ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Overview of the treatment of insomnia in adults

Overview of the treatment of insomnia in adults
Literature review current through: Jan 2024.
This topic last updated: Jan 19, 2024.

INTRODUCTION — Insomnia is one of the most common symptoms for which adults seek medical advice. Although sleep is a strong and highly regulated biologic drive, the ability to fall asleep at the desired time and maintain sleep without excessive waking is fragile and influenced by multiple factors. Identification of these factors is at the core of insomnia treatment.

Management of insomnia requires a stepwise approach, beginning with attempts to eliminate, or at least minimize, the multiple contributing factors and comorbid illnesses that can interfere with optimal sleep. Insomnia often responds only partially to treatment of sleep-impairing influences, however, and many patients require treatment directed towards sleep itself. However, successful behavioral and pharmacologic approaches to insomnia should only be implemented once all contributing factors are recognized and attempts to address them are made.

This topic is an overview of the approach to management of acute and chronic insomnia in adults. Specific medications and behavioral therapies for insomnia are reviewed in more detail separately. (See "Pharmacotherapy for insomnia in adults" and "Cognitive behavioral therapy for insomnia in adults".)

INITIAL ASSESSMENT AND COUNSELING — Insomnia etiology is best conceptualized as a combination of predisposing, precipitating, and perpetuating factors that vary over time. Each of these factors should be assessed to formulate an individualized treatment plan.

Predisposing and precipitating factors — The sleep history should include an examination of the social, medical, and psychiatric events that may have been relevant at the time insomnia began. When an individual is under psychologic or physiologic stress, such factors are variably amplified in the context of their underlying reactivity to sleep disturbance (table 1) [1,2]. (See "Evaluation and diagnosis of insomnia in adults", section on 'Common comorbidities'.)

Such events and comorbidities are important for treatment because:

Insomnia precipitated or exacerbated by a symptom of medical illness (eg, pain, nocturia, or shortness of breath) is unlikely to improve without maximal treatment of the medical disorder. (See 'Pain' below.)

Sleep disorders other than insomnia (eg, obstructive sleep apnea [OSA], circadian rhythm disorders, restless legs syndrome [RLS]) may present with insomnia but are unlikely to improve without treatment directed at the specific sleep disorder. Suspected OSA is an indication for in-laboratory polysomnography or home sleep apnea testing. (See 'Patients with comorbid sleep disorders' below and "Clinical presentation and diagnosis of obstructive sleep apnea in adults", section on 'Diagnostic evaluation'.)

Psychiatric disorders and insomnia have a bidirectional relationship, and concomitant treatment for both disorders is often necessary to hasten recovery and increase the likelihood of sustained response of both disorders. (See 'Patients with comorbid psychiatric disorders' below.)

A history of childhood trauma or chaotic home environment at night (even in the absence of posttraumatic stress disorder [PTSD]) may increase vulnerability to sleeplessness as an adult. Awareness of this history is valuable as it may shed light on etiology and help identify targets of cognitive therapy.

Medication side effects — Medications used to treat a comorbid condition may themselves precipitate insomnia through stimulation of arousal centers or other central nervous system effects (eg, stimulants, glucocorticoids, some antidepressants), nocturia (eg, diuretics), or respiratory suppression (eg, opioids) (table 2). Use of medications with the potential to disrupt sleep has been associated with an increased risk of insomnia on a population level [3] and is commonly overlooked as a contributing factor.

Stimulants – The major determinants of whether stimulants such as methylphenidate or modafinil will interfere with sleep are dose and effective half-life. Many stimulants have effective half-lives greater than 10 hours and can therefore interfere with both sleep onset and sleep maintenance. Lowering the dose, choosing a shorter-acting agent, and administering the medication earlier in the day may attenuate the sleep disturbance.

Antidepressants – Selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs) are associated with treatment-induced insomnia in approximately 20 percent of patients. While morning dosing is commonly recommended to mitigate sleep disturbances, it is not clear that this reduces the risk of incident insomnia. Lowering the dose may be of value. Sometimes, the sleep disruptive effects of antidepressants are transient, and temporary use of a sedating medication may be indicated to address this side effect.

Glucocorticoids – Oral or inhaled glucocorticoids commonly produce insomnia [4]. Lowering the dose and administering the medication earlier in the day may attenuate the sleep disturbance. If sleeplessness does not improve over time, co-administration of a sedating medication is a common approach.

