioral sleep problems in young children with down syndrome.
treatment of sleep dysfunction in patients with the Rett Journal of Applied Research in Intellectual Disabilities, 17,
syndrome. Brain & Development, 13, 232–237. 61–70.
Posey, D.J., Guenin, K.D., Kohn, A.E., Swiezy, N.B., & Stuttard, L., Beresford, B., Clarke, S., Beecham, J., & Curtis, J.
McDougle, C.J. (2001). A naturalistic open-label study of (2015). A preliminary investigation into the effectiveness of a
mirtazapine in autistic and other pervasive developmental group-delivered sleep management intervention for parents of
disorders. Journal of Child and Adolescent Psychopharma- children with intellectual disabilities. Journal of Intellectual
cology, 11, 267–277. Disabilities: JOID, 19, 342–355.
Pranzatelli, M.R., Tate, E.D., Dukart, W.S., Flint, M.J., Hoff- Tassniyom, K., Paholpak, S., Tassniyom, S., & Kiewyoo, J.
man, M.T., & Oksa, A.E. (2005). Sleep disturbance and rage (2010). Quetiapine for primary insomnia: A double blind,
attacks in opsoclonus-myoclonus syndrome: Response to randomized controlled trial. Journal of the Medical Associa-
trazodone. The Journal of Pediatrics, 147, 372–378. tion of Thailand =Chotmaihet Thangphaet, 93, 729–734.
20 Oliviero Bruni et al.

Thirumalai, S.S., Shubin, R.A., & Robinson, R. (2002). Rapid treatment of delayed sleep phase syndrome and impaired
eye movement sleep behavior disorder in children with sleep maintenance in children with neurodevelopmental
autism. Journal of Child Neurology, 17, 173–178. disabilities. Journal of Pineal Research, 44, 57–64.
Van de Wouw, E., Evenhuis, H.M., & Echteld, M.A. (2012). Weiss, M.D., Wasdell, M.B., Bomben, M.M., Rea, K.J., &
Prevalence, associated factors and treatment of sleep prob- Freeman, R.D. (2006). Sleep hygiene and melatonin treat-
lems in adults with intellectual disability: A systematic ment for children and adolescents with ADHD and initial
review. Research in Developmental Disabilities, 33, 1310– insomnia. Journal of the American Academy of Child and
1332. Adolescent Psychiatry, 45, 512–519.
Van der Heijden, K.B., Smits, M.G., Van Someren, E.J.W., & Wichniak, A., Wierzbicka, A., & Jernajczyk, W. (2012). Sleep
Gunning, W.B. (2005). Idiopathic chronic sleep onset and antidepressant treatment. Current Pharmaceutical
insomnia in attention-deficit/hyperactivity disorder: A cir- Design, 18, 5802–5817.
cadian rhythm sleep disorder. Chronobiology International, Wiggs, L., & France, K. (2000). Behavioural treatments for
22, 559–570. sleep problems in children and adolescents with physical
Van der Heijden, K.B., Smits, M.G., Van Someren, E.J.W., illness, psychological problems or intellectual disabilities.
Ridderinkhof, K.R., & Gunning, W.B. (2007). Effect of Sleep Medicine Reviews, 4, 299–314.
melatonin on sleep, behavior, and cognition in ADHD Younus, M., & Labellarte, M.J. (2002). Insomnia in children:
and chronic sleep-onset insomnia. Journal of the American When are hypnotics indicated? Paediatric Drugs, 4, 391–
Academy of Child and Adolescent Psychiatry, 46, 233–241. 403.
Van Geijlswijk, I.M., van der Heijden, K.B., Egberts, A.C.G., Youssef, J., Singh, K., Huntington, N., Becker, R., & Kothare,
Korzilius, H.P.L.M., & Smits, M.G. (2010). Dose finding of S.V. (2013). Relationship of serum ferritin levels to sleep
melatonin for chronic idiopathic childhood sleep onset fragmentation and periodic limb movements of sleep on
insomnia: An RCT. Psychopharmacology (Berl), 212, 379– polysomnography in autism spectrum disorders. Pediatric
391. Neurology, 49, 274–278.
Ware, J.C., Brown, F.W., Moorad, P.J., Pittard, J.T., & Cobert, Zell-Kanter, M., Toerne, T.S., Spiegel, K., & Negrusz, A. (2000).
B. (1989). Effects on sleep: A double-blind study comparing Doxepin toxicity in a child following topical administration.
trimipramine to imipramine in depressed insomniac The Annals of Pharmacotherapy, 34, 328–329.
patients. Sleep, 12, 537–549. Zisapel, N. (2012). Drugs for insomnia. Expert Opinion on
Wasdell, M.B., Jan, J.E., Bomben, M.M., Freeman, R.D., Emerging Drugs, 17, 299–317.
Rietveld, W.J., Tai, J., & Weiss, M.D. (2008). A randomized,
placebo-controlled trial of controlled release melatonin Accepted for publication: 26 July 2017

© 2017 Association for Child and Adolescent Mental Health.

You might also like