https://doi.org/10.1177/0269881119855343
Journal of Psychopharmacology
2019, Vol. 33(8) 923 –947
©
The Author(s) 2019
Article reuse guidelines:
sagepub.com/journals-permissions
DOI: 10.1177/0269881119855343
journals.sagepub.com/home/jop
British Association for Psychopharmacology
consensus statement on evidence-based
treatment of insomnia, parasomnias and
circadian rhythm disorders: An update
Sue Wilson
1
, Kirstie Anderson
2
, David Baldwin
3
, Derk-Jan Dijk
4
,
Audrey Espie
5
, Colin Espie
6
, Paul Gringras
7
, Andrew Krystal
8
,
David Nutt
1
, Hugh Selsick
9
and Ann Sharpley
10
Abstract
This British Association for Psychopharmacology guideline replaces the original version published in 2010, and contains updated information and
recommendations. A consensus meeting was held in London in October 2017 attended by recognised experts and advocates in the field. They were
asked to provide a review of the literature and identification of the standard of evidence in their area, with an emphasis on meta-analyses, systematic
reviews and randomised controlled trials where available, plus updates on current clinical practice. Each presentation was followed by discussion,
aiming to reach consensus where the evidence and/or clinical experience was considered adequate, or otherwise to flag the area as a direction for
future research. A draft of the proceedings was circulated to all speakers for comments, which were incorporated into the final statement.
Keywords
Sleep disorders, insomnia, circadian rhythm disorders, parasomnias, guidelines, evidence-based treatment
855343
JOP0010.1177/0269881119855343Journal of PsychopharmacologyWilson et al.
research-article2019
BAP Guidelines
Table of Contents
Introduction 2
Method 2
Insomnia 2
Scope of the guidelines 2
Table 1 Levels of evidence 3
Epidemiology of insomnia 3
Table 2 Insomnia: diagnostic criteria 4
Diagnosis of insomnia 4
Figure 1 Diagnosis of insomnia 5
Comorbidity 5
Costs and consequences of insomnia 5
Figure 2 The ideal sleeping pill 6
Recommendation 6
Psychological treatment of insomnia 7
Underpinning principles – cognitive
behavioural therapy 7
Recommendation 7
Drug treatments for insomnia 7
Underpinning principles - pharmacology 8
Underpinning principles – pharmacokinetics 8
Figure 3 treatment of insomnia 9
Tolerance, dependence and withdrawal 9
Pharmacological treatment of insomnia 10
Recommendations 10
Long-term use of sleeping medications 10
Recommendation 11
Using drugs for depression to treat insomnia 11
Recommendations 12
Drugs for psychosis for treatment of insomnia 12
Recommendations 13
Antihistamines (H1 antagonists) 13
Recommendations 13
Circadian rhythm disorders 13
Diagnosis of circadian rhythm disorders 14
Treating circadian rhythm disorders 14
Recommendations 14
Parasomnias 15
Diagnosis of parasomnias 15
Treatment of parasomnias 15
Special populations 16
Sleep disorders in women: effects of
menopause and pregnancy 16
Menopause 16
Recommendations 16
Pregnancy 16
Recommendations 16
Treatment of insomnia in older adults 17
Recommendation 17
Sleep problems in children 17
Recommendations 18
Sleep disturbance in adults with intellectual disability 18
Assessment 18
Treatment considerations 18
Recommendations 19
References 19
Appendix 1 25
924 Journal of Psychopharmacology 33(8)
Introduction
Sleep disorders are common in the general population, and even
more so in clinical practice, yet are relatively poorly understood
by doctors and other health care practitioners. These British
Association for Psychopharmacology (BAP) guidelines address
this problem by providing an accessible yet up-to-date and evi-
dence-based outline of the major issues, especially those relating
to reliable diagnosis and appropriate treatment. We limited our-
selves to discussion of sleep problems that are not regarded as
being secondary to sleep disordered breathing; National Institute
of Clinical Excellence (NICE) guidelines for this are summarised
on the NICE website and an updated guideline will be available
in 2020; a comprehensive toolkit is available at the British Sleep
Society website, http://www.sleepsociety.org.uk. We also did not
consider certain sleep disorders for which sets of guidelines
already exist, such as narcolepsy (Billiard et al., 2006) and rest-
less legs syndrome (Picchietti et al., 2015). Thus, the main scope
of this document is to address insomnia, circadian rhythm disor-
ders (CRDs) and the more common parasomnias which are likely
to present to primary care physicians and psychiatrists.
The BAP is an association of psychiatrists, psychopharma-
cologists and preclinical scientists who are interested in the broad
field of drugs and the brain. BAP is the largest national organisa-
tion of its kind worldwide, and publishes the Journal of
Psychopharmacology. The association started publishing con-
sensus statements more than two decades ago, and the first BAP
guidelines on depression were considered a landmark publication
when they appeared in 1993 (Montgomery et al., 1993). There
are now guidelines for the treatment and management of most of
the disorders encountered in psychiatry; all guidelines are avail-
able to download from the BAP website (http://www.bap.org.uk).
Method
This British Association for Psychopharmacology guideline
replaces the original version published in 2010, (Wilson et al
2010) and contains updated information and recommendations. A
consensus meeting was held in London in October 2017, attended
by recognised experts and advocates in the field. They were
asked to provide a review of the literature and identification of
the standard of evidence in their area, with an emphasis on meta-
analyses, systematic reviews and randomised controlled trials
(RCTs) where available, plus updates on current clinical practice.
Each presentation was followed by discussion, aiming to reach
consensus where the evidence and/or clinical experience was
considered adequate, or otherwise to flag the area as a direction
for future research. The previous consensus statement was then
updated with the new evidence and references.
Categories of evidence for causal relationships, observational
relationships and strength of recommendations are given in
Table 1 and are taken from (Shekelle et al., 1999). The strength
of recommendation reflects not only the quality of the evidence,
but also the importance of the area under study. For example, it is
possible to have methodologically sound (category I) evidence
about an area of practice that is clinically irrelevant, or has such
a small effect that it is of little practical importance and therefore
attracts a lower strength of recommendation. However, more
commonly, it has been necessary to extrapolate from the available
evidence leading to weaker levels of recommendation (B, C or
D) based upon category I evidence statements. The costs of the
meeting were defrayed by BAP. All speakers completed conflict
of interest statements that are held at the BAP office according to
BAP policy.
Insomnia
Scope of the guidelines
Our intention is to provide an updated statement to guide clini-
cians who manage patients in primary or secondary medical care.
There have been three sets of guidelines for the treatment of
insomnia since the previous BAP consensus (Qaseem et al.,
2016; Riemann et al., 2017; Sateia et al., 2017). The first set of
guidelines concerns adults with insomnia and includes insomnia
comorbid with other disorders such as depression; the second set
addresses primary insomnia without comorbidity; the third set
covers all adults with chronic insomnia disorder. These sets were
discussed by the expert group and where appropriate some ele-
ments were incorporated in the present consensus.
Since the publication of the 2010 BAP guideline, there has
been an important shift in thinking about the diagnosis and clas-
sification of insomnia. The historical perspective that insomnia
could be either ‘primary’ or ‘secondary’, is no longer regarded as
valid or evidence-based. Rather, the expanding literature has led
the American Psychiatric Association (APA) (Diagnostic and
Statistical Manual of Mental Disorders (DSM)-5) and the
American Academy of Sleep Medicine (AASM) (International
Classification of Sleep Disorders (ICSD)-3) to recommend that
chronic insomnia disorder (APA) should be considered as a dis-
order in its own right.
This means that insomnia disorder should be diagnosed
whenever insomnia diagnostic criteria are met, irrespective of
any concurrent physical disorder or mental disorder; and, impor-
tantly, also irrespective of any other concurrent sleep disorder. It
is anticipated that International Classification of Diseases 11
th
Revision (ICD-11) will reflect the same conclusions when it is
presented at the World Health Assembly for adoption by member
states in 2019.
1
Centre for Psychiatry, Imperial College London, London, UK
2
Regional Sleep Service, Freeman Hospital, Newcastle Upon Tyne, UK
3
Clinical and Experimental Sciences, University of Southampton,
Southampton, UK
4
Sleep Research Centre, University of Surrey, Guildford, UK
5
Psychology Department, NHS Fife, Dunfermline, UK
6
Nuffield Department of Clinical Neurosciences, University of Oxford,
Oxford, UK
7
Guy’s and St Thomas’ NHS Foundation Trust, London, UK
8
Psychiatry and Behavioral Science, University of California, San
Francisco, CA, USA
9
Royal London Hospital for Integrated Medicine, London, UK
10
Department of Psychiatry, University of Oxford, Oxford, UK
Corresponding author:
Sue Wilson, Centre for Psychiatry, Division of Brain Sciences, Imperial
College, Burlington Danes Building, Hammersmith Hospital campus,
160 Du Cane Road, London, W12 0NN, UK.
Wilson et al. 925
The complex relationship between insomnia and psychiat-
ric disorders has been the subject of much recent research. It is
increasingly recognised that sleep plays a central role in the
regulation of emotion and emotion processing (Palmer and
Alfano, 2017; Tempesta et al., 2018). Therefore, it is not sur-
prising to see a bidirectional relationship between insomnia
and mental disorder. There is considerable evidence that pre-
existing insomnia confers risk for the development of (or
relapse into) depression). This makes it all the more important
to consider the time-course of how insomnia and other psychi-
atric symptoms develop and resolve (Sánchez-Ortuño and
Edinger, 2012).
Insomnia often starts with a specific problem, for example
a stressful life event such as the loss of a job or change to a
more demanding one; or through something that changes sleep
patterns, such as the birth of a child or starting shift work. In
some people this acute insomnia persists into a chronic state.
Factors involved in the persistence of insomnia are not fully
established, but include anxiety about sleep, maladaptive sleep
habits and the possibility of an underlying vulnerability in
sleep-regulating mechanisms. Persistence of the precipitating
stressor can also contribute. Some cases of insomnia are pre-
cipitated by, or co-morbid with, other psychiatric disorders
especially anxiety and depression, or by physical illness such
as cancer or arthritis.
The nature of sleep changes with age. Older age is associated
with poorer objectively-measured sleep with shorter sleep time,
diminished sleep efficiency, and more arousals. These changes
may be more marked in men than women according to a very
large study of elderly people living at home in the USA (Sleep
Heart Health Study; Unruh et al., 2008). In the same study, the
association of subjective report of poor sleep with older age was
stronger in women. The higher prevalence of chronic health con-
ditions, including sleep apnoea, in older adults did not explain
changes of sleep parameters with aging and age-sex differences
in these relationships.
There is now greater consensus about how long insomnia
should have been present before it merits intervention. Chronic
insomnia is regarded as established after three months of persis-
tent poor sleep. There is also general agreement that when insom-
nia causes significant personal distress or marked impairment
then some form of treatment is appropriate. The cause of insom-
nia may be known or not, and knowledge of causation is not nec-
essary for a diagnosis.
Epidemiology of insomnia
Studies of prevalence of insomnia in the general population indi-
cate that one third of adults in Western countries experience dif-
ficulty with sleep initiation or maintenance at least once a week
(LeBlanc et al., 2009; Leger and Poursain, 2005; Sateia et al.,
2000), and 6–15% are thought to meet the criteria of insomnia in
that they report sleep disturbance as well as significant daytime
dysfunction (LeBlanc et al., 2009; Sivertsen et al., 2009). One-
year incidence rates have been reported to be 30.7% for insomnia
symptoms and 7.4% for insomnia syndrome. These rates
decreased to 28.8% and 3.9% for those without a prior lifetime
episode of insomnia (LeBlanc et al., 2009). There is much evi-
dence that insomnia can be a long-term disorder. In one large UK
study, about three-quarters of patients reported symptoms lasting
at least a year (Morphy et al., 2007) and, in a population-based
three-year longitudinal study, 46% of subjects who had insomnia
at baseline still had it at the three-year time point. The course of
insomnia was more likely to be persistent in those with more
severe insomnia at baseline and in women and older adults
(Morin et al., 2009). Two studies have described an increase of
insomnia over time: in the UK, insomnia diagnosis increased
from 3.1% to 5.8% (National Psychiatric Morbidity Surveys
1993–2007; Calem et al., 2012); and in Norway, insomnia diag-
nosis increased from 11.9% to 15.5% between two surveys in
2000–2010 (Pallesen et al., 2014).
Table 1. Levels of evidence.
Category of evidence:
Ia — evidence for meta-analysis of randomised controlled trials.
Ib — evidence from at least one randomised controlled trial.
IIa — evidence from at least one controlled study without randomisation.
IIb — evidence from at least one other type of quasi-experimental study.
III — evidence from non-experimental descriptive studies, such as comparative studies, correlation studies, and case-control studies.
IV — evidence from expert committee reports or opinions or clinical experience of respected authorities, or both.
Strength of recommendation:
A — directly based on category I evidence.
B — directly based on category II evidence or extrapolated recommendation from category I evidence.
C — directly based on category III evidence or extrapolated recommendation from category I or II evidence.
D — directly based on category IV evidence or extrapolated recommendation from category I, II or III evidence.
What is known about prevalence of insomnia:
Estimates of prevalence of insomnia vary according to the
definition used (Ia).
Prevalence of symptoms varies with age, with increase of
nocturnal awakenings but decrease in complaints of non-
restorative sleep as people age (Ib).
Prevalence is between 1.5–2 times higher in women than in
men (Ia).
Insomnia is a long-term disorder; many people have had
insomnia for more than two years (Ib).
Approximately half of all diagnosed insomnia is comorbid with
a psychiatric disorder (Ib).
What is not known:
What is the prevalence of distress about sleep?
What is the significance of duration of symptoms on distress?
926 Journal of Psychopharmacology 33(8)
There is a higher incidence of insomnia in women, and the
incidence increases in men and women as they get older (see
below – special populations). The symptom prevalence changes
with age, so that people aged over 65 years show more sleep
maintenance problems but a decrease in reported daytime prob-
lems compared with younger age groups, with little change in the
prevalence of sleep onset insomnia.
Diagnosis of insomnia
Diagnostic criteria from the APA, AASM and World Health
Organization (WHO) are summarised in Table 2. They agree that
insomnia is a complaint of unsatisfactory sleep, either in terms of
sleep onset, sleep maintenance or early waking. In DSM-5 and
ICSD-3 this complaint must be present three or more nights per
week, for at least three months, and be associated with impair-
ment to day-time functioning or well-being. In this sense insom-
nia can be considered a ‘24-hour disorder, because a complaint
of unsatisfactory sleep without reported functional sequelae
would not meet clinical diagnostic criteria.
Like other disorders and conditions classified within DSM-5,
insomnia is largely a subjectively determined disorder.
Polysomnographic (PSG) and actigraphic studies do indicate that
people with insomnia take longer to fall asleep and have sleep that
is more fragmented than healthy good sleepers. However, these
parameters do not reflect the level of sleep disturbance reported by
people with insomnia; and they do not sample daytime experience.
Moreover, PSG and actigraphy are not indicated for use in insom-
nia – except if other sleep disorders are suspected (Sateia et al.,
2017) – and, in any case, are seldom available in routine care.
Like depression, anxiety or pain, there is no objective test for
insomnia, and in practice it is evaluated clinically. Diagnosis,
therefore, is through appraisal against diagnostic criteria, clinical
observations and the use of validated rating scales.
There are a number of ways in which sleep can be assessed.
The simplest is by asking the patient about their sleep. Are they
having difficulty getting to sleep and/ or staying asleep? Is this
occurring most nights? Is this persistent and affecting how they
feel during the day?
An extension of this interview enquiry is to administer a
clinical rating scale. The Sleep Condition Indicator (SCI) is
one such scale being based on contemporary diagnostic criteria
and has been validated in over 200,000 adults. It also has a
short-form screening version comprising only two items (Espie
et al., 2014, 2018b; Luik et al., 2019; see Appendix 1).
A further step is to provide the patient with a sleep diary. This
allows the assessment of sleep difficulties over time, and gauges
the potential contribution of poor sleep and lifestyle habits to
daytime impairment. Some patients appreciate completing a
diary to capture the nature of their sleep problems, including the
unpredictability of their sleep from night to night. The SCI and
the diary may also be useful to assess treatment-related change.
Finally, it is important to determine if another sleep disorder (see
preliminary questions below), or a physical (such as pain, heart or
metabolic disease), neurological (such as Parkinson’s disease or cer-
ebrovascular disease) or psychiatric (such as depressive illness,
anxiety disorder or substance use disorder) disorder is present along-
side the insomnia. The insomnia problem should be actively treated,
but consideration of the interplay between conditions is good clinical
practice. A diagram illustrating diagnosis is given in Figure 1.
Table 2. Insomnia: diagnostic criteria.
International Classification
of Sleep Disorders (ICSD-3)
and (Sateia etal. 2017)
A
The patient reports (or the patient’s
parent or caregiver reports) marked
concern about, or dissatisfaction
with, sleep comprising one or more
of the following:-difficulty initiating
sleep, difficulty maintaining sleep,
waking up earlier than desired,
resistance in going to bed on the
appropriate schedule,
difficulty sleeping without the
parent or caregiver present.
B
Occurs despite adequate
opportunity and
circumstances for sleep.
C
At least one form of daytime
impairment e.g.
fatigue; mood disturbance;
interpersonal problems; reduced
cognitive function; reduced
performance; daytime sleepiness;
behavioural problems (e.g.
hyperactivity, impulsivity,
aggression); reduced motivation/
initiative; proneness to errors/
accidents.
International Classification
of Diseases (ICD)-10; World
Health Organization (WHO),
1992
Difficulty
- falling asleep,
- maintaining sleep or
- non-refreshing sleep
Three times a week and for
longer than 1 month
Marked personal distress or
interference with personal
functioning in daily living
Diagnostic and Statistical
Manual of Mental Disorders
(DSM-5; insomnia disorder)
Unhappiness with the quality or
quantity of sleep, which can include
trouble falling asleep, staying asleep
or waking up early and being unable
to get back to sleep. The problem
occurs despite ample opportunity to
sleep. The difficulty cannot be better
explained by other physical, mental
or sleep-wake disorders. The problem
cannot be attributed to substance
use or medication.
Three nights a week for at
least 3 months.
The sleep disturbance causes
significant distress or
impairment in functioning,
such as within the individual’s
working or personal life,
behaviourally or emotionally.