Opioids – Although opioids are often sedating, chronic use can lead to increased sleep fragmentation and nocturnal awakenings related to changes in sleep architecture and respiratory control. Polysomnography is required to diagnose sleep-disordered breathing due to chronic opioids, as central apneas or irregular breathing patterns may not be apparent to the patient or a bed partner. Lowering or eliminating the opioid dose and positive airway pressure therapy are the primary treatment strategies. (See "Sleep-disordered breathing in patients chronically using opioids".)

Perpetuating factors — Behavioral and cognitive compensatory responses to sleeplessness, some of which can be maladaptive for sleep, may further disturb sleep onset and maintenance, and perpetuate insomnia.

Many patients with insomnia have poor sleep habits (table 3), substantial anxiety about falling or staying asleep, unrealistic expectations of sleep, or inappropriate attributions about the association of daytime symptoms and nocturnal sleep. When these counterproductive processes are substantial, they should be addressed, either alone or in addition to other therapeutic strategies.

Determine consistency of timing of bedtime and waking time and amount of time allotted to attempts to sleep. A sleep diary (table 4A-B), filled out by the patient for at least two weeks, is an important component of the evaluation and is often informative for both patient and clinician. Regular timing of bedtime and, particularly, waking time (seven days per week) is essential for those with insomnia. The amount of time in bed, from initial attempts to sleep until final waking time, should not exceed the estimated total sleep time.

Ask if the patient is actually sleepy at bedtime. Attempts to sleep should occur only when sleepy (similar to attempts to empty the bowel or bladder).

Ask about napping and dozing during the day or evening, which should be avoided to maximize sleep drive at night.

Determine the level of anxiety regarding sleeplessness, both in the evening before bed as well as while awake in bed attempting to sleep. If substantial anxiety is present ("insomnia phobia"), methods to reduce anxiety should be instituted. (See 'Overview of cognitive behavioral therapy' below.)

Ask about clock-watching while in bed.

Ask about nocturnal environmental disturbances (children, pets, bed partner, electronics).

Determine if expectations of sleep onset time, number of awakenings, and total sleep time are realistic for age, and, if not, educate about appropriate expectations (figure 1). (See "Insufficient sleep: Definition, epidemiology, and adverse outcomes", section on 'How much sleep do we need?'.)

Assess perceived consequences of sleeplessness and attributions of daytime function and health to sleep. Educate about appropriate attributions of the effects of insomnia.

Maladaptive habits (behaviors) and attitudes (cognitions) that may perpetuate sleeplessness are the targets of cognitive behavioral therapy for insomnia (CBT-I) and should be addressed in all patients with chronic insomnia. However, it should be noted that by the time patients present to a clinician, it is often difficult to determine if such compensations are the most important cause of the insomnia or simply an inappropriate response to it.

APPROACH TO ACUTE INSOMNIA — Short-term insomnia is a common form of insomnia and usually results from psychologic or physiologic stress. Patients with acute insomnia can often identify the immediate precipitant to the insomnia. Symptoms most often last for less than a month, although formal diagnostic criteria allow for symptoms lasting up to three months [5]. The clinical approach to acute insomnia is twofold:

Discuss the role that the stressor is playing in disturbing sleep and assess the level of distress. Education can provide some control or at least acceptance of the temporary sleeplessness. For patients with mild or manageable levels of distress, we provide reassurance and a plan for follow up if insomnia does not improve.

When the insomnia is severe or associated with substantial distress, we offer short-term use of an insomnia medication to help address immediate interference with daytime function and to control escalating anxiety about sleep. Selection of a medication is individualized using the same principles as those for chronic insomnia medication selection. (See "Pharmacotherapy for insomnia in adults", section on 'Our approach'.)

The goal of pharmacologic therapy in this setting is to minimize the additional psychologic and physical stress that sleeplessness produces. In addition, medication treatment of short-term insomnia may reduce the development of dysfunctional cognitive and behavioral responses to sleeplessness that could otherwise increase the risk of more chronic insomnia.

Follow-up in two to four weeks is encouraged to reassess sleep-related symptoms and anxiety about sleep, reinforce good sleep habits, and consider additional causes of insomnia. If insomnia is persistent, we encourage evaluation and treatment with cognitive behavioral therapy for insomnia (CBT-I). (See 'Overview of cognitive behavioral therapy' below.)