Wilson et al. 927
Comorbidity
There is a generally high incidence of sleep disorders in psychi-
atric conditions. The most widely reported are shown below:
Costs and consequences of insomnia
Insomnia is now recognised as reliably associated with mental
health disorders including risk of depression and suicide
(Baglioni and Riemann, 2012; Pigeon et al., 2017), cardiovascu-
lar disease (Khan and Aouad, 2017) and type 2 diabetes
(Cappuccio et al., 2010; Vgontzas et al., 2009). Increased fatigue,
impaired work productivity, reduced quality of life, and relation-
ship dissatisfaction are also common in those with insomnia
(Espie et al., 2012; Kyle et al., 2010; Roth and Ancoli-Israel,
1999). Indeed, such impaired functioning is an important driver
for help-seeking behaviour (Morin et al., 2006).
There is at least a two-fold increased risk of subsequent
depression and anxiety disorder in patients with pre-existing
insomnia (Baglioni and Riemann, 2012). Insomnia has been asso-
ciated with: (a) an increased risk of developing subsequent depres-
sion; (b) an increased duration of established depression; and (c)
relapse following treatment for depression (Riemann, 2009). Poor
sleep quality also seems to correlate with high negative and low
positive emotions, both in clinical and subclinical samples. Good
sleep seems to be associated with high positive emotions, though
not necessarily with low negative emotions (Baglioni et al., 2010).
The strong relationship between insomnia and emotional vul-
nerability has been established for 30 years. The National
Institute of Mental Health Epidemiologic Catchment Area inter-
viewed 7954 adults on two occasions a year apart, and high-
lighted the strong association between sleep disturbance and
subsequent depression. It was found that 14% of those with
insomnia at the first interview had developed a new major depres-
sive episode one year later (Ford and Kamerow, 1989). This
increased risk of developing depression has been confirmed in
numerous investigations: in a survey of 1200 young adults in
Michigan, the odds ratio of new depression was four times
greater in those subjects who had insomnia three years earlier
(Breslau et al., 1996) and of new anxiety disorder the risk was
Figure 1. Diagnosis of insomnia.
Asking about another sleep disorder: preliminary questions:
Are you a very heavy snorer? Does your partner say that you
sometimes stop breathing at night? (obstructive sleep apnoea
syndrome (OSAS)).
When you try to relax in the evening or sleep at night, do you
ever have unpleasant, restless feelings in your legs that can be
relieved by walking or movement? (restless legs syndrome (RLS)).
Do you sometimes fall asleep in the daytime completely without
warning? Do you have collapses or extreme muscle weakness
triggered by emotion, for instance when you’re laughing?
(narcolepsy).
Do you tend to sleep well but just at the ‘wrong times’; and are
these sleeping and waking times regular? (circadian rhythm
sleep disorder (CRD); evidence also from sleep diary).
Do you have unusual or unpleasant experiences or behaviours
associated with your sleep that trouble you or that are
dangerous? (parasomnias).
Major depressive disorder Up to 70% insomnia
Up to 15% hypersomnia
Post-traumatic stress disorder
(PTSD)
Insomnia
Nightmares
Non-REM parasomnia
Schizophrenia CRD
Dementia Insomnia
CRD
Substance abuse Insomnia
Parkinson’s disease, Lewy Body
dementia
REM behaviour disorder
RLS
What is known about detrimental effects of insomnia:
Quality of life is impaired in insomnia (Ia).
There is an increased risk of subsequent first-episode
depression, and of relapse into depression, in those with a pre-
existing chronic insomnia (Ia).
There is an increased risk of type 2 diabetes and hypertension in
insomnia with objectively-measured short sleep duration (II).
‘Presenteeism’ (lack of productivity at work), absenteeism, acci-
dents at work and road accidents are increased in insomnia (II).
What is not known:
What are the potential confounding effects of medication and
comorbid disorders in reports of increased accidents?
To what extent do insomnia treatments rectify risk markers for
emotional, metabolic and cardiovascular disease, or prevent
development of disorders at a clinical level?
928 Journal of Psychopharmacology 33(8)
two-fold greater. In a questionnaire survey of adults in the UK,
there was a three-fold increased risk of new depression and a
two-fold risk of new anxiety disorder if subjects had reported one
sleep problem occurring ‘on most nights’ a year earlier (Morphy
et al., 2007). In a much longer study in Norway, with two surveys
10 years apart (Neckelmann et al., 2007), the risk of having an
anxiety disorder diagnosis at the second time point increased by
about one and a half times if insomnia had been present at the
first time point, and about five times if insomnia was present at
both time points. Doctors in a prospective study who had com-
plained of insomnia whilst studying at medical school in the
1950s and 1960s were twice as likely to have developed depres-
sion at follow-up in the 1990s (Chang et al., 1997).
Insomnia is associated with activation of the hypothalamic-
pituitary-adrenal (HPA) axis, with increased adrenocorticotrophin
(ACTH) and cortisol in most studies (Varkevisser et al., 2005;
Vgontzas et al., 1998; Vgontzas et al., 2001). When the complaint
of insomnia is accompanied by short duration of sleep measured
objectively, there is a 3–5-fold increase in overall risk of hyperten-
sion which is comparable to that seen with other common sleep
disorders, such as sleep disordered breathing (Vgontzas et al.,
2009). In France, Japan and the USA, insomnia patients scored
significantly lower on all eight domains of the SF-36 quality of life
questionnaire, compared with good sleepers (Leger, 2012).
People with a diagnosis of insomnia also have subjective
complaints of poor daytime function. When compared with
matched controls, they show increased subjective sleepiness but
decreased objective sleepiness, due to the fact that they are usu-
ally over-aroused, but feel subjectively tired. Objectively, they
show poorer performance on psychomotor tasks, particularly
those requiring the switching of attention (e.g. frontal/executive
tasks) (Edinger et al., 2008): objectively measured time awake
after sleep onset (WASO) was the best predictor of impaired
daytime performance. Likewise, Altena et al. (Altena et al.,
2008) reported that people with insomnia perform more poorly
on complex cognitive tasks, an effect which normalises follow-
ing cognitive behavioural therapy (CBT) intervention. A meta-
analysis of 24 studies (Fortier-Brochu et al., 2012). comparing
the daytime cognitive performance of people with insomnia and
good sleeper controls found that those with insomnia exhibited
performance impairments of small to moderate magnitude in
working memory, episodic memory and some aspects of execu-
tive functioning. However no significant group differences were
observed for tasks assessing general cognitive function, percep-
tual and psychomotor processes, procedural learning, verbal
functions, different dimensions of attention (alertness, complex
reaction time, speed of information processing, selective atten-
tion, sustained attention/vigilance) and some aspects of execu-
tive functioning (verbal fluency, cognitive flexibility).
The economic burden of insomnia is high, with overall costs
thought to exceed US$100 billion per year in the USA (Wickwire
et al., 2016). In Europe, the economic costs of insufficient sleep,
including disruption from insomnia and other sleep difficulties
were modelled across five countries; in the UK costs were esti-
mated at 1.86% of Gross Domestic Product or US$50 billion to
the economy (Hafner et al., 2017). Costs are both direct (pre-
scription costs, appointments and inpatient care) and indirect
(lost workplace productivity/presenteeism, absenteeism, and
increased risk of traffic and workplace accidents) (Daley et al.,
2009; Leger et al., 2014). Increased insomnia severity has also
been shown to be associated with increased healthcare utilisation
(Wickwire et al., 2016) and people with insomnia have higher
healthcare costs than controls (Wickwire et al, 2019). The major-
ity of studies indicate that the cost of treating insomnia is less
than the cost of not treating insomnia, and that treatment costs
appear to be recouped within 6–12 months (Morgan et al., 2004;
Wickwire et al., 2016).
Recommendations
It is important to treat insomnia because the condition
causes decreased quality of life, is associated with impaired
functioning in many areas, and leads to increased risk of
depression, anxiety and possibly diabetes and cardiovascu-
lar disorders (A).
Goal of treatment:
To lessen suffering.
Improve daytime function.
Type of treatment:
Cognitive-behavioural therapy for insomnia (CBTi)
should be offered as first line treatment.
In case of treatment failure, unavailability of CBTi, or
inability to engage with CBTi, pharmacological treatment
with an evidence base should be offered (see Figure 2).
Figure 2. Treatment of insomnia.
Wilson et al. 929
Psychological treatment of insomnia
Underpinning principles – CBT. Psychological treatment of
insomnia should be considered appropriate for a number of
reasons.
First, insomnia has across diagnostic and classification sys-
tems been long regarded as a ‘psychophysiological’ disorder in
which mental and behavioural factors play crucial roles as predis-
posing, precipitating and perpetuating factors (Espie, 2002; Espie
et al., 2006; Kalmbach et al., 2018; Riemann et al., 2010;
Spielman et al., 1987). Core features of insomnia are heightened
arousal and learned sleep-preventing associations. Arousal can
reflect a general cognitive hypervigilance and many patients
describe ‘racing thoughts’ as a problem when they are trying to
sleep. A cycle develops in which the more one strives to sleep,
the more agitated one becomes, and the less able one is to fall
asleep. All of these sleep-related behaviours and attitudes con-
trast with those of the ‘good sleeper who seems to sleep without
much thought or planned behaviour.
Second, CBTi directly addresses these cognitive and behav-
ioural factors, particularly those that maintain insomnia. CBTi
employs a package of interventions designed to encourage ‘poor
sleepers’ to think and behave like ‘good sleepers’. The therapy is
manualised and health professionals can be trained to administer
it either individually or in a group setting. Therapies are multi-
modal, embodying techniques such as sleep restriction and stim-
ulus control as well as cognitive restructuring. CBT then is a
treatment modality, as is pharmacotherapy. The latter comprises
a range of licensed medications, and the former a range of proven
psychotherapeutic methods.
Third, and most importantly, there is a substantial evidence base
for the safety, efficacy and clinical effectiveness of CBTi. Systematic
reviews and meta-analyses have consistently found that CBTi
reduces sleep latency and the duration and frequency of night-time
wakenings, as well as increasing sleep efficiency with moderate to
large effect sizes. CBT also increases total sleep time, and the ben-
efits of CBTi are durable at medium to long-term follow up.
Importantly, side-effects with CBTi are relatively minimal,
with some patients reporting mild daytime sleepiness in the early
stages of sleep restriction and stimulus control. However, there is
considerable evidence of generalised benefit to mood, wellbeing,
and to social and occupational functioning in controlled trials
(Espie et al., 2018a; Van Houdenhover, 2011). Recent insomnia
CBT studies have demonstrated a causal relationship between
improvements in sleep and improvements in mental health symp-
toms, wellbeing and quality of life (Espie et al., 2018a, b;
Freeman et al., 2017)
CBTi is therefore lastingly effective, and is the recommended
treatment of first choice for chronic insomnia in guideline docu-
ments (e.g. American College of Physicians: Qaseem et al., 2016;
European Insomnia Guidelines, Riemann et al., 2017).
A longstanding problem for CBTi is not its effectiveness but
its availability. At the time of writing of the 2010 BAP guideline,
reference was made to this ongoing challenge and how it hin-
dered real world implementation. CBT is traditionally offered
face-to-face and so has been restricted by the number of available
therapists to provide treatment. The advent of digital CBT
(dCBT; Web and mobile delivery) has changed this landscape.
There are now four published meta-analyses of dCBT which
indicate comparable effectiveness to in-person CBTi and demon-
strate the emerging opportunity for patients to access this form of
CBT treatment (Cheng and Dizon, 2012; Seyffert et al., 2016; van
Straten et al., 2018; Zachariae et al., 2016). Two dCBT interven-
tions in particular have a considerable evidence base. There are
seven published RCTs of the SHUT-i product (with a total sample
size n~2100 and eight published RCTs of the Sleepio product
(total n~6900). The available evidence base is not just for CBT (in
general), but also for discrete dCBT ‘products’; a situation that
more closely aligns with the pharmaceutical literature where spe-
cific drugs can be seen as discrete pharmacotherapy products that
can be offered as part of a formulary-driven pharmacopeia.
Recommendations
CBT-based treatment packages for chronic insomnia
including sleep restriction and stimulus control are effective
and therefore should be offered to patients as a first-line
treatment (A).
Both face to face CBTi and dCBTi are efficacious (A).
dCBTi has the potential to offer patients and clinicians a
choice amongst evidence-based alternatives (CBT or drugs)
in routine clinical care (A).
Drug treatments for insomnia
What is known about psychological treatment of insomnia:
CBTi is an effective treatment for insomnia when delivered
individually or in small group format (Ia).
CBTi is an effective treatment for insomnia when delivered
digitally as a Web/mobile intervention (1a).
CBTi is as effective as prescription medications for short-term
treatment of chronic insomnia. Moreover, there are indications
that the beneficial effects of CBT may last well beyond the
termination of active treatment (Ia).
Improvements in sleep following CBTi for chronic insomnia
mediate improvements in mental health symptoms, wellbeing
and quality of life (1a).
What is not known:
What are the predictors of failure to respond to CBTi?
Does hypnotic medication enhance the effects of CBTi and, if it
does, under what circumstances?
What is known about drug treatments for short-term treatment
of insomnia:
Gamma-aminobutyric acid (GABA)-positive allosteric modulators
(PAMs) are efficacious for insomnia (Ia).
Safety concerns (adverse events and carry-over effects) are
fewer and less serious in hypnotics with shorter half-lives (Ib).
Prolonged release melatonin improves sleep onset latency and
quality in patients over 55 years (Ib).
Suvorexant is efficacious in insomnia (Ia).
Doxepin in very low doses (3 mg and 6 mg) is efficacious in
insomnia (Ia).
What is not known:
Does improvement in insomnia last after treatment is stopped?
Does treatment reduce the risk of subsequent depression?
Who responds and who does not?
930 Journal of Psychopharmacology 33(8)
Underpinning principles – pharmacology.
Most drugs which
affect the brain do so by affecting neurotransmitter function in
the brain, which they can do by:
simulating the action of a brain neurotransmitter on the
receptor (agonists, partial agonists)
blocking neurotransmitter action on postsynaptic recep-
tors (antagonists)
changing receptor sensitivity (allosteric modulators)
increasing the amount of neurotransmitter present in the
synapse, either by increasing the release of it into the syn-
aptic cleft, blocking its transportation out of the cleft, or
preventing the action of enzymes which break it down.
Arousal is maintained by parallel neurotransmitter systems with
cell bodies located in brainstem or midbrain centres, with projec-
tions to the thalamus and forebrain. These activating neurotrans-
mitters are noradrenaline, serotonin (5-HT), acetylcholine,
dopamine, histamine and the orexin system with cell bodies in
the hypothalamus, which together promote wakefulness through
regulating arousal pathways (and inhibiting sedating ones). For
all of these arousing neurotransmitters, waking can be promoted
by increasing their function, and sleep or sedation by decreasing
their function in the brain.
The promotion of sleep is regulated by a number of other neu-
rotransmitters; primary amongst these is GABA, the main inhibi-
tory neurotransmitter in the brain. The majority of brain cells are
inhibited by GABA so increasing its function reduces arousal and
produces sleep, and eventually anaesthesia. There are many sub-
sets of GABA neurones distributed throughout the brain but a
particular cluster in the hypothalamus (ventrolateral preoptic
nucleus) can be considered to be the sleep ‘switch’ (Saper et al.,
2005). These neurones ‘switch off brain arousal systems at the
level of the cell bodies and therefore promote sleep. GABA
receptors in the cortex can also promote sedation and sleep by
inhibiting the target neurones of the arousal system.
Benzodiazepines, so-called ‘Z drugs’ and barbiturates all enhance
the effects of GABA at the GABA
A
receptor (GABA-PAMs).
There are a number of subtypes of this receptor which are rele-
vant for sleep not only because of their different location in the
brain but also because of the fact that some drugs for insomnia
are selective for a particular subtype. The alpha-1 subtype is
highly expressed in the cortex and probably mediates the seda-
tive and hypnotic effects of many drugs that act at the benzodiaz-
epine site; zolpidem targets this subtype preferentially (Sanna
et al., 2002). The alpha-3 subtype predominates in the reticular
nucleus of the thalamus which plays an important role in regulat-
ing sleep. This subtype is particularly targeted by eszopiclone
(Jia et al., 2009). Traditional benzodiazepine drugs for insomnia
act on the alpha-1, alpha-2, alpha-3 and alpha-5 subtypes.
The other main sleep-promoting neurotransmitter is adeno-
sine. Brain levels rise during the day and are thought to lead to
sleepiness, which increases the longer the time since the last
sleep. The arousing and sleep-impairing effects of caffeine
(Landolt et al., 2004) are thought to be due to blockade of aden-
osine-A2 receptors, so attenuating this natural process (Porkka-
Heiskanen et al., 2002).
Histamine neurons form part of the neurotransmitter network
promoting arousal. Histamine levels are high in daytime and low
during sleep. Drugs which reduce histamine function (H1 recep-
tor antagonists or antihistamines) reduce arousal. Antihistamine
medications which cross the blood-brain barrier, such as promet-
hazine and diphenhydramine, are widely used ‘over-the-counter’
and prescribed to promote sleep. Unfortunately, they are not
selective for histamine, and their actions at other brain receptors
(particularly antagonism at cholinergic, noradrenergic and (pro-
methazine) dopaminergic receptors contribute to an adverse
effect profile. The most selective available medication is very
low dose doxepin, which, at doses from 1–6 mg, has little or no
effect at brain receptors other than H1 receptor antagonism. This
drug is approved in the USA for insomnia, but is not available in
Europe. Esmirtazapine, the S-enantiomer of mirtazapine is also
selective for H1 receptors, and is undergoing evaluation (Ivgy-
May et al., 2015; Ruwe et al., 2016).
Orexin is a neurotransmitter intimately involved in sleep and
waking. When the orexin receptors 1 and 2 (OR1 and OR2) in the
hypothalamus are activated they promote waking, and antago-
nists for these have been found to promote sleep. Several recep-
tor antagonists have been developed and one, suvorexant, is
licensed in the USA for insomnia (Herring et al., 2016). These
drugs are not yet available in Europe but several agents are being
evaluated in clinical trials.
Melatonin is produced in the pineal gland and has an important
role in regulating circadian rhythms (Cajochen et al., 2003; Dijk
and von Schantz, 2005). The circadian ‘pacemaker’ is the supra-
chiasmatic nucleus (SCN) of the hypothalamus and when active it
inhibits melatonin secretion in the pineal gland. Once melatonin
appears in the plasma it enters the brain and binds to melatonin
receptors in the hypothalamus. Melatonin has both phase-shifting
effects, so changing the timing of the biological clock, and direct
sleep-facilitating effects. Administering exogenous melatonin or
analogues such as ramelteon (licensed in the USA) can promote
sleep onset. A slow-release formulation of melatonin has been
licensed on the basis of improved sleep continuity and daytime
well-being in people aged over 55 years with insomnia. Melatonin
production is reported to decline with age and to be lower in mid-
dle-aged and elderly patients with insomnia than in good sleepers
(Attenburrow et al., 1996; Dowling et al., 2008; Haimov, 2001;
Leger et al., 2004). Beta-adrenergic receptor blockers and non-
steroidal anti-inflammatory drugs inhibit melatonin secretion.