APPROACH TO CHRONIC INSOMNIA — Cognitive behavioral therapy (CBT) and pharmacotherapy are the main treatment options for chronic insomnia that persists despite appropriate identification and management of precipitating and perpetuating factors.

Choice of initial therapy — CBT for insomnia (CBT-I) is preferred as first-line therapy for chronic insomnia in most patients. However, CBT-I is not effective for all patients and is not accessible to many, either due to lack of therapists or to limitations of insurance or time. In such cases, long-term use of medications is an acceptable approach if the patient is thoroughly evaluated beforehand, is followed regularly during the course of treatment, and continues to respond positively to the medication [6].

Several prospective studies have assessed the comparative efficacy of CBT-I, medications, and combination therapy as an initial approach. In short-term trials, CBT-I alone and CBT-I in combination with medications demonstrate relatively equivalent outcomes, and both are superior to medication alone [7-9]. Longer-term outcome studies (12 to 24 months) also demonstrate superior efficacy of CBT-I alone or in combination with medication compared with medication-alone approaches for subjective sleep outcomes [10]. CBT-I without medication has the advantage that it does not expose patients to side effects and potential drug interactions, and it provides patients with lifelong skills should insomnia recur [6,11,12]. However, it is unclear that CBT-I produces benefits for objective sleep parameters [13]. A preference for CBT-I or other behavioral therapies over medication as initial therapy has been endorsed in clinical practice guidelines of the American Academy of Sleep Medicine [12], the British Association for Psychopharmacology [14], the American College of Physicians [11,15], and the European Sleep Research Society [16]. (See 'Society guideline links' below.)

For patients who require a rapid response for clinical reasons (eg, deterioration in daytime function) or who have excessive anxiety regarding sleeplessness (which may interfere with the ability to follow time in bed restriction and stimulus control aspects of CBT-I), a combination approach can be used initially (given the faster response with medication approaches), with a plan to taper the medication over time (eg, six to eight weeks). (See 'Follow-up and monitoring' below.)

There are no reliable predictors of treatment response to CBT-I or medications [17]. One might predict that deviations from optimal thoughts and behaviors would provide greater opportunity for benefit from CBT-I; however, counterproductive beliefs and behaviors may have developed as a consequence of more severe sleep disturbance, more disorganized lifestyle, or underlying anxiety disorder and therefore may actually be markers of treatment resistance. One study suggested that greater distress at baseline or prolonged times to fall asleep may predict beneficial response to CBT-I, whereas short total sleep time (<6 hours) may predict poor response [18,19].

Predictors of treatment success with medications for insomnia are also poorly characterized. In one meta-analysis of nonbenzodiazepine benzodiazepine receptor agonists (BZRAs), younger age and female sex were associated with greater improvements in sleep onset latency [20]. Anecdotally, previous nonresponse to high doses of hypnotics or substantial discrepancy between self-reported and objectively recorded total sleep time (referred to as paradoxical insomnia) are indicators of poor future response to medications.

Overview of cognitive behavioral therapy — Cognitive behavioral therapy for insomnia (CBT-I) is the preferred form of treatment for chronic insomnia in adults and has been endorsed as first-line therapy by multiple societies and guideline panels [6,11,14,15,21,22].

CBT-I is a multicomponent approach to chronic insomnia that addresses common thoughts and behaviors that interfere with optimal sleep. It is traditionally delivered in face-to-face individual or group settings, over four to eight sessions; remote delivery (online or telephone) can also be effective, although adherence is less robust in this context. CBT-I is distinct from other forms of CBT (eg, for anxiety and mood disorders), and referring providers should confirm that the therapist has been specifically trained in this modality.

The behavioral components of CBT-I include:

Establishment of a stable bedtime and waking time seven days per week

Reduction in time in bed to approximate the total hours of estimated sleep (time in bed restriction) (table 5)

Encouragement to use the bed only for sleep and sex; try to sleep only when sleepy; and get out of bed if anxiety occurs while unable to sleep (stimulus control) (table 6)

Sleep hygiene, which includes avoidance of substances that interfere with sleep, avoidance of naps to maximize sleep drive, and optimization of the comfort of the sleep environment (table 3)

The cognitive approaches of CBT-I address:

Anxious and catastrophic thoughts that are associated with sleeplessness

Inappropriate expectations about hours of sleep

Misattributions regarding the effects of sleeplessness

Relaxation through progressive muscle relaxation, mindfulness, and meditation (table 7)

CBT-I and other behavioral approaches to chronic insomnia are reviewed in more detail separately. (See "Cognitive behavioral therapy for insomnia in adults".)