Underpinning principles – pharmacokinetics. The principles
of the ideal drug for insomnia have been discussed for decades
and are outlined in Figure 3. All licensed drugs for insomnia
improve one or more aspects of subjective sleep, and some also
improve daytime functioning (see below – but note that many
drugs have not been evaluated on this parameter).
Kinetic aspects are important both in terms of how quickly the
drug enters the brain and how long its effects last (for comparison
of drugs see Tables 3 and 4 in Wilson et al., 2010). The faster the
drug enters the brain, the sooner sleep is induced. Some agents
used for insomnia have not been active in this aspect of sleep
because of poor kinetic properties: for example, temazepam tablets
have a poorer bioavailability and slower absorption (and thus a
longer presence in the body) than the previous gel formulations.
Drugs that enter the brain very quickly, though effective, may need
to be taken in the bedroom or even in bed to prevent people falling
asleep before they are in bed (see Zentiva Pharma UK Limited,
2002; zolpidem Summary of Product Characteristics (SPC)).
Wilson et al. 931
The ease of waking and the propensity to daytime carry-over
(‘hangover’) effects are determined by the duration of action –
with GABA-PAMs this is typically defined by the elimination
half-life of the drugs (see Tables 3 and 4 in Wilson et al., 2010)
and the dose taken. Drugs with half-lives of more than six hours
tend to leave sufficient residual drug in the brain to cause hango-
ver effects in the morning. This was particularly the case with the
first benzodiazepine drugs for insomnia such as nitrazepam,
which was associated with daytime sedation and falls (Trewin
et al., 1992). The rationale for developing zopiclone, zolpidem
and zaleplon was to make shorter half-life drugs with minimal
carry-over effects (Nutt, 2005b). This was largely achieved,
although some hangover effects are seen with zopiclone and esz-
opiclone (Boyle et al., 2012; Staner et al., 2005).
A very short half-life limits a drug’s duration of action on
sleep, and zolpidem is less effective at maintaining sleep
throughout the night than drugs with a longer half-life. A con-
trolled release formulation of zolpidem (currently only availa-
ble in the USA) prolongs its nocturnal actions and enhances
sleep continuity, though only by 10s of minutes (Greenblatt
et al., 2006). Individual factors seem important and some peo-
ple are more susceptible to carry-over effect than others, prob-
ably due to individual differences either in the rate of drug
clearance, which can vary by as much a two-fold between sub-
jects, or sensitivity to drug actions. In particular, women tend to
have higher blood concentrations of zolpidem, and greater
impairment of driving ability, the following morning than do
men (Farkas et al., 2013). The US Food and Drugs Administration
responded to this finding by requiring manufacturers to recom-
mend gender-specific labelling with dosing for women being
half that of men.
Tolerance, dependence and withdrawal. Dose escalation
above recommended doses in patients with insomnia alone
appears uncommon, and tolerance to the effects of GABA-PAM
drugs for insomnia is not a frequently encountered problem in
clinical experience. Many patients use the same dose for months
or years and still feel it works. However, a temporary worsening
of sleep, usually with increased sleep onset latency, is reported
during the withdrawal period for most GABAergic agents (Hajak
et al., 2009; Soldatos et al., 1999; Voshaar et al., 2004). Although
there have been no head-to-head studies of this question, there is
some lower level evidence in humans that subtype-selective
drugs such as eszopiclone produce less tolerance and rebound
(Krystal et al., 2003; Nutt and Stahl, 2010).
Animal and human research demonstrates that brain receptor
function changes in response to chronic treatment with benzodi-
azepine receptor agonists, and this takes time to return to pre-
medication levels after cessation of medication. There is evidence
from animal studies that chronic administration of benzodiaz-
epines produces adaptive changes in the receptor which attenuate
the effects of the endogenous neurotransmitter GABA, and so
produce symptoms on withdrawal (Bateson, 2002). It may be
possible to develop drugs with a lower propensity to such effects:
through targeting specific subtypes of the benzodiazepine recep-
tor by changing the chemical structure to produce a different
interaction at the pharmacophore; or by making partial agonists
(Doble et al., 2004).
Considerations of dependence on GABA-PAMs are contingent
on what happens when treatment is stopped. A psychological
dependence is seen in many patients and some are reluctant to stop
treatment. If they do stop, there can be relapse, where the patient’s
original symptoms return; or rebound of symptoms, where for one
Figure 3. The ideal sleeping pill.
932 Journal of Psychopharmacology 33(8)
What is known about long-term treatment:
Insomnia is often long-lasting and is in practice often treated
with hypnotics for long periods in clinical practice (Ib).
Studies suggest that dependence (tolerance/withdrawal) is
not inevitable with hypnotic therapy up to one year with
eszopiclone, zolpidem, ramelteon (Ib).
Intermittent dosing may reduce the risk of tolerance and
dependence (Ib).
What is not known:
How can we predict the needed treatment duration?
How and when should treatment be discontinued?
Should dosing for longer periods be nightly or intermittent?
How do we detect the abuse prone individual in the clinic?
Does hypnotic therapy affect the course of insomnia or
associated conditions?
or two nights there is a worsening of sleep disturbance, with longer
sleep onset latency and increased waking during sleep; this is com-
monly reported by patients and has been documented in some
research studies (Hajak et al., 2009; Soldatos et al., 1999). More
rarely, there is a longer withdrawal syndrome. All of these can be
ameliorated by resuming medication. The withdrawal syndrome is
characterised by the emergence of symptoms not previously
reported, such as agitation, headache, dizziness, dysphoria, irrita-
bility, fatigue, depersonalization, hypersensitivity to noise and
visual stimuli. Physical symptoms which have been described
include nausea, vomiting, muscle cramps, sweating, weakness,
muscle pain or twitching and ataxia. This syndrome usually
resolves within a few weeks, but persists in some patients, and this
persistence may be related to personality traits and cognitive fac-
tors (Murphy and Tyrer, 1991).
Pharmacological treatment of insomnia. All licensed drugs
licensed for insomnia are efficacious (I). As explained above,
some may improve sleep earlier in the night, as they enter the
brain more quickly, and thus reduce sleep onset latency. Duration
of action depends to a great extent on half-life and for instance in
a patient with predominantly sleep-onset insomnia, a shorter act-
ing drug such as zolpidem or melatonin might be appropriate,
and for those with awakenings throughout the night a slightly
longer acting drug such as zopiclone may be preferable.
Most of the drugs approved for insomnia enhance GABA
function in the brain. As well as promoting sleep these drugs are
anxiolytic, anticonvulsant and myorelaxant, and can cause ataxia
and memory problems when taken other than just before a period
in bed. If their effect in the brain persist after waking in the morn-
ing they are sometimes described as ‘hangover effects’ therefore
differences in the pharmacokinetics of individual benzodiaz-
epines (or ‘Z drugs’) are particularly important. Melatonin does
not give rise to motor or memory effects; however the synthetic
melatonin agonist ramelteon gives rise to sleepiness which can
persist for 12–14 h (Cohen et al., 2010). Recent clinical trials
have measured daytime outcomes after drugs for insomnia, and
beneficial effects have been reported for melatonin in those over
55 years, and for zolpidem, zopiclone, eszopiclone and lormetaz-
epam. These measures have not been used in studies of other
drugs, so their effects on daytime function are not documented.
In systematic reviews of GABA-PAMs, adverse events/side
effects are less common and less severe for the Z-drugs zolpidem
and eszopiclone (Buscemi et al., 2007). Controlled studies meas-
uring cognitive and psychomotor function (such as digit-symbol
substitution test, and memory) in insomnia patients have only
shown next-day deleterious effects consistently after use of flu-
razepam (very long-acting) or very high doses of other benzodi-
azepines (Buscemi et al., 2005). Evidence for hypnotic effects on
next day driving in insomnia patients is limited, however epide-
miological studies show that road accidents are increased in peo-
ple taking benzodiazepines or zopiclone (Barbone et al., 1998;
Neutel, 1995). Studies in healthy volunteers show that residual
effects of drugs for insomnia increase with their half-life duration
(Verster et al., 2006). Effects of insomnia itself on driving have
not been studied extensively, though sleep deprivation does
impair driving performance (Connor et al., 2002). In a controlled
study of patients with insomnia in a driving simulator, there was
next-day impairment after zopiclone and lormetazepam but not
zolpidem, when compared with placebo (Staner et al., 2005).
Very low doses of doxepin, (1, 2 or 6 mg when doxepin acts
only as a histamine antagonist) improve sleep in adult (18–65
years) and elderly (65 years and older) insomnia patients (Yeong
et al., 2015). Doxepin has a preferential effect on reducing awak-
enings in the latter half of the night (Krystal et al., 2010) and does
not appear to give rise to residual daytime effects (Krystal et al.,
2011). Suvorexant, an antagonist at OR1 and OR2 receptors,
improves sleep in adult and elderly insomnia patients. It increases
subjective total sleep time and decreases subjective wake time in
the middle and end of the night and subjective time to sleep
onset; a few healthy volunteers had impairment of driving ability
nine hours after higher doses of suvorexant (Vermeeren et al.,
2015). Neither of these drugs is currently available in Europe.
Recommendations
Factors which clinicians need to take into account when
prescribing are efficacy, safety, and duration of action (A).
Other factors are previous efficacy of the drug or adverse
effects, history of substance abuse or dependence (D).
Long-term use of sleeping medications
The question of long-term hypnotic treatment is one of the more
controversial areas in psychopharmacology. It has long been
stated that sleeping medication should not be used long-term for
the treatment of insomnia. This was the consensus view of the
panel of a 1983 National Institute of Health (NIH; 1983)
Consensus Conference on the medication treatment of insomnia,
which became a guideline for clinical practice in the USA, and
later the UK Committee on Safety of Medicines and the Royal
College of Psychiatrists both recommended only short-term use.
While it was appreciated that benzodiazepine hypnotic agents
had a favourable risk-benefit ratio and were first-line agents for
insomnia management, all of these reports expressed concerns
about the risks of physical dependence and recommended their
use should be limited to periods of 2–3 weeks. Despite the rec-
ommendation for treatment with hypnotic drugs being only 2–4
weeks, many millions of patients worldwide remain on long-term
treatment (Ohayon et al., 1999; Maust et al., 2019).
The reasons for longer term use are complicated and difficult
to research but are probably similar to those which affect
Wilson et al. 933
understanding of longer term benzodiazepine treatment in
patients with anxiety disorders. We do not know the proportions
of longer term users that have continuing insomnia requiring
daily drug treatment, or who do not need the drug at all, or who
are afraid to try discontinuing because of fear or experience of
rebound insomnia. In one study where people were successful in
discontinuing benzodiazepine hypnotics, a follow-up after two
years revealed approximately 40% had resumed regular use
(Belanger et al., 2005; Morin et al., 2005a), which suggests some
people have enduring problems with sleep which benefit from
treatment. Insomnia may have some similarities with depression,
in that both represent long-term disorders in which maintenance
treatment may be needed in many patients (Jindal et al., 2004). A
related issue is whether early intervention at the onset of insom-
nia might reduce the likelihood of it persisting.
Placebo-controlled trials of treatment for durations longer than
three weeks that can more definitely assess safety and efficacy,
and determine whether dependence phenomena occur, have been
undertaken. Trials of nightly dosing for up to 12 months duration
suggest that tolerance and withdrawal do not generally occur with
some hypnotics: zolpidem (one study of 12 months and one of
eight months duration) eszopiclone (two studies of six months
duration); ramelteon (a six-month study with outcome assessed
with polysomnography but not self report); and temazepam (a
two-month study) (Ancoli-Israel et al., 2010; Bastien et al., 2003;
Krystal et al., 2003; Mayer et al., 2009; Morin et al., 1999; Roehrs
2012; Walsh et al., 2007). Others agents have not been studied for
longer durations. The available evidence does not suggest there is
an unfavourable risk/benefit transition at 3–4 weeks for any agent.
Open-label studies of nightly dosing for periods up to one year
with the agents studied (zolpidem, eszopiclone, and ramelteon)
suggest that discontinuation symptoms are generally mild and
infrequent (Ancoli-Israel et al., 2005; Randall, 2012; Richardson
et al., 2009). Intermittent, non-nightly, dosing is also an important
consideration with respect to long-term hypnotic treatment. Many
individuals do not have nightly insomnia and treatment only when
needed can decrease the risks and costs of therapy and reduce psy-
chological dependence/treatment withdrawal anxiety. There is
evidence from a placebo-controlled trial for sustained efficacy
and safety for six months of ‘as needed’ treatment (subjects being
required to take at least three doses per week) with controlled
release zolpidem 12.5 mg (Krystal et al., 2008).
In conclusion, insomnia is often long-lasting and often treated
with hypnotics for long periods in clinical practice. Controlled
trials of longer-term use are being undertaken and these suggest
dependence (tolerance/withdrawal) is not inevitable with hyp-
notic therapy up to one year, and is not characteristic of the sev-
eral agents studied. The longer-term safety and efficacy of many
other commonly used hypnotics remains uncertain.
A number of critical issues remain unresolved. We currently
lack the means to determine who should receive longer-term
treatment and to predict the required treatment duration. Lacking
the means to determine the optimal duration of therapy, a rational
approach is to carry out periodic trials of tapering and discon-
tinuing medication to determine if continued therapy is indi-
cated (Krystal, 2009). As such, the duration of treatment is
decided by a series of risk/benefit decisions based on trial dis-
continuations. This approach provides an ‘exit strategy’ and
addresses concerns that once started hypnotic therapy could be
unending. Concomitant CBT during tapered discontinuation is
helpful (Morin et al., 2004; Parr et al., 2009; Belleville et al.,
2007). Another unresolved issue is whether to implement nightly
or intermittent dosing of hypnotics for a given patient. It is clear
that the medication works on the nights it is taken but poor sleep
occurs when it is not. In many instances this is a practical deci-
sion based on whether the patient can predict when they go to
bed whether they will have sleep difficulty. In addition, some
have argued that intermittent dosing may reinforce psychologi-
cal dependence on the drug.
Recommendations
Use as clinically indicated (A).
In general, hypnotic discontinuation should be based on
slowly tapering off medication (A).
CBTi during taper improves outcome (A).
Using drugs for depression to treat insomnia
Tricyclic antidepressants have long been used for insomnia, with
little evidence of efficacy (see Everitt et al., 2018), whereas the
serotonin reuptake inhibitors (SRIs) as a class generally disrupt
sleep early in the course of treatment (Mayers and Baldwin, 2005).
The alerting effect of SRIs can sometimes be offset by co-adminis-
tration of sedating drugs for depression such as trazodone, probably
because they block 5-HT2 receptors that are being overstimulated
by an increase in 5-HT (Kaynak et al., 2004); alpha-1 adrenergic
antagonism may also contribute. Other 5-HT2 antagonist drugs for
depression such as nefazodone (now discontinued) (Hicks et al.,
2002) and mirtazapine (Winokur et al., 2003) have been shown to
reduce insomnia in depression, especially early in treatment.
Low doses (sub-therapeutic for depression) of sedating tricy-
clics, particularly amitriptyline, have been used for decades to
treat insomnia. This is particularly common practice in primary
care in the UK, where amitriptyline 10 or 25 mg is also used for
long periods in many patients with chronic illness, particularly
those with pain syndromes. At this dose, amitriptyline is proba-
bly acting mostly as a histamine H1 receptor antagonist although
a degree of 5-HT2 and cholinergic muscarinic antagonism may
also contribute. There are no controlled studies of hypnotic effi-
cacy of low-dose amitriptyline in insomnia, and tricyclics are
more likely to be lethal than licensed hypnotics in overdose
(Nutt, 2005a). Controlled trials demonstrate an effect of doxepin
What is known about the use of drugs for depression to treat
insomnia:
There is limited evidence for efficacy of trimipramine,
trazodone and paroxetine in insomnia (Ib).
Older drugs for depression may affect a wide range of brain
receptors and have longer lasting carry-over effects than
traditional drugs for insomnia. They are associated with
increased risks of road accidents especially early in treatment
in depression (Ib).
What is not known:
Duration of effect (particularly as they are often prescribed for
long periods).
How their effects compare with approved medications for
insomnia.
934 Journal of Psychopharmacology 33(8)
in insomnia at sub-antidepressant dose (25 mg) (Hajak et al.,
2001) for four weeks, with rebound insomnia.
Trazodone is an agonist at 5-HT1A receptors, an antagonist at
5-HT2 and α1 adrenergic receptors and a weak 5-HT reuptake
inhibitor: it is the second most prescribed medication for insomnia
in the USA. It has a perceived absence of risk and is cheap, but
25–30% patients experience difficulty tolerating trazodone and
dropout rates tend to be higher than for benzodiazepine hypnotics
or Z-drugs. Although there have been 18 trazodone studies meas-
uring sleep outcomes, only two were in primary insomnia, and
only one was a controlled study (Walsh et al., 1998). This study
used 50 mg trazodone vs placebo, and found a significant effect
on sleep maintenance parameters at week 1 but not week 2, and a
high incidence of daytime somnolence. Trimipramine is a drug for
depression which blocks α-1 adrenergic, histamine H1, dopamine
D2, 5-HT2 and cholinergic receptors (Gross et al., 1991;
Richelson, 1994). There is one controlled trial (Riemann et al.,
2002) in insomnia at doses of 50–200 mg for four weeks which
found a significant improvement in sleep efficiency as measured
by polysomnography, paralleled by subjective improvements.
Side effects were described as marginal. Paroxetine, an SRI, was
studied in patients with insomnia aged over 55 years, at a median
dose of 20 mg for six weeks (Reynolds, III et al., 2006), there
being a 50% response rate (placebo 38%) with subjective sleep
quality and daytime well-being improved. This seeming paradoxi-
cal action of paroxetine to improve sleep is probably related to its
good efficacy in many anxiety disorders where it seems to reduce
recurrent thinking and ruminations.
Taking SRIs, venlafaxine, mianserin or mirtazapine increases
the risk of restless legs syndrome and periodic limb movements
of sleep (Hoque and Chesson, Jr., 2010) and SRIs can induce or
exacerbate sleep bruxism (Wilson and Argyropoulos, 2005).
Recommendations
Use drugs according to a knowledge of pharmacology (A).
Consider drugs for depression when there is co-existent
mood disorder (A).
Beware toxicity of TCAs in overdose even when low unit
doses prescribed (A).
Drugs for psychosis for treatment of insomnia
Dopamine-5-HT antagonists, particularly quetiapine and olan-
zapine, have become widely used in the treatment of sleep prob-
lems with very little controlled trial evidence. For example,
prescriptions of quetiapine for sleep disturbances increased by
300% in Canada between 2005–2012 (Pringsheim and Gardner,
2014) and a cross-sectional study conducted using the US
National Health and Nutrition Examination Survey data from
1999–2010 found that quetiapine was the fourth most common
drug prescribed for insomnia (11%) (Bertisch et al., 2014).
PSG sleep studies in healthy participants have shown changes
in sleep architecture. These include increases in slow wave sleep
and decreases in rapid eye movement (REM) sleep with ziprasi-
done and olanzapine (Cohrs et al., 2004; Cohrs et al., 2005;
Sharpley et al., 2000). Some positive changes in measures of sleep
maintenance have been shown with clozapine, quetiapine, olan-
zapine and risperidone (Gimenez et al., 2007; Lindberg et al.,
2002; Sharpley et al., 2000). Subjective sleep was improved by
risperidone, olanzapine and quetiapine.
In models of transient insomnia, such as acoustic stress and
phase advance in healthy volunteers, quetiapine, ziprasidone and
lurasidone have shown improvements in sleep initiation and
maintenance (Cohrs et al., 2004, 2005; Karsten et al., 2017;
Krystal and Zammit, 2016).
In patients with schizophrenia (see Monti et al., 2017) clozap-
ine increased total sleep time (Kluge et al., 2014); olanzapine
increased slow-wave sleep and improved sleep-continuity meas-
ures (Gao et al., 2013; Kluge et al., 2014; Monti et al., 2017;
Muller et al., 2004; Salin-Pascual et al., 1999, 2004). Quetiapine
administration has generated mixed results in patients with schiz-
ophrenia, with Loebel et al. (2014) finding increases in daytime
sleepiness after six weeks quetiapine extended release (XR) 600
mg compared with placebo, whereas Keshavan et al. (2007) found
reduced sleep continuity compared with drug naïve patients.
In bipolar patients (see Monti 2016) six months add-on clozap-
ine treatment increased total sleep time compared with baseline
(Armitage et al., 2004). Risperidone and olanzapine both improved
sleep continuity when added to an SRI in depression (Sharpley
et al., 2003; 2005; Lazowski et al., 2014). Furthermore, Moreno
et al. (2007) found that olanzapine improved sleep continuity in
patients with bipolar disorder during a manic episode. Using actig-
raphy, Todder et al. (2006) and Kim et al. (2014) showed that
adjunctive or monotherapy quetiapine improved sleep continuity
in patients with unipolar or bipolar disorder. A PSG study with
ziprasidone augmentation in bipolar patients with an acute depres-
sive episode improved sleep induction, sleep continuity and sleep
architecture (Baskaran et al. 2013). Overall, these results indicate a
potential beneficial effect on sleep of dopamine -serotonin antago-
nists in patients prescribed them for a labelled indication.
In patients with insomnia, a small open study of quetiapine (25
mg in most patients) for six weeks (Wiegand et al., 2008) showed
improvements in sleep, with transient adverse effects of morning
hangover and dry mouth. A double-blind, placebo- controlled
study which investigated the efficacy of quetiapine (25 mg) in pri-
mary insomnia (Tassniyom et al., 2010) had a small sample size
(n=13) and subjective improvements in sleep were not significant.
Side effects are well documented, and include weight gain, met-
abolic syndrome, extrapyramidal symptoms and risk of tardive
dyskinesia. There are some case reports of abuse of quetiapine in
inpatients and prisoners (Sansone and Sansone, 2010). A review of
quetiapine for use in insomnia (Anderson and Vande Griend, 2014),
concluded that its benefit in the treatment of insomnia has not been
What is known about use of drugs for psychosis in treatment of
insomnia:
Dopamine serotonin antagonists particularly olanzapine and
quetiapine improve sleep in healthy participants (Ib).
Quetiapine, ziprasidone and lurasidone improve sleep in models
of transient insomnia (Ib).
When used on label, clozapine, olanzapine, quetiapine,
risperidone and ziprasidone improve sleep continuity in
schizophrenia, unipolar and bipolar disorder (I).
Minor improvements in sleep in primary insomnia are seen after
quetiapine (IIb).
Side effects are common because of the broad pharmacological
actions of these drugs (I).
What is not known:
How drugs for psychosis compare with approved drugs for
insomnia?
Wilson et al. 935
proven to outweigh potential risks, even in patients with a comorbid
labelled indication for quetiapine. An American Academy of Sleep
Medicine clinical practice guideline for the pharmacologic treat-
ment of chronic insomnia in adults also concluded that quetiapine
was not indicated for use in insomnia (Sateia et al., 2017).
Recommendations
Side effects are common because of the pharmacological
actions of drugs for psychosis and there are a few reports of
abuse.
Together these indicate no indication for use as first-line
treatment (D).
Antihistamines (H1 antagonists). Antihistamines are sedating
and are sold as ‘over the counter (OTC) sleeping medi cations.
There is only limited evidence that these older non-selective anti-
histamines work, although some modest benefits have been
reported after two weeks dosing with diphenhydramine in mild
insomnia (Morin et al., 2005b). More profound acute effects on
sleep have been reported for both promethazine and hydroxyzine
in healthy volunteers (Adam and Oswald, 1986; Alford et al.,
1992) but the latter is not available OTC, and both have quite long
durations of action so are likely to cause hangover effects.
Antihistamines are sometimes used in alleviation of insomnia
in drug and alcohol withdrawal where traditional hypnotics are
less suitable due to the risk of cross-dependence, although there
are no controlled trials in this setting. These drugs have anticho-
linergic side effects, making them dangerous in overdose.
The selective antihistamine (low-dose) doxepin 1–6 mg has
been shown to be effective in insomnia in adults and the elderly,
with few residual effects, and is approved in the USA but not in
Europe. It has been shown to particularly reduce awakenings in
the latter part of the night. Another selective agent, low-dose
esmirtazapine, is currently undergoing evaluation.
Recommendations
The selective antihistamine doxepin (very low dose) is
effective in insomnia (A).
Non-selective histamine antagonists have a limited role in
psychiatric and primary care practice for the management of
insomnia (D).
Circadian rhythm disorders
Daily rhythms of sleeping and waking are controlled by a variety
of brain mechanisms, the most prominent of these being the cir-
cadian process (the ‘body clock’ signalling time for sleep) and
the homeostatic process (a build-up of sleep pressure during the
hours of wakefulness). These two processes work together to
consolidate sleep and wakefulness (Diik and von Schantz, 2005).
Circadian rhythm is a roughly 24-hour cycle in the physiolog-
ical processes of living beings. It is internally generated, although
it can be modulated by external cues (zeitgebers) such as sun-
light, and feeding and drinking, and in humans by daily routines
of work, exercise etc. Circadian rhythms are controlled by a brain
pacemaker in the SCN of the anterior hypothalamus, which has a
direct input from the retina signalling light levels. Most body sys-
tems, not only sleeping and waking, are to some degree modu-
lated by input from this circadian pacemaker.
The innate frequency of the circadian clock in humans is
slightly longer than 24 h, and synchronisation with the external
24-hour physical environment and day- to-day activities requires
daily adjustments to the internal clock. The light-dark cycle is the
strongest synchronising agent for the circadian system. In humans,
light exposure in the evening produces delays, and in the early
morning produces advances. The SCN also receives internal sig-
nals from the pineal gland, via the nocturnal release of melatonin.
Endogenous melatonin release begins to increase 2–3 hours
before sleep onset, and peaks in the middle of the night. Exogenous
melatonin given during the early morning delays the timing of
circadian rhythms and, given during the early evening, induces
advances in this timing, which contrasts with the effects of light.
There are differences within humans in their preference for
sleep times (chronotype). Some people preferring to rise early and
are at their best in the morning (‘morningness’, or ‘larks’) and
those who rise later and are at their peak later in the day (‘evening-
ness’, or ‘owls) These preferences be determined in part by genet-
ics. Children are early chronotypes and become progressively
later (delaying) as they grow older, reaching a maximum in their
‘lateness’ at around the age of 20 years. After 20 years, they
become earlier again (advancing) with increasing age. Young
women reach their maximum in lateness earlier than men; men
continue to delay their sleep until around the age of 21 years and
remain later chronotypes for most of their adulthood (Roenneburg
et al., 2004). This gender difference disappears at around the age
of 50 years, which coincides with the average age of menopause.
Circadian rhythm sleep-wake disorders (CRSWDs) occur
when there is an alteration of the endogenous circadian system or
a misalignment between the endogenous circadian rhythm and
the sleep-wake schedule required by the physical environment or
social or work timetable. This results in insomnia when they are
needing to sleep, and sleepiness when alertness is required, caus-
ing significant distress and impairment of function.
There are six CRSWDs defined in ICSD:
Jet Lag Disorder occurs when the circadian clock has a tem-
porary misalignment after rapid transition across time zones.
Symptoms of insomnia, daytime sleepiness and physical
discomfort normally disappear once there is exposure to
zeitgebers in the new environment, within a length of time
proportional to the number of time zones crossed. Thus after
an eastbound trip from the USA west coast the circadian clock
might take several days to settle into the UK routine. This
condition tends not to present to health professionals for treat-
ment, although it is likely to have some destabilising impact
on conditions such as bipolar disorder.
Delayed Sleep Wake Phase Disorder (DSWPD) affects 2–8%
of the population and is most prevalent in young adults. There
is usually difficulty falling asleep before 02:00–03:00 (some-
times later), and on days without work/school/college the
preferred wake time is after 10:00, resulting in sleep-onset
insomnia and difficulty waking up in the morning on days
when an early bedtime for an early start time is necessary.
There is a high incidence of comorbidity with psychiatric dis-
order in those with DSWPD with up to 60% of people having
a diagnosis of depression, substance abuse, or anxiety (Abe
et al., 2011; Murray et al., 2017; Reid et al., 2012).
Advanced Sleep Wake Phase Disorder (ASWPD) is much rarer,
with reports of an average sleep onset from 18:00–21:00 and
936 Journal of Psychopharmacology 33(8)
Current understanding of circadian rhythms and sleep physiology
provides a strong theoretical basis for the use of melatonin in
some, but not all CRDs. Empirical evidence for efficacy is strong
in some CRDs, but weak or absent in others. Melatonin agonists
may be promising in the treatment of CRDs, e.g. non-24-hour
SWRD (Lockley et al., 2015), but there remains a need for RCTs
in well-characterised CRD populations.
There is solid evidence to support the use of melatonin in
jet lag (Spiegelhalder et al., 2017) but (immediate release)
melatonin has to be taken near desired bedtime otherwise there
may undesired daytime sleepiness. An evidence-based strategy
for minimising jetlag which includes strategic scheduling of
sleep combined with melatonin is given by Sack (Sack, 2010).
In DSWPD, there is both a theoretical and an empirical basis for
use of melatonin, which is effective in practice, shown in two sys-
tematic reviews (MacMahon et al., 2005; Sack et al., 2007a); how-
ever studies in these reviews vary in the physiological and subjective
outcomes measured. Direct comparison with other therapies such
as timed-light exposure, for which there is a little evidence of effi-
cacy (see below), or chronotherapy, for which there are no con-
trolled trials, has not been reported. In one trial, efficacy of
melatonin combined with sleep scheduling was compared to sleep
scheduling alone and found to be effective (Sletten et al., 2018).
In non-24-hour sleep rhythm disorder, in sighted individuals, case
reports (n=5) suggest a positive benefit of melatonin. The evidence in
blind people is more compelling where case reports and two small,
single-blind placebo-controlled studies are positive (Sack et al.,
2007a; Skene and Arendt, 2007; Skene et al., 1999). Two RCTs of the
selective melatonin agonist tasimelteon in totally blind individuals
showed a positive effect on entrainment (Lockley et al., 2015).
There is no evidence of the efficacy of melatonin in irregular
sleep wake rhythm, or in shift work disorder, although there have
been some reports of use in shift workers with varying results
(Sack et al., 2007b).
Bright-light therapy has been used effectively in delayed
sleep phase syndrome (Shirani and St Louis, 2009). Exposure to
bright light of 2500 lux for two hours in the early morning,
combined with light restriction after 16:00 (dark goggles) is an
effective treatment for delayed sleep phase syndrome and a
light mask offering exposure to gradually increasing light inten-
sity through closed eyelids over the last four hours of habitual
sleep time has been shown to be effective in these patients.
In the elderly patient with dementia with an irregular sleep
wake disorder there is a little evidence to support the use of bright-
light therapy in combination with behavioural interventions, but
the use of melatonin is discouraged (Auger et al., 2015).
Recommendations
Clinical assessment is essential in delayed sleep wake
phase disorder, non-24-hour sleep rhythm disorder [A].
Melatonin may be useful in delayed sleep wake phase dis-
order, non-24-hour sleep rhythm disorder in non-sighted
individuals and jet lag disorder [B].
Other approaches such as behavioural regimes and sched-
uled light exposure (in sighted individuals) can also be used
[B/C].
Because of the necessity for careful timing of interven-
tions, patients with these disorders need to be treated in spe-
cialised sleep disorders centres (D).
wake time of 02:00–05:00. However, if allowed to sleep during
their preferred times, the sleep quality and duration are normal
for age. Prevalence is around 1%, but this may be a low estimate
as the lifestyle disruption may be less of a problem with earlier
timings. There sometimes a hereditary component to advanced
sleep wake phase disorder, and mutations of clock genes have
been identified in familial cohorts (Hirano et al., 2016).
In Irregular Sleep Wake Rhythm Disorder (ISWRD), there is
no clear main sleep period, but sleep and wake periods distrib-
uted irregularly through the 24-hour period with at least three
sleep bouts. There is thought to be a disruption of either the
central pacemaker, or of perception of environmental cues,
and this disorder is most common among young people with
neurodevelopmental disorders and adults with neurodegen-
erative disorders and occasionally with traumatic brain injury.
Non-24-hour Sleep Wake Rhythm Disorder (SWRD) occurs
when individuals are unable to entrain to the 24-hour day but
follow their endogenous circadian period, which is usually
slightly longer than 24 h. Their sleep-wake routine moves pro-
gressively later each day, and sleep complaints may consist of
insomnia, excessive daytime sleepiness, or both. A large pro-
portion of people with this disorder are visually impaired, and
without light perception, so the resetting of endogenous rhythms
through the retino-hypothalamic pathway is not possible.
Shift work sleep disorder (SWSD) is characterised by exces-
sive sleepiness during work and insomnia when trying to sleep
between shifts. There are many shift work schedules; they
may be permanent, rotating or irregular, so the presentation
of SWSD is variable. It is not known why some people adapt
adequately to changing work period timing, and others do not.
Disruption of circadian rhythms may also be present in other
sleep disorders, such as insomnia (Flynn-Evans et al., 2017)
Diagnosis of circadian rhythm disorders
Assessment of these disorders involves interview, sleep diaries
(from parents/carers if necessary) and actigraphy for 14 days. In
the case of DSWPD and ASWPD it is recommended to give the
morningness-eveningness questionnaire (Horne and Ostberg,
1976.
Treating circadian rhythm disorders
What is known:
Melatonin is effective in jet lag disorder (1a), delayed sleep phase
syndrome (Ib) and non-24-hour sleep rhythm disorder (IIa).
Light therapy is effective in delayed sleep phase syndrome (III).
What is not known:
What are the best efficacy measures – subjective or objective?
Is there a need to distinguish between adults and adolescents
in DSWPD since sleep times are somewhat delayed in normal
adolescence?
Is there a need to distinguish between sighted and blind
individuals?
Is melatonin or light therapy more effective for DSWPD?
Wilson et al. 937
Parasomnias
Parasomnias are unusual episodes or behaviours occurring dur-
ing sleep which disturb the patient or others and this document
addresses those that cause significant distress and therefore pre-
sent for treatment. Violent or unusual night-time attacks may
arise from deep non-REM sleep (night terrors and sleepwalking)
or from REM sleep [sleep paralysis, severe recurrent nightmares,
REM behaviour disorder(RBD)] and treatments depend on which
disorder is present.
Night terrors (also called sleep terrors) are recurrent episodes
of abrupt awakening from deep non-REM sleep, usually in first
third of night, usually with a scream and signs of intense fear and
autonomic arousal, and the patient is unresponsive to comforting.
They may sit up in bed and sometimes engage in automatic
behaviour associated with fear and escape. There is usually no
detailed recall, and if the patient wakes from a terror (not com-
mon), there is confusion and disorientation and only a vague
memory of fear. Night terrors are common in children with 30–
40% having at least one episode and repeated episodes in about
5%. The peak age for these is at about 2–7 years with a gradual
diminution up to early adolescence (Stallman et al., 2018). In
some cases they persist into adult life; the prevalence in adults is
unknown. Almost all adult patients have had night terrors or
sleepwalking as a child (Crisp, 1996) there is a strong genetic
component (Nguyen et al., 2008), and night terrors and sleep-
walking in the same patient is fairly common. Sleepwalking
alone probably has 15–20% lifetime prevalence. The main symp-
tom is of automatic behaviour at night with the sufferer unre-
sponsive to surroundings and other people. The behaviour is
most commonly walking around, but can include other behav-
iours which are highly familiar to the subject such as dressing,
washing, making tea, arranging objects in the house. Some cases
of sleepwalking seem related to use of certain drugs e.g. alcohol
and hypnotics, especially zolpidem and triazolam, opiates (pos-
sibly related to sleep-disordered breathing) (Pressman et al.,
2007), or other sleep disorders such as sleep apnoea. It is rare for
affected individuals to present for treatment, except if they have
injured themselves or a partner, have put themselves into poten-
tial danger, or have excessive daytime fatigue because of night
time disturbance. Another reason for presentation is anxiety and
disruption of sleep of partner, family or housemates.
Sleep paralysis, nightmares and REM sleep behaviour disor-
der (RBD) are disorders arising from REM sleep. Sleep paralysis
and nightmares are recalled by the patient. REM behaviour epi-
sodes are sometimes recalled but more often only apparent to the
bed partner.