Overview of pharmacotherapy — For such patients who have not responded to CBT-I, the risks of untreated insomnia (eg, falls at night, cognitive impairment, development of mood/anxiety disorder, worsening of a medical disorder) also factor into decision-making [23-25]. The choice among various medications for insomnia is individualized based on a variety of factors, including patient age and comorbidities, type of insomnia complaint, side effect profiles, cost, and clinician and patient preference (algorithm 1 and figure 2). The choice of therapy, specific drugs, side effects, and risks are reviewed separately. (See "Pharmacotherapy for insomnia in adults", section on 'Drug selection'.)

Medications with regulatory approval for insomnia treatment span multiple classes and agents that can be categorized based on their mechanism of action or original indication: benzodiazepine receptor agonists (BZRAs, including benzodiazepines and nonbenzodiazepine BZRAs, such as zolpidem), histamine receptor antagonists (eg, low-dose doxepin), melatonin receptor agonists (eg, ramelteon), and dual orexin receptor antagonists (eg, suvorexant) (table 8A-C).

Use of other sedating medications for insomnia should be considered for patients who do not have an adequate therapeutic response to first-line medications with regulatory approval for insomnia, when a different mechanism of action is desired, or when there is a specific reason to avoid BZRAs. Medications with sedating effects, such as trazodone and gabapentin, when used at much lower doses than those studied and approved for other indications, may represent a relatively safe alternative to BZRAs in selected patients, even if the evidence base for efficacy is not as robust.

FOLLOW-UP AND MONITORING

Response assessment — Insomnia is a clinical diagnosis, and therefore treatment response is based on history and patient self-report. A sleep diary (table 4A-B) is a valuable supplement to the history, as some patients may remember only the nights when they slept poorly.

Consumer wearable devices that report sleep parameters are increasingly popular and may be of some value, but only to compare sleep before and after treatment, as their accuracy for absolute sleep metrics is often poor. (See "Actigraphy in the evaluation of sleep disorders", section on 'Consumer wearable devices'.)

Follow-up after the first month of medication use is indicated if continued treatment is planned. Thereafter, follow-up at least every six months is recommended.

Poor response to initial therapy — In patients who fail insomnia treatment, it is important to discuss expectations of sleep, particularly in those with advanced age and comorbidities. In the presence of unreasonable expectations, all treatments have a high likelihood of failure.

Examination of sleep diaries for timing of sleep and duration of time in bed may demonstrate conditions that make optimal sleep unlikely (eg, excess time in bed, variable bedtimes or waking times) or that sleep has in fact improved. Similarly, persistent medical, sleep, or psychiatric disorders can reduce the efficacy of both CBT-I and medications, and these potential contributors should be reassessed. (See 'Perpetuating factors' above.)

Cognitive behavioral therapy for insomnia (CBT-I) requires discipline, perseverance, and a belief that counterintuitive behaviors (eg, reduction in time in bed, getting out of bed when not asleep) can be therapeutic. These factors contribute to suboptimal adherence to, and benefit from, CBT-I in some patients. In such cases, a trial of medication is often reasonable as second-line therapy. Maintenance of as many of the components of CBT-I as possible remains important over time.

When medication treatments fail, clinicians should determine whether failure is due to lack of efficacy or treatment-emergent side effects. Both of these limitations can often be addressed by altering the dose, timing, or specific medication that is prescribed. This topic is reviewed in more detail separately. (See "Pharmacotherapy for insomnia in adults", section on 'Patients with inadequate response'.)

Tapering medications — Attempts to discontinue sedative medications are a valuable but difficult task, as patients may be reluctant to fix what is not broken and are very anxious about the return of sleeplessness. A supportive discussion about the patient's concerns and about the probability of successful discontinuation is recommended.

The duration of insomnia prior to medication treatment is the best predictor of the likelihood of continued need for medication. Nonetheless, approximately 75 percent of those with insomnia have symptoms for less than one year [26]. Conveying this message may be reassuring when patients are considering whether medication remains necessary.