Sleep paralysis is a brief state of involuntary immobility usu-
ally occurring on waking from a night’s sleep or a nap (more
rarely at sleep onset). It is often accompanied by dream imagery,
sometimes of a frightening kind, and sometimes a feeling of
chest tightness. It is attributed to waking abruptly from a REM
sleep episode with the REM atonia persisting briefly. It appears
to be more common in those with narcolepsy, and in those with
irregular sleep-wake routine and after drinking alcohol.
RBD is a disorder first described in the late 1980s with violent
complex behaviour at night. There are two sleep abnormalities:
lack of atonia during REM sleep (which can be quantified using
video-polysomnography), and increased vividness and/or nasty
content of dreams. The violent behaviour is described as ‘acting
out of dreams’, made possible by the lack of the normal muscle
paralysis in REM sleep causes injury to self or bed partner in up
to 70% of patients. Its incidence is estimated at 0.5–1% of those
over 55 years), occurs in older people with a steady rise after 55
years and has a male preponderance in older patients. It is now
well recognised as the most robust prodromal, non-motor symp-
tom of a subsequent neurodegeneration, typically an alpha synu-
cleinopathy. Several cohorts under long term follow-up have
shown that 50% at five years and 91% at 15 years will have
developed another neurodegenerative problem. It is often associ-
ated with Parkinson’s disease (PD) (it is seen in up to 50% of PD
patients), Lewy body dementia (~70%), multiple system atrophy
(>90%). RBD often precedes other symptoms of neurodegenera-
tion by several years (Iranzo et al., 2014).
Diagnosis of parasomnias
Assessment of parasomnia may be possible with a detailed his-
tory from patient or witness, but in general for adequate diag-
nosis, referral to a specialist sleep centre for polysomnography
and video recording may be necessary especially for RBD
where loss of REM atonia is seen.
Treatment of parasomnias
There is little high-level evidence for treatments in these disor-
ders. There are no controlled trials of treatment of non-REM
parasomnias in adults (Harris and Grunstein, 2009). Priorities
are to minimise possible trigger factors such as noise, frighten-
ing films, caffeine, alcohol or meals late at night; and to make
sure there is a stable and adequate sleep-wake schedule. It is
important to safeguard against harm to the patient, such as by
locking windows, bolting doors, or sleeping on the ground
floor, and safety of the bed partner or nearby children also
requires attention.
Drug treatment decisions should be based on the frequency
and severity of events. Clonazepam in doses up to 3 mg per night
has been reported to be effective (case series, n=69) (Schenck
and Mahowald, 1996). Smaller case series have reported good
effects of paroxetine (Wilson et al., 1997) and imipramine
(Cooper, 1987) (both effective immediately) and there are several
case series of hypnotherapy in sleepwalkers (Becker PM, 2015).
A randomised controlled study of three weeks’ treatment with
5-hydroxytryptophan in children found evidence of efficacy at
six-month follow-up (Bruni et al., 2004).
For nightmares, psychological treatments are effective and
these focus on exposure – writing down dreams – or guided
imagery, pleasant images, and ‘changing the ‘ending’ (Burgess
et al., 1998; Hansen et al., 2013; Krakow et al., 2001). There is
good evidence of beneficial effects of the alpha-1 adrenergic
blocker prazosin in reducing nightmares related to PTSD in both
military and civilian settings and in the paediatric population
(Keeshin et al., 2017; Nadorff et al., 2014). Nightmares have
been reported to be triggered or worsened by many drug treat-
ments including cholinesterase inhibitors, beta-blockers, SRIs
(especially paroxetine) levodopa, and following withdrawal from
drugs for depression.
938 Journal of Psychopharmacology 33(8)
For RBD, all data on treatment comes from retrospective
case notes review and for melatonin, a single, small crossover
randomised trial. There are no prospective or controlled studies
of clonazepam for RBD, but large case series suggest a good
effect when used at doses between 1–4 mg (Aurora et al., 2010;
Boeve et al., 2004) in reducing number of episodes and injury
during them. Dose-limiting side effects are common in those
with dementia, disorders of gait or balance, or concomitant
OSAS (Anderson and Schneerson, 2009). Beneficial effects
have been reported for melatonin 3–12 mg but with fewer
adverse events. A single, small RCT has shown benefit for mel-
atonin including in quantitative measure of REM atonia (Kunz
and Mahlberg, 2010). Single case studies and small series have
reported beneficial effects of clonidine (Nash et al., 2003),
donepezil (Massironi et al., 2003), and sodium oxybate (Kosky
et al., 2008).
Drugs which can worsen RBD or provoke its symptoms
include SRIs, venlafaxine, mirtazapine, bisoprolol and tramadol
(Gagnon et al., 2006).
Special populations
Sleep disorders in women: effects of
menopause and pregnancy
Menopause. Insomnia increases as women approach and pass
through the menopause ( Bixler et al., 2009; Kuh et al., 1997;
Owens and Matthews, 1998). Post-menopausal women have a
longer sleep latency and decreased slow-wave sleep. This is due
to a variety of reasons – climacteric symptoms e.g. hot flushes
due to hormonal changes and psychiatric disorders are most often
cited. Hormone therapy appears to protect women from these
changes (Bixler et al., 2009).
CBTi has been shown to be effective in insomnia in this
group, with long lasting benefits up to six months post-treatment
(McCurry et al., 2016). There are modest benefits from some but
not all studies of pharmacotherapy with SRIs including escitalo-
pram, citalopram and venlafaxine although short duration of
follow-up limits the conclusions that can be drawn from the
studies (Ensrud et al., 2012; Davari-Tahna et al., 2016).
Post-menopause, there is also a rise in the incidence of
sleep-disordered breathing (Young et al., 2003) (Bixler et al.,
2001). Post-menopausal women with sleep-disordered breath-
ing are more likely to complain of depression and insomnia
than men with a similar degree of OSAS (Shepertycky et al.,
2005).
Recommendations
Clinicians should appreciate that there is a rise in inci-
dence of sleep-disordered breathing after the menopause and
that clinical presentation, often including insomnia, in women
is different to men (D).
The use of hormone therapy should involve informed indi-
vidualised treatment of symptoms, looking at risks and ben-
efits in light of recent studies (A).
Follow recommendations for insomnia in other sections (A).
Pregnancy. Many women report poor sleep during preg-
nancy with the reasons varying depending on the trimester. In
the first trimester, nausea, backache and urinary frequency
can cause sleep disturbance. The second trimester tends to be
easier but foetal movements and heartburn may be trouble-
some. By the third trimester, sleep is more disturbed with
complaints again of urinary frequency, backache in addition
to cramps, itching and unpleasant dreams. Most women fall
asleep fairly easily but wake more frequently (Sedov et al.,
2018).
If a patient suffers from intractable insomnia and a phar-
macological agent is required, it is helpful to note that zolpi-
dem and diphenhydramine are in FDA class B (foetal harm
possible, but unlikely; no evidence of foetal harm in animal
studies) (for review see (Pien and Schwab, 2004). However,
non-selective histamine antagonists such as diphenhydramine
can exacerbate restless legs syndrome (RLS) and have anticho-
linergic actions. Zolpidem may be preferable as it is short-
acting and does not have anticholinergic side effects. The
hypnotics temazepam and zopiclone have not been associated
with any increase in congenital malformations (Ban et al.,
2014).
RLS is common in pregnancy, with a prevalence between
15–25%, peaking in the third trimester with improvement in
the last two weeks and often resolving post-partum. It is some-
times associated with anaemia (Hubner et al., 2013; Neau
et al., 2010). Iron replacement is safe and may be effective
based on small case series, with carbidopa/levodopa or clonaz-
epam only recommended in severe and refractory cases
(Picchietti et al., 2015).
Snoring and sleep disordered breathing, especially in obese
subjects, is increasingly recognised, is associated with worse foe-
tal outcomes and affects up to 8% of women by the third trimes-
ter. (O’Brien et al., 2014).
Recommendations
Good sleep hygiene and lifestyle (D).
Manage general pregnancy associated complaints e.g.
decrease fluid intake, pillow support (D).
The effects of CBTi in pregnancy have only been assessed
in one small open-label study, but this approach seems sensi-
ble (B).
Recognise RLS by careful history and investigations if
necessary.
Dopamine agonists are contraindicated (FDA category C
or greater).
Iron supplementation has been shown to be effective in
RLS. Supplementation is suggested even if levels are not
low (D).
Keep caffeine low as it can exacerbate RLS (D).
Mild-moderate exercise in the early evening, stretching,
massage (D).
If patient suffers from intractable insomnia and a phar-
macological agent is required, zolpidem or zopiclone should
be used short-term after discussion on potential risks and
benefits (D).
Wilson et al. 939
What is known about treatment of insomnia in older adults:
CBTi is effective in older adults and is associated with minimal
side effects (Ia).
Eszopiclone, suvorexant and doxepin improve global and sleep
outcomes (Ib).
Prolonged release melatonin given for three weeks improves sleep
onset latency and sleep quality in patients over 55 years (Ib).
Drugs with sedative effects increase the risk of falls in older
adults (III).
Benzodiazepine hypnotics have an unfavourable risk/benefit
ratio (B).
Insomnia increases the risk of falls and fractures in nursing
homes independently of medication (III).
What is not known:
Is there an effective treatment for sleeplessness in older adults
with dementia?
Treatment of insomnia in older adults
Insomnia in elderly patients often responds well to CBTi (see
psychological treatment section above). Meta-analyses compar-
ing CBT outcomes in older adults (55 years plus), have reported
moderate to large effect sizes, whether or not insomnia is comor-
bid with other disorders (Alessi and Vitiello, 2015).
A meta-analysis (Glass et al., 2005) concluded that benzo-
diazepine hypnotics had an unfavourable risk/benefit ratio in
elderly patients, but the different methods of collection and cate-
gorisation of drug-related side effects in the studies included
makes them difficult to interpret. Individual randomised con-
trolled studies with short-acting Z drugs show little evidence of
adverse effects, particularly cognitive side effects in the morning.
However if a patient needs to rise within a few hours after taking
a benzodiazepine agonist drug there may be undesired effects on
motor control. Falls are increased after sedatives and hypnotics,
drugs for depression or psychosis, benzodiazepines, nonsteroidal
anti-inflammatory drugs and calcium channel antagonists
(Woolcott et al., 2009) but this study did not specify daytime/
night-time use. There is a 2.5-fold risk of falls in hospital after
zolpidem (Rhalimi et al., 2009), but in nursing homes the situation
may be different; in a large study (Avidan et al., 2005) insomnia
itself, but not hypnotic use, was associated with an increase in
falls and hip fractures. Therefore the development of sleep-pro-
moting drugs without motor side effects has been welcomed.
Prolonged release melatonin has been shown to reduce sleep
onset latency and increase subjective sleep quality in patients
over 55 years (Lemoine et al., 2007; Wade et al., 2007, 2011); its
effects are modest but it has no known motor side effects.
The antihistamine drug doxepin (in very low dose 3 mg, see
above) has been found effective in insomnia in the older patient, in
particular decreasing awakenings in the latter half of the night with-
out daytime effects (Lankford et al., 2012; Krystal et al., 2013).
A Cochrane review, looking at pharmacotherapies for sleep
disturbances in dementia (McCleery et al., 2016) found a lack of
evidence to help guide drug treatment of sleep problems in
dementia. Few RCTs have been conducted. There is some evi-
dence to support the use of low dose trazodone (Camargos,
2014). This is an area with a high need for pragmatic trials, par-
ticularly of those drugs that are in common clinical use for sleep
problems in dementia.
Recommendations
CBTi is effective and should be offered as a first line treat-
ment where available (A).
When a hypnotic is indicated in patients over 55 years
prolonged release melatonin should be tried first (B).
If a GABA-A hypnotic is used then a shorter half-life will
minimise unwanted hangover effects (A).
Sleep problems in children
Sleep problems are commonly associated with certain genetic
and neuro-developmental problems seen in childhood including
ADHD, autism, learning difficulties and epilepsy. Training and
awareness of paediatric sleep disorders is poor and accurate
diagnoses and hence appropriate treatments are often delayed.
Settling and sleep maintenance problems may be exacerbated
by a sleep disorder such as obstructive sleep apnoea or RLS.
Evidence from systematic review suggests that most sleep dis-
orders in childhood respond well to behavioural treatments
(Mindell et al., 2006). Appropriate sleep hygiene measures and
more specific techniques of extinction, or graduated extinction, are
all more effective than placebo at improving sleep and reducing the
number of weekly night wakes in otherwise healthy children who
regularly wake up in the night (Ramchandani et al., 2000). These
interventions hold for both typically developing children and chil-
dren with learning difficulties and sleep problems. These interven-
tions may not change sleep parameters in the child, but instead
improve outcomes related to impact on parents and other carers.
The sedative side effects of antihistamines may speed up
behavioural programmes over short periods (France et al., 1991)
but seem not to work without behavioural interventions; in a
placebo-controlled double-blind trial in infants aged 6–27 months
the same authors found no significant effect of 15 mg or 30 mg
trimeprazine tartrate, and concluded that it is not recommended
as a pharmacological treatment for infant sleep disturbance
unless as an adjunct to a behavioural therapy program (France
et al., 1999). Clinically the short term use of an H1 blocker for
transient or extreme insomnia is frequently employed: however,
tolerance can develop quickly and some children can experience
dramatic and paradoxical over-arousal. The TIRED RCT specifi-
cally investigated the use of diphenhydramine in infants aged
from 6–15 months and found it was no more effective than pla-
cebo in reducing night-time awakening (Merenstein et al., 2006).
It is important to consider the effects of these medications at neu-
rotransmitter systems other than H1 receptors, particularly in
relation to side effects due to anticholinergic or dopamine antag-
onist action.
What is known:
Most sleep settling and maintenance problems in childhood
respond well to behavioural treatments (I).
Melatonin reduces long sleep latency (following appropriate
behavioural interventions) in children with sleep onset
insomnia or delayed sleep phase syndrome and learning
difficulties, autism and attention deficit hyperactivity disorder
(ADHD) (II).
What is not known:
What are the long-term effects of melatonin?
940 Journal of Psychopharmacology 33(8)
The evidence supporting use of melatonin to reduce long
sleep latency (following appropriate behavioural interventions)
in populations of children with idiopathic sleep onset insomnia
(Smits et al., 2003) or delayed sleep phase syndrome and learning
difficulties, autism and ADHD (Gringras et al., 2017; Maras
et al., 2018; Rossignol and Frye, 2011; van der Heijden et al.,
2007) is increasingly robust.
There is evidence to support a behavioural intervention both
before a trial of melatonin (Gringras et al., 2012) (as many will
respond without requiring melatonin), and for continuing a
behavioural intervention whilst administering melatonin. The
combination of both has been shown to be more effective than
either one alone (Cortesi et al., 2012).
The most recent randomised controlled study showed a paedi-
atric mini-pill sustained release melatonin at a dose of 2–10 mg
was well-tolerated, efficacious and safe compared to placebo for
treatment of insomnia in children with autistic spectrum disorder
(ASD). These studies have resulted in the first licensed sleep med-
ication for insomnia in children with ASD. They showed clini-
cally meaningful improvements in total sleep time (TST), duration
of uninterrupted sleep (longest sleep episode) and sleep latency
(SL) with corresponding behavioural improvements for the chil-
dren, and improved quality of life measures in their parents over a
two-year period. (Gringras et al., 2017; Maras et al., 2018)
Melatonin at doses between 0.5–12 mg is commonly used as a
sleep-promoting agent in children undergoing procedures such as
EEG, as an alternative to sleep deprivation to induce drowsiness
and sleep that does not affect the EEG morphology. A melatonin-
induced sleep EEG was as useful as a sleep-deprived EEG but chil-
dren’s behaviour on the day of the melatonin-induced sleep EEG
recording was more acceptable to parents (Wassmer et al., 2001).
Clonidine is an antihypertensive agent with sedative side
effects that is licensed for children with ADHD and improves
sleep maintenance in some children. The therapeutic window is
narrow, both for adverse effects on sleep architecture and tolera-
bility. Tolerance to the sleep-inducing effects develops over time
leading to the need for increased doses with concomitant risk of
adverse effects.
Chloral hydrate and triclofos are still popular hypnotics for
children but have a very long half-life and considerable potential
for ‘hang-over effects in children. The half-life of chloral hydrate
itself is short (a few minutes), but the half-lives of its active
metabolites are longer, being 8–12 h for trichloroethanol and 67
h for trichloroacetic acid. Toxicity is an important concern due to
central nervous system depressant action, arrhythmogenic poten-
tial and stomach irritation.
Recommendations
Behavioural strategies should be tried first in children
with disturbed sleep (A).
Melatonin improves sleep in children with ASDs (A).
Melatonin administration can be used to advance sleep
onset to normal values in children with ADHD who are not on
stimulant medication (B).
Sleep disturbance in adults with intellectual
disability
There is little clarity in the definition of sleep problems in adults
with intellectual disability. This is primarily because it is
difficult to obtain subjective measures from the patient who may
be unable to communicate verbally or even perceive that they
are having a problem. Reports of sleep difficulties tend to be
from carers as they struggle to cope with issues which only seem
to be exacerbated when they, and the person they care for, expe-
rience sleep disturbance. There is a compelling need to develop
a more accurate, standardised measure of sleep for this popula-
tion (Meltzer and Mindell, 2014), to obtain essential information
about prevalence.
Difficulties in assessing sleep disorders include diagnostic
overshadowing, where behaviours such as daytime sleepiness,
inattention and challenging behaviour are regarded as sympto-
matic of the intellectual disability as opposed to being indicative
of a sleep disorder. In these circumstances therefore, clinicians
may fail to consider the possibility of sleep disturbance and thus
neglect to undertake more detailed investigations.
The situation is further compounded by the fact that health
and behavioural problems can increase as sleep problems
develop. For example, the inability to problem solve combined
with daytime somnolence exacerbates cognitive processing prob-
lems in those already compromised intellectually (Carr et al.,
2003; Symons et al., 2000).
Within this population there are additional high-risk groups
including those with co-morbidities such as Down syndrome
where people may present with sleep related breathing disorders,
or Smith-Magenis syndrome where melatonin rhythm is inverted.
In general, there are a wide range of precipitating and per-
petuating factors to consider including maladaptive coping strat-
egies, the impact of medication e.g. drugs used in psychosis,
depression, epilepsy including side-effects and the impact of
polypharmacy on sleep and daytime functioning. For those in
institutional or residential living environments the environment
itself may contribute to disturbed or inconsistent sleep patterns,
as other patients/residents wake others up or staff shift patterns
determine wake/sleep times rather the individuals themselves.