CBT-I treatment is encouraged prior to medication taper to reinforce optimal sleep habits and manage dysfunctional thoughts that will often arise during the process. In general, a slow reduction in medication dose is the best approach, with roughly 25 percent reduction of the original dose each week [27]. For patients who have used a medication for sleep chronically, a taper over many months may be warranted, with allowance for as-needed use when first attempting to stop regular nightly use.

Reasons for subspecialty referral — Referral to a subspecialist is appropriate under the following circumstances:

There is suspicion that a relevant comorbidity (medical, psychiatric, or sleep) is an important contributor to the insomnia and requires further expertise and management in that context.

A patient is not responding to treatments that are generally effective for insomnia.

TREATMENT CONSIDERATIONS FOR SPECIFIC POPULATIONS

Patients with comorbid psychiatric disorders — At least one-third of patients with chronic insomnia have a psychiatric disorder, and, conversely, the majority of those with a psychiatric disorder have insomnia. Although it is worthwhile examining the temporal relationship between the sleep disturbance and the psychiatric illness, establishing a clear causal relationship between the two is often difficult if not impossible. The Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5), defines insomnia disorder as a distinct entity, encouraging independent diagnosis and treatment of insomnia in the context of psychiatric illness [28].

In the context of an untreated psychiatric disorder with substantial insomnia, treatment should be directed towards both sets of symptoms, both to reduce suffering and to enhance psychiatric treatment. For example, concurrent treatment of insomnia with eszopiclone in patients undergoing initial treatment of mood and anxiety disorders has been shown to enhance response time and overall efficacy for the underlying psychiatric illness [29,30].

Treatments that address both sets of symptoms (eg, sedating antidepressants) are often considered when insomnia is a prominent feature of a mood or anxiety disorder. However, the short-term advantages of this approach need to be weighed against the potential side effects (eg, weight gain with mirtazapine, dry mouth with most drugs, daytime sedation), considering that long-term use of the medication for the psychiatric disorder may be required. In addition, the doses of sedating antidepressants used for insomnia (eg, trazodone) are usually far from therapeutic from the standpoint of the mood disorder.

Mood and anxiety disorders — Treatment of mood and anxiety disorders with either pharmacotherapy or psychotherapy produces improvements in insomnia in most patients. However, persistent or treatment-emergent insomnia may occur even after improvement of the psychiatric illness in many patients. Sleep disturbance is the most common persistent symptom in treated depression and is a risk factor for recurrence of a mood episode in euthymic depressed or bipolar patients [31,32]. Treatment of insomnia in this context should be directed towards the presumed causes of the sleep disturbance, assessing the roles of persistent mood disorder, a comorbid sleep, medical or psychiatric disorder, behavioral factors, and the antidepressant medication itself [33]. (See 'Predisposing and precipitating factors' above and 'Perpetuating factors' above.)

The value of cognitive behavioral therapy for insomnia (CBT-I) in patients with mood disorders is unsettled. Initial studies suggested that adding this approach to psychiatric medications improved both sleep disturbance and the underlying depression or bipolar disorder [34,35]. However, subsequent studies, while confirming the benefits of CBT-I for insomnia in this context, have shown mixed results on outcomes related to psychiatric illness or suggest that the benefit for the mood disorder is independent of the effect on sleep [36-38]. There are few controlled data on the use of CBT-I in anxiety disorders comorbid with insomnia.

Posttraumatic stress disorder — Insomnia is nearly universal in patients with posttraumatic stress disorder (PTSD), consistent with the hypervigilance and nightmares that are diagnostic criteria for the disorder. Insomnia is also one of the most common persistent symptoms after evidence-based treatment for PTSD [39]. Consistent with general guidelines, CBT-I is considered first-line therapy for insomnia treatment in PTSD, and one controlled study demonstrated benefit of CBT-I for multiple sleep outcomes compared with a waitlist control group [40]. (See "Posttraumatic stress disorder in adults: Psychotherapy and psychosocial interventions".)

No controlled trials have specifically examined the use of pharmacotherapy to address insomnia in PTSD. However, the most commonly used medications for insomnia in PTSD are trazodone, prazosin, and nonbenzodiazepine benzodiazepine receptor agonists (BZRAs). Treatment of nightmares is discussed separately. (See "Nightmares and nightmare disorder in adults".)