Assessment. Sound clinical assessment should elicit any aetio-
logical or exacerbating factors which can be reversed. This may
be best undertaken by direct observation initially. Carers should
be supported to keep a structured 24-hour record of sleep pattern
and behaviour. Actigraphy or EEG may be useful when a sleep
disorder other than insomnia or settling difficulties is suspected,
but clinicians should be aware that the recording equipment may
not be tolerated by people with more moderate to severe levels of
intellectual disability. The possibility of a CRD should also be
excluded in individuals with additional visual impairment.
Treatment considerations. There is a varying degree of evi-
dence for treatments of sleep difficulties in this heterogeneous
population. Systematic review (van de Wouw et al., 2012) used
SIGN 50 methodology but no statistical analysis in 50 studies
using behavioural interventions in adults with intellectual dis-
abilities. They concluded there was some indication of the effec-
tiveness of behavioural interventions. In addition, they reported
that in some cases sleep problems were associated with challeng-
ing behaviour and medication.
The relatively small number of controlled studies in this area
give support to parental/carer education and modifying environ-
mental factors (Montgomery et al., 2004). Behavioural regimes
such as chronotherapy, bedtime fading, extinction, distancing/
desensitisation and sleep-wake scheduling (Wiggs and France,
Wilson et al. 941
2000; Gunning and Espie, 2003) may also prove bene ficial. The use
of light therapy has been described (Short and Carpenter, 1998).
There is little evidence for effectiveness of sleep-promoting
drugs, apart from melatonin. A meta-analysis (Braam et al., 2009)
indicated that melatonin (1–9 mg) decreases sleep latency and
number of wakes per night, and increases total sleep time in indi-
viduals with intellectual disabilities. There were few adverse
events in the relatively short-term studies included, however long
term safety requires further research.
Recommendations
Clinical assessment should describe sleep disturbance
and elicit aetiological and exacerbating factors (A).
Environmental, behavioural and educational approaches
should be used first line (A).
Melatonin is effective in improving sleep (A).
Treatment should be planned within a capacity/best inter-
ests framework (D).
Acknowledgements
Although the first author prepared the document for publication, all
authors contributed equally to the consensus.
Declaration of conflicting interests
The author(s) declared no potential conflicts of interest with respect to
the research, authorship, and/or publication of this article.
Funding
The authors received no financial support for the research, authorship,
and/or publication of this article.
References
Abe T, Inoue Y, Komada Y, et al. (2011) Relation between morningness-
eveningness score and depressive symptoms among patients with
delayed sleep phase syndrome. Sleep Med 12: 680–684.
Adam K and Oswald I (1986) The hypnotic effects of an antihistamine:
Promethazine. Br J Clin Pharmacol 22: 715–717.
Alessi C and Vitiello MV (2015) Insomnia (primary) in older people:
Non-drug treatments. BMJ Clin Evid 2015: 2302.
Alford C, Rombaut N, Jones J, et al. (1992) Acute effects of hydroxyzine
on nocturnal sleep and sleep tendency the following day: A C-EEG
study. Hum Psychopharmacol Clin Exp 7: 25–35.
Altena E, Van Der Werf YD, Strijers RL, et al. (2008) Sleep loss affects
vigilance: Effects of chronic insomnia and sleep therapy. J Sleep Res
17: 335–343.
Ancoli-Israel S, Krystal AD, McCall WV, et al. (2010) A 12-week, ran-
domized, double-blind, placebo-controlled study evaluating the effect
of eszopiclone 2 mg on sleep/wake function in older adults with pri-
mary and comorbid insomnia. Sleep 33: 225–234.
Ancoli-Israel S, Richardson GS, Mangano RM, et al. (2005) Long-term
use of sedative hypnotics in older patients with insomnia. Sleep Med
6: 107–113.
Anderson KN and Shneerson JM (2009) Drug treatment of REM sleep
behavior disorder: The use of drug therapies other than clonazepam.
J Clin Sleep Med 5: 235–239.
Anderson SL and Vande Griend JP (2014) Quetiapine for insomnia: A
review of the literature. Am J Health Syst Pharm 71: 394–402.
Armitage R, Cole D, Suppes T, et al. (2004) Effects of clozapine on sleep
in bipolar and schizoaffective disorders. Prog Neuropsychopharma-
col Biol Psychiatry 28: 1065–1070.
Attenburrow M, Dowling B and Sharpley A (1996) Case-control study
of evening melatonin concentration in primary insomnia. Br Med J
312: 1263–1264.
Auger RR, Burgess HJ, Emens JS, et al. (2015) Clinical practice guideline
for the treatment of intrinsic circadian rhythm sleep-wake disorders:
Advanced sleep-wake phase disorder (ASWPD), delayed sleep-wake
phase disorder (DSWPD), non-24-hour sleep-wake rhythm disorder
(N24SWD), and irregular sleep-wake rhythm disorder (ISWRD). An
update for 2015: an American academy of sleep medicine clinical
practice guideline. J Clin Sleep Med 11: 1199–1236.
Aurora RN, Zak RS, Maganti RK, et al. (2010) Best practice guide for
the treatment of REM sleep behavior disorder (RBD). J Clin Sleep
Med 6: 85–95.
Avidan AY, Fries BE, James ML, et al. (2005) Insomnia and hypnotic
use, recorded in the minimum data set, as predictors of falls and hip
fractures in Michigan nursing homes. J Am Geriatr Soc 53: 955–962.
Baglioni C and Riemann D (2012) Is chronic insomnia a precursor to
major depression? Epidemiological and biological findings. Curr
Psychiatry Rep 14: 511–518.
Baglioni C, Spiegelhalder K, Lombardo C, et al. (2010) Sleep and emo-
tions: A focus on insomnia. Sleep Med Rev 14: 227–238.
Ban L, West J, Gibson JE, et al. (2014) First trimester exposure to anxio-
lytic and hypnotic drugs and the risks of major congenital anomalies: A
United Kingdom population-based cohort study. PLoS One 9: e100996.
Barbone F, McMahon AD, Davey PG, et al. (1998) Association of road-
traffic accidents with benzodiazepine use. Lancet 352: 1331–1336.
Baskaran A, Summers D, Willing SL, et al. (2013) Sleep architecture
in ziprasidone-treated bipolar depression: A pilot study. Ther Adv
Psychopharmacol 3: 139–149.
Bastien CH, LeBlanc M, Carrier J, et al. (2003) Sleep EEG power spectra,
insomnia, and chronic use of benzodiazepines. Sleep 26: 313–317.
Bateson AN (2002) Basic pharmacologic mechanisms involved in benzo-
diazepine tolerance and withdrawal. Curr Pharm Des 8: 5–21.
Becker PM (2015) Hypnosis in the management of sleep disorders. Sleep
Med Clin 10: 85–92.
Belanger L, Morin CM, Bastien C, et al. (2005) Self-efficacy and compli-
ance with benzodiazepine taper in older adults with chronic insom-
nia. Health Psychol 24: 281–287.
Belleville G, Guay C, Guay B, et al. (2007) Hypnotic taper with or with-
out self-help treatment of insomnia: A randomized clinical trial. J
Consult Clin Psychol 75: 325–335.
Bertisch SM, Herzig SJ, Winkelman JW, et al. (2014) National use of
prescription medications for insomnia: NHANES 1999–2010. Sleep
37: 343–349.
Billiard M, Bassetti C, Dauvilliers Y, et al. (2006) EFNS guidelines on
management of narcolepsy. Eur J Neurol 13: 1035–1048.
Bixler EO, Papaliaga MN, Vgontzas AN, et al. (2009) Women sleep
objectively better than men and the sleep of young women is more
resilient to external stressors: Effects of age and menopause. J Sleep
Res 18: 221–228.
Bixler EO, Vgontzas AN, Lin HM, et al. (2001) Prevalence of sleep-
disordered breathing in women: Effects of gender. Am J Respir Crit
Care Med 163: 608–613.
Boeve BF, Silber MH and Ferman TJ (2004) REM sleep behavior disor-
der in Parkinson's disease and dementia with Lewy bodies. J Geriatr
Psychiatry Neurol 17: 146–157.
Boyle J, Groeger JA, Paska W, et al. (2012) A method to assess the
dissipation of the [corrected] residual effects of [corrected] hyp-
notics: Eszopiclone versus zopiclone. J Clin Psychopharmacol 32:
704–709.
Braam W, Smits MG, Didden R, et al. (2009) Exogenous melatonin for
sleep problems in individuals with intellectual disability: A meta-
analysis. Dev Med Child Neurol 51: 340–349.
Breslau N, Roth T, Rosenthal L, et al. (1996) Sleep disturbance and psy-
chiatric disorders: A longitudinal epidemiological study of young
adults. Biol Psychiatry 39: 411–418.
942 Journal of Psychopharmacology 33(8)
Bruni O, Ferri R, Miano S, et al. (2004) L -5-Hydroxytryptophan treat-
ment of sleep terrors in children. Eur J Pediatr 163: 402–407.
Burgess M, Gill M and Marks I (1998) Postal self-exposure treatment of
recurrent nightmares. Randomised controlled trial. Br J Psychiatry
172: 257–262.
Buscemi N, Vandermeer B, Friesen C, et al. (2005) Manifestations and
management of chronic insomnia in adults. Evid Rep Technol Assess
(Summ.): 125: 1–10.
Buscemi N, Vandermeer B, Friesen C, et al. (2007) The efficacy and
safety of drug treatments for chronic insomnia in adults: A meta-
analysis of RCTs. J Gen Intern Med 22: 1335–1350.
Cajochen C, Krauchi K and Wirz-Justice A (2003) Role of melatonin in
the regulation of human circadian rhythms and sleep. J Neuroendo-
crinol 15: 432–437.
Calem M, Bisla J, Begum A, et al. (2012) Increased prevalence of insom-
nia and changes in hypnotics use in England over 15 years: Analysis
of the 1993, 2000, and 2007 National Psychiatric Morbidity Surveys.
Sleep 35: 377–384.
Camargos EF, Louzada LL, Quintas JL, et al. (2014) Trazodone improves
sleep parameters in Alzheimer disease patients: A randomized, dou-
ble-blind, and placebo-controlled study. Am J Geriatr Psychiatry 22:
1565–1574.
Cappuccio FP, D'Elia L, Strazzullo P, et al. (2010) Quantity and quality
of sleep and incidence of type 2 diabetes: A systematic review and
meta-analysis. Diabetes Care 33: 414–420.
Carr EG, Magito McLaughlin D, Giacobbe-Grieco T, et al. (2003) Using
mood ratings and mood induction in assessment and intervention for
severe problem behavior. Am J Ment Retard 108: 32–55.
Chang PP, Ford DE, Mead LA, et al. (1997) Insomnia in young men and
subsequent depression. The Johns Hopkins Precursors Study. Am J
Epidemiol 146: 105–114.
Cheng SK and Dizon J (2012) Computerised cognitive behavioural ther-
apy for insomnia: A systematic review and meta-analysis. Psycho-
ther Psychosom 81: 206–216.
Cohen DA, Wang W, Klerman EB, et al. (2010) Ramelteon prior to a
short evening nap impairs neurobehavioral performance for up to 12
hours after awakening. J Clin Sleep Med 6: 565–571.
Cohrs S, Meier A, Neumann AC, et al. (2005) Improved sleep continuity
and increased slow wave sleep and REM latency during ziprasidone
treatment: A randomized, controlled, crossover trial of 12 healthy
male subjects. J Clin Psychiatry 66: 989–996.
Cohrs S, Rodenbeck A, Guan Z, et al. (2004) Sleep-promoting properties
of quetiapine in healthy subjects. Psychopharmacology (Berl) 174:
421–429.
Connor J, Norton R, Ameratunga S, et al. (2002) Driver sleepiness and
risk of serious injury to car occupants: Population based case control
study. BMJ 324: 1125.
Cooper AJ (1987) Treatment of coexistent night-terrors and somnam-
bulism in adults with imipramine and diazepam. J Clin Psychiatry
48: 209–210.
Cortesi F, Giannotti F, Sebastiani T, et al. (2012) Controlled-release
melatonin, singly and combined with cognitive behavioural ther-
apy, for persistent insomnia in children with autism spectrum
disorders: A randomized placebo-controlled trial. J Sleep Res 21:
700–709.
Crisp AH (1996) The sleepwalking/night terrors syndrome in adults.
Postgrad Med J 72: 599–604.
Daley M, Morin CM, LeBlanc M, et al. (2009) Insomnia and its relation-
ship to health-care utilization, work absenteeism, productivity and
accidents. Sleep Med 10: 427–438.
Davari-Tanha F, Soleymani-Farsani M, Asadi M, et al. (2016) Com-
parison of citalopram and venlafaxine’s role in treating sleep
disturbances in menopausal women, a randomized, double-blind,
placebo-controlled trial. Arch Gynecol Obstet 293: 1007–1013.
Dijk DJ and von Schantz M (2005) Timing and consolidation of human
sleep, wakefulness, and performance by a symphony of oscillators. J
Biol Rhythms 20: 279–290.
Doble A, Martin IL and Nutt DJ (2004) Calming the Brain: Benzodiaze-
pines and Related Drugs from Laboratory to Clinic. London: Martin
Dunitz Limited.
Dowling GA, Burr RL, Van Someren EJ, et al. (2008) Melatonin and
bright-light treatment for rest-activity disruption in institutionalized
patients with Alzheimer’s disease. J Am Geriatr Soc 56: 239–246.
Edinger JD, Means MK, Carney CE, et al. (2008) Psychomotor perfor-
mance deficits and their relation to prior nights’ sleep among indi-
viduals with primary insomnia. Sleep 31: 599–607.
Ensrud KE, Joffe H, Guthrie KA, et al. (2012) Effect of escitalopram
on insomnia symptoms and subjective sleep quality in healthy peri-
menopausal and postmenopausal women with hot flashes: A ran-
domized controlled trial. Menopause 19: 848–855.
Espie CA (2002) Insomnia: Conceptual issues in the development, per-
sistence, and treatment of sleep disorder in adults. Annu Rev Psychol
53: 215–243.
Espie CA, Broomfield NM, MacMahon KM, et al. (2006) The attention-
intention-effort pathway in the development of psychophysiologic
insomnia: A theoretical review. Sleep Med Rev 10: 215–245.
Espie CA, Emsley R, Kyle SD, et al. (2018a) Effect of digital cognitive
behavioral therapy for insomnia on health, psychological well-being,
and sleep-related quality of life: A randomized clinical trial. JAMA
Psychiatry. DOI: 10.1001/jamapsychiatry.2018.2745
Espie CA, Farias Machado P, Carl JR, et al. (2018b) The sleep condition
indicator: Reference values derived from a sample of 200 000 adults.
J Sleep Res 27: e12643.
Espie CA, Kyle SD, Hames P, et al. (2012) The daytime impact of DSM-5
insomnia disorder: Comparative analysis of insomnia subtypes from
the Great British Sleep Survey. J Clin Psychiatry 73: e1478–1484.
Espie CA, Kyle SD, Hames P, et al. (2014) The sleep condition indicator:
A clinical screening tool to evaluate insomnia disorder. BMJ Open 4:
e004183.
Everitt H, Baldwin DS, Stuart B, et al. (2018) Antidepressants for insom-
nia in adults. Cochrane Database Syst Rev 5: CD010753.
Farkas RH, Unger EF and Temple R (2013) Zolpidem and driving impair-
ment—identifying persons at risk. N Engl J Med 369: 689–691.
Flynn-Evans EE, Shekleton JA, Miller B, et al. (2017) Circadian phase
and phase angle disorders in primary insomnia. Sleep 40(12): 1–11.
Ford DE and Kamerow DB (1989) Epidemiological study of sleep dis-
turbances and psychaitric disorders: An opportunity for prevention?
JAMA 262: 1479–1484.
Fortier-Brochu E, Beaulieu-Bonneau S, Ivers H, et al. (2012) Insomnia
and daytime cognitive performance: A meta-analysis. Sleep Med Rev
16: 83–94.
France KG, Blampied NM and Wilkinson P (1991) Treatment of infant
sleep disturbance by trimeprazine in combination with extinction. J
Dev Behav Pediatr 12: 308–314.
France KG, Blampied NM and Wilkinson P (1999) A multiple-baseline,
double-blind evaluation of the effects of trimeprazine tartrate on
infant sleep disturbance. Exp Clin Psychopharmacol 7: 502–513.
Freeman D, Sheaves B, Goodwin GM, et al. (2017) The effects of
improving sleep on mental health (OASIS): A randomised controlled
trial with mediation analysis. Lancet Psychiatry 4: 749–758.
Gagnon JF, Postuma RB and Montplaisir J (2006) Update on the phar-
macology of REM sleep behavior disorder. Neurology 67: 742–747.
Gao K, Mackle M, Cazorla P, et al. (2013) Comparison of somnolence
associated with asenapine, olanzapine, risperidone, and haloperidol
relative to placebo in patients with schizophrenia or bipolar disorder.
Neuropsychiatr Dis Treat 9: 1145–1157.
Gimenez S, Clos S, Romero S, et al. (2007) Effects of olanzapine, ris-
peridone and haloperidol on sleep after a single oral morning dose in
healthy volunteers. Psychopharmacology (Berl) 190: 507–516.
Glass J, Lanctot KL, Herrmann N, et al. (2005) Sedative hypnotics in
older people with insomnia: Meta-analysis of risks and benefits. BMJ
331: 1169.
Greenblatt DJ, Legangneux E, Harmatz JS, et al. (2006) Dynamics and
kinetics of a modified-release formulation of zolpidem: Comparison
Wilson et al. 943
with immediate-release standard zolpidem and placebo. J Clin Phar-
macol 46: 1469–1480.
Gringras P, Gamble C, Jones AP, et al. (2012) Melatonin for sleep prob-
lems in children with neurodevelopmental disorders: Randomised
double masked placebo controlled trial. BMJ 345: e6664.
Gringras P, Nir T, Breddy J, et al. (2017) Efficacy and safety of pediatric
prolonged-release melatonin for insomnia in children with autism
spectrum disorder. J Am Acad Child Adolesc Psychiatry 56: 948–957
e944.
Gross G, Xin X and Gastpar M (1991) Trimipramine: Pharmacological
reevaluation and comparison with clozapine. Neuropharmacology
30: 1159–1166.
Gunning MJ and Espie CA (2003) Psychological treatment of reported
sleep disorder in adults with intellectual disability using a multiple
baseline design. J Intellect Disabil Res 47: 191–202.
Hafner M, Stepanek M, Taylor J, et al. (2017) Why sleep matters-the
economic costs of insufficient sleep: A cross-country comparative
analysis. Rand Health Q 6: 11.
Haimov I (2001) Melatonin rhythm abnormalities and sleep disorders in
the elderly. CNS Spectr 6: 502–506.
Hajak G, Hedner J, Eglin M, et al. (2009) A 2-week efficacy and safety
study of gaboxadol and zolpidem using electronic diaries in primary
insomnia outpatients. Sleep Med 10: 705–712.
Hajak G, Rodenbeck A, Voderholzer U, et al. (2001) Doxepin in the
treatment of primary insomnia: A placebo-controlled, double-blind,
polysomnographic study. J Clin Psychiatry 62: 453–463.
Hansen K, Hofling V, Kroner-Borowik T, et al. (2013) Efficacy of psy-
chological interventions aiming to reduce chronic nightmares: A
meta-analysis. Clin Psychol Rev 33: 146–155.
Harris M and Grunstein RR (2009) Treatments for somnambulism in
adults: Assessing the evidence. Sleep Med Rev 13: 295–297.
Herring WJ, Connor KM, Ivgy-May N, et al. (2016) Suvorexant in
patients with insomnia: Results from two 3-month randomized con-
trolled clinical trials. Biol Psychiatry 79: 136–148.
Hicks JA, Argyropoulos SV, Rich AS, et al. (2002) Randomised con-
trolled study of sleep after nefazodone or paroxetine treatment in out-
patients with depression. Br J Psychiatry 180: 528–535.
Hirano A, Shi G, Jones CR, et al. (2016) A Cryptochrome 2 mutation
yields advanced sleep phase in humans. eLife 5 pii: e16695.
Hoque R and Chesson AL, Jr (2010) Pharmacologically induced/exacer-
bated restless legs syndrome, periodic limb movements of sleep, and
REM behavior disorder/REM sleep without atonia: Literature review,
qualitative scoring, and comparative analysis. J Clin Sleep Med 6:
79–83.
Horne JA and Ostberg O (1976) A self-assessment questionnaire to deter-
mine morningness-eveningness in human circadian rhythms. Int J
Chronobiol 4: 97–110.
Hubner A, Krafft A, Gadient S, et al. (2013) Characteristics and determi-
nants of restless legs syndrome in pregnancy: A prospective study.
Neurology 80: 738–742.
Iranzo A, Fernandez-Arcos A, Tolosa E, et al. (2014) Neurodegenerative
disorder risk in idiopathic REM sleep behavior disorder: Study in
174 patients. PLoS One 9: e89741.
Ivgy-May N, Roth T, Ruwe F, et al. (2015) Esmirtazapine in non-elderly
adult patients with primary insomnia: Efficacy and safety from a
2-week randomized outpatient trial. Sleep Med 16: 831–837.
Jia F, Goldstein PA and Harrison NL (2009) The modulation of synaptic
GABA(A) receptors in the thalamus by eszopiclone and zolpidem. J
Pharmacol Exp Ther 328: 1000–1006.
Jindal RD, Buysse DJ and Thase ME (2004) Maintenance treatment of
insomnia: What can we learn from the depression literature? Am J
Psychiatry 161: 19–24.
Kalmbach DA, Cuamatzi-Castelan AS, Tonnu CV, et al. (2018) Hyper-
arousal and sleep reactivity in insomnia: Current insights. Nat Sci
Sleep 10: 193–201.
Karsten J, Hagenauw LA, Kamphuis J, et al. (2017) Low doses of
mirtazapine or quetiapine for transient insomnia: A randomised,
double-blind, cross-over, placebo-controlled trial. J Psychopharma-
col 31: 327–337.
Kaynak H, Kaynak D, Gozukirmizi E, et al. (2004) The effects of tra-
zodone on sleep in patients treated with stimulant antidepressants.
Sleep Med 5: 15–20.
Keeshin BR, Ding Q, Presson AP, et al. (2017) Use of prazosin for pedi-
atric PTSD-associated nightmares and sleep disturbances: A retro-
spective chart review. Neurol Ther 6: 247–257.
Keshavan MS, Prasad KM, Montrose DM, et al. (2007) Sleep quality and
architecture in quetiapine, risperidone, or never-treated schizophre-
nia patients. J Clin Psychopharmacol 27: 703–705.
Khan MS and Aouad R (2017) The effects of insomnia and sleep loss on
cardiovascular disease. Sleep Med Clin 12: 167–177.
Kim SJ, Lee YJ, Lee YJ, et al. (2014) Effect of quetiapine XR on depres-
sive symptoms and sleep quality compared with lithium in patients
with bipolar depression. J Affect Disord 157: 33–40.
Kluge M, Schacht A, Himmerich H, et al. (2014) Olanzapine and clozap-
ine differently affect sleep in patients with schizophrenia: Results
from a double-blind, polysomnographic study and review of the lit-
erature. Schizophr Res 152: 255–260.
Kosky C, Bonakis A, Merritt S, et al. (2008) Sodium oxybate improves
coexisting REM behavior disorder in narcolepsy with cataplexy. J
Sleep Res 17: 97.
Krakow B, Hollifield M, Johnston L, et al. (2001) Imagery rehearsal
therapy for chronic nightmares in sexual assault survivors with post-
traumatic stress disorder: A randomized controlled trial. JAMA 286:
537–545.
Krystal AD (2009) A compendium of placebo-controlled trials of the
risks/benefits of pharmacological treatments for insomnia: The
empirical basis for U.S. clinical practice. Sleep Med Rev 13: 265–274.
Krystal AD, Durrence HH, Scharf M, et al. (2010) Efficacy and safety of
doxepin 1 mg and 3 mg in a 12-week sleep laboratory and outpatient
trial of elderly subjects with chronic primary insomnia. Sleep 33:
1553–1561.
Krystal AD, Erman M, Zammit GK, et al. (2008) Long-term efficacy
and safety of zolpidem extended-release 12.5 mg, administered 3 to
7 nights per week for 24 weeks, in patients with chronic primary
insomnia: A 6-month, randomized, double-blind, placebo-controlled,
parallel-group, multicenter study. Sleep 31: 79–90.
Krystal AD, Lankford A, Durrence HH, et al. (2011) Efficacy and safety
of doxepin 3 and 6 mg in a 35-day sleep laboratory trial in adults with
chronic primary insomnia. Sleep 34: 1433–1442.
Krystal AD, Richelson E and Roth T (2013) Review of the histamine
system and the clinical effects of H1 antagonists: Basis for a new
model for understanding the effects of insomnia medications. Sleep
Med Rev 17: 263–272.
Krystal AD, Walsh JK, Laska E, et al. (2003) Sustained efficacy of
eszopiclone over 6 months of nightly treatment: Results of a random-
ized, double-blind, placebo-controlled study in adults with chronic
insomnia. Sleep 26: 793–799.
Krystal AD and Zammit G (2016) The sleep effects of lurasidone: A pla-
cebo-controlled cross-over study using a 4-h phase-advance model
of transient insomnia. Hum Psychopharmacol 31: 206–216.
Kuh DL, Hardy R and Wadsworth M (1997) Women’s health in midlife:
The influence of the menopause, social factors and health in earlier
life. Br J Obstet Gynaecol 104: 1419.
Kunz D and Mahlberg R (2010) A two-part, double-blind, placebo-con-
trolled trial of exogenous melatonin in REM sleep behaviour disor-
der. J Sleep Res 19: 591–596.
Kyle SD, Morgan K and Espie CA (2010) Insomnia and health-related
quality of life. Sleep Med Rev 14: 69–82.
Landolt HP, Retey JV, Tonz K, et al. (2004) Caffeine attenuates waking
and sleep electroencephalographic markers of sleep homeostasis in
humans. Neuropsychopharmacology 29: 1933–1939.
Lankford A, Rogowski R, Essink B, et al. (2012) Efficacy and safety of
doxepin 6 mg in a four-week outpatient trial of elderly adults with
chronic primary insomnia. Sleep Med 13: 133–138.
944 Journal of Psychopharmacology 33(8)
Lazowski LK, Townsend B, Hawken ER, et al. (2014) Sleep architecture and
cognitive changes in olanzapine-treated patients with depression: A dou-
ble blind randomized placebo controlled trial. BMC Psychiatry 14: 202.
LeBlanc M, Merette C, Savard J, et al. (2009) Incidence and risk factors
of insomnia in a population-based sample. Sleep 32: 1027–1037.
Leger D and Poursain B (2005) An international survey of insomnia:
Under-recognition and under-treatment of a ploysymptomatic condi-
tion. Curr Med Res Opin 21: 1785–1792.
Leger D, Bayon V, Ohayon MM, et al. (2014) Insomnia and accidents:
Cross-sectional study (EQUINOX) on sleep-related home, work and
car accidents in 5293 subjects with insomnia from 10 countries. J
Sleep Res 23: 143–152.
Leger D, Laudon M and Zisapel N (2004) Nocturnal 6-sulfatoxymelato-
nin excretion in insomnia and its relation to the response to melato-
nin replacement therapy. Am J Med 116: 91–95.
Leger D, Morin CM, Uchiyama M, et al. (2012) Chronic insomnia, qual-
ity-of-life, and utility scores: Comparison with good sleepers in a
cross-sectional international survey. Sleep Med 13: 43–51.
Lemoine P, Nir T, Laudon M, et al. (2007) Prolonged-release melatonin
improves sleep quality and morning alertness in insomnia patients
aged 55 years and older and has no withdrawal effects. J Sleep Res
16: 372–380.
Lindberg N, Virkkunen M, Tani P, et al. (2002) Effect of a single-dose of
olanzapine on sleep in healthy females and males. Int Clin Psycho-
pharmacol 17: 177–184.
Lockley SW, Dressman MA, Licamele L, et al. (2015) Tasimelteon for
non-24-hour sleep-wake disorder in totally blind people (SET and
RESET): Two multicentre, randomised, double-masked, placebo-
controlled phase 3 trials. Lancet 386: 1754–1764.
Loebel AD, Siu CO, Cucchiaro JB, et al. (2014) Daytime sleepiness asso-
ciated with lurasidone and quetiapine XR: Results from a randomized
double-blind, placebo-controlled trial in patients with schizophrenia.
CNS Spectr 19: 197–205.
Luik AI, Machado PF, Siriwardena N, et al. (2019) Screening for insom-
nia in primary care: Using a two-item version of the Sleep Condition
Indicator. Br J Gen Pract 69: 79–80.
MacMahon KM, Broomfield NM and Espie CA (2005) A systematic
review of the effectiveness of oral melatonin for adults (18 to 65
years) with delayed sleep phase syndrome and adults (18 to 65 years)
with primary insomnia. Curr Psychiatry Rev 1: 103–113.
Maras A, Schroder CM, Malow BA, et al. (2018) Long-term efficacy and
safety of pediatric prolonged-release melatonin for insomnia in chil-
dren with autism spectrum disorder. J Child Adolesc Psychopharmacol.
Epub ahead of print 11 October 2018. DOI:10.1089/cap.2018.0020.
Massironi G, Galluzzi S and Frisoni GB (2003) Drug treatment of REM
sleep behavior disorders in dementia with Lewy bodies. Int Psycho-
geriatr 15: 377–383.
Maust DT, Lin LA and Blow FC (2019) Benzodiazepine use and misuse
among adults in the United States. Psychiatr Serv 70: 97–106.
Mayer G, Wang-Weigand S, Roth-Schechter B, et al. (2009) Efficacy and
safety of 6-month nightly ramelteon administration in adults with
chronic primary insomnia. Sleep 32: 351–360.
Mayers AG and Baldwin DS (2005) Antidepressants and their effect on
sleep. Hum Psychopharmacol 20: 533–559.
McCleery J, Cohen DA and Sharpley AL (2016) Pharmacotherapies for
sleep disturbances in dementia. Cochrane Database Syst Rev 11:
CD009178.
McCurry SM, Guthrie KA, Morin CM, et al. (2016) Telephone-based
cognitive behavioral therapy for insomnia in perimenopausal and
postmenopausal women with vasomotor symptoms: A MsFLASH
randomized clinical trial. JAMA Intern Med 176: 913–920.
Meltzer LJ and Mindell JA (2014) Systematic review and meta-analysis
of behavioral interventions for pediatric insomnia. J Pediatr Psychol
39: 932–948.
Merenstein D, ener-West M, Halbower AC, et al. (2006) The trial of
infant response to diphenhydramine: The TIRED study—a randomized,
controlled, patient-oriented trial. Arch Pediatr Adolesc Med 160:
707–712.
Mindell JA, Kuhn B, Lewin DS, et al. (2006) Behavioral treatment of
bedtime problems and night wakings in infants and young children.
Sleep 29: 1263–1276.
Montgomery P, Stores G and Wiggs L (2004) The relative efficacy of
two brief treatments for sleep problems in young learning disabled
(mentally retarded) children: A randomised controlled trial. Arch Dis
Child 89: 125–130.
Montgomery SA, Bebbington P, Cowen P, et al. (1993) Guidelines for treat-
ing depressive illness with antidepressants: A statement from the British
Association for Psychopharmacology. J Psychopharmacol 7: 19–23.
Monti JM, Torterolo P and Pandi Perumal SR (2017) The effects of
second generation antipsychotic drugs on sleep variables in healthy
subjects and patients with schizophrenia. Sleep Med Rev 33: 51–57.
Monti JM (2016) The effect of second-generation antipsychotic drugs on
sleep parameters in patients with unipolar or bipolar disorder. Sleep
Med 23: 89–96.
Moreno RA, Hanna MM, Tavares SM, et al. (2007) A double-blind com-
parison of the effect of the antipsychotics haloperidol and olanzapine
on sleep in mania. Braz J Med Biol Res 40: 357–366.
Morgan K, Dixon S, Mathers N, et al. (2004) Psychological treatment for
insomnia in the regulation of long-term hypnotic drug use. Health
Technol Assess 8: iii-iv, 1–68.
Morin CM, Bastien C, Guay B, et al. (2004) Randomized clinical trial of
supervised tapering and cognitive behavior therapy to facilitate ben-
zodiazepine discontinuation in older adults with chronic insomnia.
Am J Psychiatry 161: 332–342.
Morin CM, Belanger L, Bastien C, et al. (2005a) Long-term outcome
after discontinuation of benzodiazepines for insomnia: A survival
analysis of relapse. Behav Res Ther 43: 1–14.
Morin CM, Belanger L, LeBlanc M, et al. (2009) The natural history of
insomnia: A population-based 3-year longitudinal study. Arch Intern
Med 169: 447–453.
Morin CM, Colecchi C, Stone J, et al. (1999) Behavioral and pharma-
cological therapies for late-life insomnia: A randomized controlled
trial. JAMA 281: 991–999.
Morin CM, Koetter U, Bastien C, et al. (2005b) Valerian-hops combi-
nation and diphenhydramine for treating insomnia: A randomized
placebo-controlled clinical trial. Sleep 28: 1465–1471.
Morin CM, LeBlanc M, Daley M, et al. (2006) Epidemiology of insom-
nia: Prevalence, self-help treatments, consultations, and determi-
nants of help-seeking behaviors. Sleep Med 7: 123–130.
Morphy H, Dunn KM, Lewis M, et al. (2007) Epidemiology of insomnia:
A longitudinal study in a UK population. Sleep 30: 274–280.
Muller MJ, Rossbach W, Mann K, et al. (2004) Subchronic effects of
olanzapine on sleep EEG in schizophrenic patients with predomi-
nantly negative symptoms. Pharmacopsychiatry 37: 157–162.
Murphy SM and Tyrer P (1991) A double-blind comparison of the effects
of gradual withdrawal of lorazepam, diazepam and bromazepam in
benzodiazepine dependence. Br J Psychiat 158: 511–516.
Murray JM, Sletten TL, Magee M, et al. (2017) Prevalence of circa-
dian misalignment and its association with depressive symptoms in
delayed sleep phase disorder. Sleep 40.
Nadorff MR, Lambdin KK and Germain A (2014) Pharmacological and
non-pharmacological treatments for nightmare disorder. Int Rev Psy-
chiatry 26: 225–236.
Nash JR, Wilson SJ, Potokar JP, et al. (2003) Mirtazapine induces REM
sleep behavior disorder (RBD) in parkinsonism. Neurology 61: 1161.
Neau JP, Marion P, Mathis S, et al. (2010) Restless legs syndrome and
pregnancy: Follow-up of pregnant women before and after delivery.
Eur Neurol 64: 361–366.
Neckelmann D, Mykletun A and Dahl AA (2007) Chronic insomnia as a
risk factor for developing anxiety and depression. Sleep 30: 873–880.
Neutel CI (1995) Risk of traffic accident injury after a prescription for a
benzodiazepine. Ann Epidemiol 5: 239–244.
Wilson et al. 945
Nguyen BH, Perusse D, Paquet J, et al. (2008) Sleep terrors in children: A
prospective study of twins. Pediatrics 122: e1164-e1167.
Nutt DJ and Stahl SM (2010) Searching for perfect sleep: The continuing
evolution of GABAA receptor modulators as hypnotics. J Psycho-
pharmacol 24: 1601–1612.
Nutt DJ (2005a) Death by tricyclic: The real antidepressant scandal? J
Psychopharmacol 19: 123–124.
Nutt DJ (2005b) NICE: The National Institute of Clinical Excellence —
or Eccentricity? Reflections on the Z-drugs as hypnotics. J Psycho-
pharmacol 19: 125–127.
O’Brien LM, Bullough AS, Chames MC, et al. (2014) Hypertension,
snoring, and obstructive sleep apnoea during pregnancy: A cohort
study. BJOG 121: 1685–1693.
Ohayon MM, Caulet M, Arbus L, et al. (1999) Are prescribed medica-
tions effective in the treatment of insomnia complaints? J Psychosom
Res 47: 359–368.
Owens JF and Matthews KA (1998) Sleep disturbance in healthy middle-
aged women. Maturitas 30: 41–50.
Pallesen S, Sivertsen B, Nordhus IH, et al. (2014) A 10-year trend of
insomnia prevalence in the adult Norwegian population. Sleep Med
15: 173–179.
Palmer CA and Alfano CA (2017) Sleep and emotion regulation: An
organizing, integrative review. Sleep Med Rev 31: 6–16.
Parr JM, Kavanagh DJ, Cahill L, et al. (2009) Effectiveness of current
treatment approaches for benzodiazepine discontinuation: A meta-
analysis. Addiction 104: 13–24.