Psychotic disorders — Insomnia is common and often multifactorial in patients with schizophrenia, both prior to and with treatment of the psychotic illness. Contributing factors may include the underlying psychotic disorder, erratic sleep schedules and suboptimal sleep environment, comorbidity with other sleep disorders (eg, sleep apnea), and comorbid psychiatric (eg, substance abuse/dependence) or medical illness.

Clinicians will often use sedating antipsychotics in such patients, either switching from an existing agent or as an adjunctive medication. Unfortunately, these medications have a propensity for weight gain and metabolic disturbances.

Small, controlled trials support the use of CBT-I or eszopiclone in the treatment of insomnia in schizophrenia [41,42]. In the eszopiclone trial, eight weeks of nightly treatment was associated with improvement in overall insomnia severity in patients with clinically stable schizophrenia [42].

Substance use disorders — Substance use disorders, like many other psychiatric disorders, have a bidirectional relationship with insomnia. Insomnia is a risk factor for substance abuse and occurs with increased frequency during active use and in early or late recovery [43,44]. Insomnia is also a risk factor for relapse.

As in other patients with chronic insomnia, use of CBT-I is preferred, particularly as patients with a substance use disorder may have "forgotten" how to initiate sleep without a sedative. Mastering CBT-I may provide confidence that sleep can be achieved using the body's own sleep drive [45,46]. Because CBT-I may take more time to become effective than medications, use of pharmacotherapy early in sobriety may be beneficial in selected patients. Gabapentin, trazodone, and quetiapine are commonly used in this setting. BZRAs are generally avoided due to concerns about relapse, dependence, and the combined toxicity of BZRAs with alcohol and other substances.

The evaluation and treatment of insomnia in patients with a substance use disorder is reviewed in more detail separately. (See "Insomnia in patients with a substance use disorder".)

Older adults — Although insomnia symptoms related to sleep maintenance and achievement of restorative sleep become more prominent with age [26], the prevalence of insomnia disorder is reduced in older adults compared with younger adults, possibly because of decreased expectations of sleep, more flexible sleep hours, or changing role expectations [47].

Treatment of insomnia in older adults requires careful attention to the role of medical, neurologic, sleep, and psychiatric comorbidities. Vulnerability to side effects increases with age, and medications for insomnia often exacerbate existing age-related impairments such as gait instability, sedation, cognitive dysfunction, urinary and bowel dysfunction, and cardiac arrhythmias. Older adults may have slower drug metabolism, more drug-drug interactions, and thus maximum and next-day serum concentrations will be increased. (See "Pharmacotherapy for insomnia in adults", section on 'Special populations'.)

Due to these concerns, CBT-I is the treatment of choice in older adults with chronic insomnia. CBT-I has shown efficacy for sleep as well as depression outcomes in trials in older adults, both in individual and group settings [37,48,49]. CBT-I is well tolerated in this context. When time in bed restriction is used as part of CBT-I, attention to next-day sleepiness is warranted [50]. Other nonpharmacologic interventions that may help sleep in older adults include exercise and tai chi [51-54]. (See "Cognitive behavioral therapy for insomnia in adults" and "Physical activity and exercise in older adults".)

Pharmacotherapy in older adults with persistent insomnia should be individualized with increased awareness of the potential for drug interactions, altered metabolism, and side effects. These considerations are reviewed separately. (See "Pharmacotherapy for insomnia in adults", section on 'Special populations'.)

Hospitalized patients — Insomnia, whether acute or chronic, is commonly encountered in hospitalized patients. Sleep disturbances in the hospital are often multifactorial, and the initial approach should focus on optimizing the sleeping environment, reducing unnecessary nocturnal assessments, treating the underlying medical illness, minimizing stimulating or sleep-related side effects of concomitant medications, and using nonpharmacologic strategies to improve sleep. When pharmacotherapy is deemed necessary, selection of an agent should be individualized based on symptom severity, age, comorbidities, side effects, and drug-drug interactions. (See "Poor sleep and insomnia in hospitalized adults".)

Dementia and other neurodegenerative disorders — Many neurologic disorders are associated with insomnia, either as a result of dysfunction in central nervous system pathways that regulate sleep or as a consequence of pain, immobility, or respiratory dysfunction.

Alzheimer disease – Insomnia is very common in Alzheimer disease (AD) and can manifest with disturbances in circadian timing of sleep, frequent awakenings, and nocturnal restlessness. Nonpharmacologic therapies are the mainstay of treatment. First-line approaches include treatment of comorbid medical and sleep disorders, adjustment of potentially sleep-disruptive medications, and behavioral approaches such as stability of the light-dark cycle and enhancement of social and physical activity during the day.

There are few studies of medication treatments for insomnia in patients with AD, a population in which the risk of side effects is increased. Evaluation and management of sleep disturbances and sleep disorders in patients with AD and other forms of dementia are reviewed in more detail separately. (See "Sleep-wake disturbances and sleep disorders in patients with dementia".)

Parkinson disease – Insomnia is also very common in Parkinson disease (PD), with primary motor symptoms of tremor, bradykinesia, and rigidity leading to impairments in sleep maintenance. Nonmotor symptoms such as depression, hallucinations, and nocturia can also contribute. A stepwise approach to evaluation and treatment of insomnia in patients with PD is presented separately. (See "Evaluation and treatment of insomnia, daytime sleepiness, and other sleep disorders in Parkinson disease", section on 'Insomnia'.)

Concussion and traumatic brain injury — Insomnia is reported by more than half of patients with traumatic brain injury and is especially common among those with milder injuries, including concussion. Circadian rhythm disturbances are also common and can be misattributed to insomnia. The approach to insomnia and other sleep disturbances in patients with traumatic brain injury is reviewed separately. (See "Sleep-wake disorders in patients with traumatic brain injury".)

Patients with comorbid sleep disorders

Obstructive sleep apnea — Insomnia and obstructive sleep apnea (OSA) are distinct sleep disorders that typically require independent treatment; however, they commonly co-occur. Approximately 15 percent of patients with insomnia have moderate to severe OSA, with higher rates in males, older adults, those who snore, and patients with metabolic syndrome, obesity, or daytime sleepiness [55]. Undiagnosed OSA is common among patients with insomnia complaints [56]. Roughly one-third of those with OSA have insomnia complaints, which are actually more common than daytime sleepiness [57]. Comorbid insomnia also interferes with adherence to continuous positive airway pressure (CPAP) and has been associated with increased risk of hypertension, cardiovascular disease, and all-cause mortality compared with either condition alone [58].

There are a limited number of controlled trials of insomnia therapy in patients with OSA, and the existing studies usually address untreated OSA. Concerns about exacerbation of respiratory status with use of BZRAs have limited their use in this context. However, short-term trials have demonstrated that neither benzodiazepines nor nonbenzodiazepine BZRAs worsen untreated, nonsevere OSA [59,60]; minimum oxygen saturation may be decreased with benzodiazepines in severe OSA.

Patients with mild to moderate sleep apnea benefit from insomnia treatment to a similar degree as those without sleep apnea [61]. Patients with insomnia may struggle more to adjust to sleep apnea treatments such as positive airway pressure (PAP) therapy than those without insomnia [62,63].

Limited prospective data suggest that CBT-I is an effective strategy for improving sleep as well as enhancing CPAP adherence in patients with insomnia and moderate to severe OSA [64-67]. Patients should be monitored closely for increased daytime sleepiness in the initial period of bedtime restriction therapy, if used. (See "Cognitive behavioral therapy for insomnia in adults", section on 'Precautions'.)

Short-term use of sedating medications appears to be safe as a means to improve CPAP adherence and potentially address underlying insomnia in patients with mild-moderate OSA [59]. In one trial, eszopiclone reduced the severity of sleep-disordered breathing and improved sleep during one to two nights of use [68,69]. CPAP treatment adherence and rates of CPAP discontinuation over the first two weeks of treatment also improved, although other studies have shown mixed results. (See "Assessing and managing nonadherence with continuous positive airway pressure (CPAP) for adults with obstructive sleep apnea", section on 'Pharmacological therapy'.)

Restless legs syndrome — Insomnia is nearly universal in patients presenting for treatment of restless legs syndrome (RLS). A single question such as "Do your legs bother you at night?" should be asked of all patients with insomnia to screen for RLS. In older adults, a very delayed sleep phase (eg, sleep onset at 4 AM) may be a clue to an underlying diagnosis of RLS. As with other insomnia contributors, addressing RLS is an important component of successful therapy for sleeplessness.