Picchietti DL, Hensley JG, Bainbridge JL, et al. (2015) Consensus clini-
cal practice guidelines for the diagnosis and treatment of restless legs
syndrome/Willis-Ekbom disease during pregnancy and lactation.
Sleep Med Rev 22: 64–77.
Pien GW and Schwab RJ (2004) Sleep disorders during pregnancy. Sleep
27: 1405–1417.
Pigeon WR, Bishop TM and Krueger KM (2017) Insomnia as a precip-
itating factor in new onset mental illness: A systematic review of
recent findings. Curr Psychiatry Rep 19: 44.
Porkka-Heiskanen T, Alanko L, Kalinchuk A, et al. (2002) Adenosine
and sleep. Sleep Med Rev 6: 321–332.
Pressman MR (2007) Factors that predispose, prime and precipitate
NREM parasomnias in adults: Clinical and forensic implications.
Sleep Med Rev 11: 5–30.
Pringsheim T and Gardner DM (2014) Dispensed prescriptions for que-
tiapine and other second-generation antipsychotics in Canada from
2005 to 2012: A descriptive study. CMAJ Open 2: E225–232.
Qaseem A, Kansagara D, Forciea MA, et al. (2016) Management of
chronic insomnia disorder in adults: A clinical practice guideline from
the American College of Physicians. Ann Intern Med 165: 125–133.
Ramchandani P, Wiggs L, Webb V, et al. (2000) A systematic review of
treatments for settling problems and night waking in young children.
BMJ 320: 209–213.
Randall S, Roehrs TA and Roth T (2012) Efficacy of eight months of nightly
zolpidem: A prospective placebo-controlled study. Sleep 35: 1551–1557.
Reid KJ, Jaksa AA, Eisengart JB, et al. (2012) Systematic evaluation of
Axis-I DSM diagnoses in delayed sleep phase disorder and evening-
type circadian preference. Sleep Med 13: 1171–1177.
Reynolds CF, III, Buysse DJ, Miller MD, et al. (2006) Paroxetine treat-
ment of primary insomnia in older adults. Am J Geriatr Psychiatry
14: 803–807.
Rhalimi M, Helou R and Jaecker P (2009) Medication use and increased
risk of falls in hospitalized elderly patients: A retrospective, case-
control study. Drugs Aging 26: 847–852.
Richardson GS, Zammit G, Wang-Weigand S, et al. (2009) Safety and
subjective sleep effects of ramelteon administration in adults and
older adults with chronic primary insomnia: A 1-year, open-label
study. J Clin Psychiatry 70: 467–476.
Richelson E (1994) The pharmacology of antidepressants at the synapse:
Focus on newer compounds. J Clin Psychiatry 55(Suppl A): 34–39.
Riemann D, Baglioni C, Bassetti C, et al. (2017) European guideline for
the diagnosis and treatment of insomnia. J Sleep Res 26: 675–700.
Riemann D, Spiegelhalder K, Feige B, et al. (2010) The hyperarousal
model of insomnia: A review of the concept and its evidence. Sleep
Med Rev 14: 19–31.
Riemann D, Voderholzer U, Cohrs S, et al. (2002) Trimipramine in pri-
mary insomnia: Results of a polysomnographic double-blind con-
trolled study. Pharmacopsychiatry 35: 165–174.
Riemann D (2009) Does effective management of sleep disorders reduce
depressive symptoms and the risk of depression? Drugs 69(Suppl
2): 43–64.
Roehrs TA, Randall S, Harris E, et al. (2012) Twelve months of nightly zolpi-
dem does not lead to rebound insomnia or withdrawal symptoms: A pro-
spective placebo-controlled study. J Psychopharmacol 26: 1088–1095.
Roenneburg T, Kuehnle T, Pramstaller PP, et al. (2004) A marker for the
end of adolescence. Curr Biol 14: R1038–R1039.
Rossignol DA and Frye RE (2011) Melatonin in autism spectrum disor-
ders: A systematic review and meta-analysis. Dev Med Child Neurol
53: 783–792.
Roth T and Ancoli-Israel S (1999) Daytime consequences and correlates
of insomnia in the United States: Results of the 1991 National Sleep
Foundation Survey. II. Sleep 22(Suppl 2): S354–S358.
Ruwe F, P IJ-B, Roth T, et al. (2016) A phase 2 randomized dose-finding
study with esmirtazapine in patients with primary insomnia. J Clin
Psychopharmacol 36: 457–464.
Sack RL (2010) Clinical practice. Jet lag. N Engl J Med 362: 440–447.
Sack RL, Auckley D, Auger RR, et al. (2007a) Circadian rhythm sleep
disorders: Part I, basic principles, shift work and jet lag disorders.
An American Academy of Sleep Medicine review. Sleep 30: 1460–
1483.
Sack RL, Auckley D, Auger RR, et al. (2007b) Circadian rhythm sleep
disorders: Part II, advanced sleep phase disorder, delayed sleep
phase disorder, free-running disorder, and irregular sleep-wake
rhythm. An American Academy of Sleep Medicine review. Sleep
30: 1484–1501.
Salin-Pascual RJ, Herrera-Estrella M, Galicia-Polo L, et al. (2004) Low
delta sleep predicted a good clinical response to olanzapine adminis-
tration in schizophrenic patients. Rev Invest Clin 56: 345–350.
Salin-Pascual RJ, Herrera-Estrella M, Galicia-Polo L, et al. (1999) Olan-
zapine acute administration in schizophrenic patients increases delta
sleep and sleep efficiency. Biol Psychiatry 46: 141–143.
Sanchez-Ortuno MM and Edinger JD (2012) Cognitive-behavioral ther-
apy for the management of insomnia comorbid with mental disor-
ders. Curr Psychiatry Rep 14: 519–528.
Sanna E, Busonero F, Talani G, et al. (2002) Comparison of the effects
of zaleplon, zolpidem, and triazolam at various GABA(A) receptor
subtypes. Eur J Pharmacol 451: 103–110.
Sansone RA and Sansone LA (2010) Is seroquel developing an illicit
reputation for misuse/abuse? Psychiatry 7: 13–16.
Saper CB, Scammell TE and Lu J (2005) Hypothalamic regulation of
sleep and circadian rhythms. Nature 437: 1257–1263.
Sateia MJ, Buysse DJ, Krystal AD, et al. (2017) Clinical practice guide-
line for the pharmacologic treatment of chronic insomnia in adults:
An American Academy of sleep medicine clinical practice guideline.
J Clin Sleep Med 13: 307–349.
Sateia MJ, Doghramji K, Hauri PJ, et al. (2000) Evaluation of chronic
insomnia. An American Academy of Sleep Medicine review. Sleep
23: 243–308.
Schenck C and Mahowald M (1996) Long-term, nightly benzodiazepine
treatment of injurious parasomnias and other disorders of disrupted
nocturnal sleep in 170 adults. Am J Med 100: 333–337.
Sedov ID, Cameron EE, Madigan S, et al. (2018) Sleep quality during
pregnancy: A meta-analysis. Sleep Med Rev 38: 168–176.
Seyffert M, Lagisetty P, Landgraf J, et al. (2016) Internet-delivered cog-
nitive behavioral therapy to treat insomnia: A systematic review and
meta-analysis. PLoS One 11: e0149139.
946 Journal of Psychopharmacology 33(8)
Sharpley AL, Attenburrow ME, Hafizi S, et al. (2005) Olanzapine
increases slow wave sleep and sleep continuity in SSRI-resistant
depressed patients. J Clin Psychiatry 66: 450–454.
Sharpley AL, Bhagwagar Z, Hafizi S, et al. (2003) Risperidone augmen-
tation decreases rapid eye movement sleep and decreases wake in
treatment-resistant depressed patients. J Clin Psychiatry 64: 192–196.
Sharpley AL, Vassallo CM and Cowen PJ (2000) Olanzapine increases
slow-wave sleep: Evidence for blockade of central 5-HT(2C) recep-
tors in vivo. Biol Psychiatry 47: 468–470.
Shekelle PG, Woolf SH, Eccles M, et al. (1999) Developing clinical
guidelines. West J Med 170: 348–351.
Shepertycky MR, Banno K and Kryger MH (2005) Differences between
men and women in the clinical presentation of patients diagnosed
with obstructive sleep apnea syndrome. Sleep 28: 309–314.
Shirani A and St Louis EK (2009) Illuminating rationale and uses for
light therapy. J Clin Sleep Med 5: 155–163.
Short CA and Carpenter PK (1998) The treatment of sleep disorders in
people with learning disabilities using light therapy. Int J Psychiatry
Clin Prac 1: 143–145.
Sivertsen B, Krokstad S, Mykletun A, et al. (2009) Insomnia symptoms
and use of health care services and medications: The HUNT-2 study.
Behav Sleep Med 7: 210–222.
Skene DJ and Arendt J (2007) Circadian rhythm sleep disorders in the
blind and their treatment with melatonin. Sleep Med 8: 651–655.
Skene DJ, Lockley SW and Arendt J (1999) Melatonin in circadian sleep
disorders in the blind. Biol Signals Recept 8: 90–95.
Sletten TL, Magee M, Murray JM, et al. (2018) Efficacy of melatonin
with behavioural sleep-wake scheduling for delayed sleep-wake phase
disorder: A double-blind, randomised clinical trial. PLoS Med 15:
e1002587.
Smits MG, van Stel HF, van der HK, et al. (2003) Melatonin improves
health status and sleep in children with idiopathic chronic sleep-
onset insomnia: A randomized placebo-controlled trial. J Am Acad
Child Adolesc Psychiatry 42: 1286–1293.
Soldatos CR, Dikeos DG and Whitehead A (1999) Tolerance and rebound
insomnia with rapidly eliminated hypnotics: A meta-analysis of
sleep laboratory studies. Int Clin Psychopharmacol 14: 287–303.
Spiegelhalder K, Nissen C and Riemann D (2017) Clinical sleep-wake
disorders II: Focus on insomnia and circadian rhythm sleep disor-
ders. Handb Exp Pharmacol. Epub ahead of print 14 July 2017. DOI:
10.1007/164_2017_40.
Spielman AJ, Caruso LS and Glovinsky PB (1987) A behavioral perspec-
tive on insomnia treatment. Psychiatr Clin North Am 10: 541–553.
Stallman HM, Kohler M and White J (2018) Medication induced sleep-
walking: A systematic review. Sleep Med Rev 37: 105–113.
Staner L, Ertle S, Boeijinga P, et al. (2005) Next-day residual effects of
hypnotics in DSM-IV primary insomnia: A driving simulator study
with simultaneous electroencephalogram monitoring. Psychophar-
macology 181: 790–798.
Symons FJ, Davis ML and Thompson T (2000) Self-injurious behavior
and sleep disturbance in adults with developmental disabilities. Res
Dev Disabil 21: 115–123.
Tassniyom K, Paholpak S, Tassniyom S, et al. (2010) Quetiapine for pri-
mary insomnia: A double blind, randomized controlled trial. J Med
Assoc Thai 93: 729–734.
Tempesta D, Socci V, De Gennaro L, et al. (2018) Sleep and emotional
processing. Sleep Med Rev 40: 183–195.
Todder D, Caliskan S and Baune BT (2006) Night locomotor activity and
quality of sleep in quetiapine-treated patients with depression. J Clin
Psychopharmacol 26: 638–642.
Trewin VF, Lawrence CJ and Veitch GB (1992) An investigation of the
association of benzodiazepines and other hypnotics with the inci-
dence of falls in the elderly. J Clin Pharm Ther 17: 129–133.
Unruh ML, Redline S, An MW, et al. (2008) Subjective and objective
sleep quality and aging in the sleep heart health study. J Am Geriatr
Soc 56: 1218–1227.
van de Wouw E, Evenhuis HM and Echteld MA (2012) Prevalence,
associated factors and treatment of sleep problems in adults with
intellectual disability: A systematic review. Res Dev Disabil 33:
1310–1332.
van der Heijden KB, Smits MG, Van Someren EJ, et al. (2007) Effect of
melatonin on sleep, behavior, and cognition in ADHD and chronic
sleep-onset insomnia. J Am Acad Child Adolesc Psychiatry 46: 233–
241.
Van Houdenhove L, Buyse B, Gabriels L, et al. (2011) Treating pri-
mary insomnia: Clinical effectiveness and predictors of outcomes
on sleep, daytime function and health-related quality of life. J Clin
Psychol Med Settings 18: 312–321.
van Straten A, van der Zweerde T, Kleiboer A, et al. (2018) Cognitive
and behavioral therapies in the treatment of insomnia: A meta-analy-
sis. Sleep Med Rev 38: 3–16.
Varkevisser M, Van Dongen HP and Kerkhof GA (2005) Physiologic
indexes in chronic insomnia during a constant routine: Evidence for
general hyperarousal? Sleep 28: 1588–1596.
Vermeeren A, Sun H, Vuurman EF, et al. (2015) On-the-road driving
performance the morning after bedtime use of suvorexant 20 and 40
mg: A study in non-elderly healthy volunteers. Sleep 38: 1803–1813.
Verster J, Veldhuijzen DS, Patat A, et al. (2006) Hypnotics and driving
safety: Meta analyses of randomised controlled trials applying the
on-the-road driving test. Curr Drug Saf 1: 63–71.
Vgontzas AN, Bixler EO, Lin HM, et al. (2001) Chronic insomnia is
associated with nyctohemeral activation of the hypothalamic-pitu-
itary-adrenal axis: Clinical implications. J Clin Endocrinol Metab
86: 3787–3794.
Vgontzas AN, Liao D, Bixler EO, et al. (2009) Insomnia with objective
short sleep duration is associated with a high risk for hypertension.
Sleep 32: 491–497.
Vgontzas AN, Tsigos C, Bixler EO, et al. (1998) Chronic insomnia and
activity of the stress system: A preliminary study. J Psychosom Res
45: 21–31.
Voshaar RC, van Balkom AJ and Zitman FG (2004) Zolpidem is not
superior to temazepam with respect to rebound insomnia: A con-
trolled study. Eur Neuropsychopharmacol 14: 301–306.
Wade AG, Crawford G, Ford I, et al. (2011) Prolonged release melatonin
in the treatment of primary insomnia: Evaluation of the age cut-off
for short- and long-term response. Curr Med Res Opin 27: 87–98.
Wade AG, Ford I, Crawford G, et al. (2007) Efficacy of prolonged
release melatonin in insomnia patients aged 55–80 years: Quality
of sleep and next-day alertness outcomes. Curr Med Res Opin 23:
2597–2605.
Walsh JK, Erman M, Erwin CW, et al. (1998) Subjective hypnotic effi-
cacy of trazodone and zolpidem in DSM-III-R primary insomnia.
Hum Psychopharm Clin Exp 13: 191–198.
Walsh JK, Krystal AD, Amato DA, et al. (2007) Nightly treatment of
primary insomnia with eszopiclone for six months: Effect on sleep,
quality of life, and work limitations. Sleep 30: 959–968.
Wassmer E, Carter PF, Quinn E, et al. (2001) Melatonin is useful for
recording sleep EEGs: A prospective audit of outcome. Dev Med
Child Neurol 43: 735–738.
Wickwire EM, Shaya FT and Scharf SM (2016) Health economics of
insomnia treatments: The return on investment for a good night's
sleep. Sleep Med Rev 30: 72–82.
Wickwire EM, Tom SE, Scharf SM, et al. (2019) Untreated insomnia
increases all-cause health care utilization and costs among Medicare
beneficiaries. Sleep 42: zsz007.
Wiegand MH, Landry F, Bruckner T, et al. (2008) Quetiapine in primary
insomnia: A pilot study. Psychopharmacology 196: 337–338.
Wiggs L and France K (2000) Behavioural treatments for sleep prob-
lems in children and adolescents with physical illness, psychological
problems or intellectual disabilities. Sleep Med Rev 4: 299–314.
Wilson S and Argyropoulos S (2005) Antidepressants and sleep: A quali-
tative review of the literature. Drugs 65: 927–947.
Wilson et al. 947
Item Score
4 3 2 1 0
Thinking about the past month, to what extent has poor sleep …
1. … troubled you in general Not at all A little Somewhat Much Very much
Thinking about a typical night in the last month …
2. … how many nights a week do you have a problem with your sleep? 0–1 2 3 4 5–7
Scoring instructions: add the item scores to obtain the SCI total (minimum 0, maximum 8). A lower score means better sleep.
Wilson SJ, Lillywhite AR, Potokar JP, et al. (1997) Adult night terrors
and paroxetine. Lancet 350: 185.
Wilson SJ, Nutt DJ, Alford C, et al. (2010) British Association for Psy-
chopharmacology consensus statement on evidence-based treatment
of insomnia, parasomnias and circadian rhythm disorders. J Psycho-
pharmacol 24: 1577–1601.
Winokur A, DeMartinis NA, III, McNally DP, et al. (2003) Comparative
effects of mirtazapine and fluoxetine on sleep physiology measures
in patients with major depression and insomnia. J Clin Psychiatry
64: 1224–1229.
Woolcott JC, Richardson KJ, Wiens MO, et al. (2009) Meta-analysis of
the impact of 9 medication classes on falls in elderly persons. Arch
Intern Med 169: 1952–1960.
World Health Organization (1992) The ICD-10 Classification of
mentaland behavioural disorders: clinical descriptions and diagnostic
guidelines. Geneva: WHO. https://www.who.int/classifications/icd/en
/bluebook.pdf
Yeung WF, Chung KF, Yung KP, et al. (2015) Doxepin for insomnia:
A systematic review of randomized placebo-controlled trials. Sleep
Med Rev 19: 75–83.
Young T, Finn L, Austin D, et al. (2003) Menopausal status and sleep-
disordered breathing in the Wisconsin Sleep Cohort Study. Am J
Respir Crit Care Med 167: 1181–1185.
Zachariae R, Lyby MS, Ritterband LM, et al. (2016) Efficacy of internet-
delivered cognitive-behavioral therapy for insomnia: A systematic
review and meta-analysis of randomized controlled trials. Sleep Med
Rev 30: 1–10.
Zentiva Pharma UK Limited (2002) Zolpidem - Summary of Product
Characteristics. Available at: https://www.medicines.org.uk/emc
/product/3975/smpc (accessed 18 June 2019).
Appendix 1
Two-item version of the Sleep Condition Indicator (SCI) 02 (Luik et al. (2019) Screening for insomnia in primary care: Using a two-
item version of the Sleep Condition Indicator. Br J Gen Pract 69: 79–80.)