Gabapentinoids (eg, gabapentin, pregabalin) are preferred as initial agents for RLS in most patients, since long-term use of dopamine agonists commonly worsens the RLS ("augmentation" of symptoms) [70]. Serotonergic agents (eg, selective serotonin reuptake inhibitors, serotonin norepinephrine reuptake inhibitors, mirtazapine) can exacerbate RLS symptoms (table 9). (See "Management of restless legs syndrome and periodic limb movement disorder in adults".)

Of note, patients with RLS may be at increased risk for complex sleep-related behaviors with use of BZRAs such as zolpidem. Treatment-emergence of a sleep-related eating disorder, in particular, is sometimes a clue that a patient has underlying RLS that was not previously recognized. (See "Disorders of arousal from non-rapid eye movement sleep in adults", section on 'Sleep-related eating disorder'.)

Patients with comorbid medical conditions — Sleep disturbance is associated with a wide range of medical conditions. Cumulative number of medical conditions (multimorbidity) predicts rates of sleep disturbance as well as use of sedating medications, independent of age [71,72]. Optimal management of these conditions often improves sleep; however, insomnia may persist even with resolution of the medical condition or with partial, but maximal, medical management.

As bidirectional relationships exist among many medical conditions and insomnia, addressing insomnia may improve both the patient's medical status and their quality of life. Considerations in some of the more common medical comorbid conditions are reviewed in the sections below.

Pain — Pain is one of the most common causes of both acute and chronic insomnia. Sources of pain that may be especially likely to disrupt sleep are gastroesophageal, headache, musculoskeletal, and neuropathic.

Management of the pain is the primary method to improve sleep, and medications directed towards sleep should be reserved until maximal pain control is established. However, as sleeplessness most likely increases pain intensity and frequency, insomnia treatment is an important goal in optimal pain relief.

Many insomnia therapies (eg, CBT, sedating antidepressants, BZRAs, gabapentin/pregabalin) have overlapping indications for various pain syndromes and are reasonable choices in patients with pain for whom a secondary sleep benefit is desired [73,74].

Chronic lung disease — Over 25 percent of patients with chronic obstructive pulmonary disease (COPD) have insomnia disorder [75]. Contributing factors include cough, dyspnea, medication use, comorbid sleep-related breathing disorders, and medical and psychiatric comorbidities.

Polysomnography (PSG) to exclude a sleep-related breathing disorder should be performed in patients who have anatomical risk factors of OSA (eg, obesity, upper airway crowding) and/or more severe overnight oxygen desaturation (or those with cyclical/sawtooth oxygenation), morning headaches after oxygen administration, obesity, or gastroesophageal reflux disease [76]. (See "Sleep-related breathing disorders in COPD".)

Studies on the treatment of insomnia in patients with COPD are limited. In the absence of sleep apnea identified on PSG, CBT-I is encouraged as first-line therapy. Medications are generally reserved for patients who have failed CBT-I, based on concerns about increased vulnerability to adverse effects on respiratory drive, diaphragmatic endurance, and oxygen saturation. Sedating antidepressants, melatonin, and gabapentin may be safer choices than BZRAs in most patients. BZRAs can be used in patients with normocapnic COPD, followed by overnight oximetry monitoring. In a systematic review of five small studies, BZRAs improved objective sleep measures but not subjective sleep in patients with COPD [77].

Renal failure — Insomnia is highly prevalent in patients with chronic kidney disease (CKD). Contributing factors may include pruritus, pain, nocturnal leg cramps, RLS, sleep apnea, comorbid medical illnesses, circadian rhythm alterations, and poor sleep hygiene. Overnight polysomnography should be performed if a sleep-related breathing disorder is suspected based on clinical symptoms (eg, excessive daytime sleepiness, snoring, witnessed apneas during sleep) and risk factors such as obesity and end-stage kidney disease (ESKD). (See "Sleep disorders in end-stage kidney disease", section on 'Sleep apnea'.)

There are no controlled data on CBT-I in patients with CKD, but supportive evidence in the general population is likely generalizable to these patients, who are at increased risk for altered drug clearance and adverse drug effects. Optimization of renal replacement therapy is an important consideration in patients with ESKD. (See "Sleep disorders in end-stage kidney disease", section on 'Treatment'.)

A few small studies have examined pharmacotherapy in patients with CKD. In short-term placebo-controlled studies, zolpidem, zaleplon, and clonazepam all improved sleep quality [78]. Melatonin improved sleep measures in a short-term, but not a one-year, study [79]. When medications are prescribed for sleep, initial low doses and slow upward titration are recommended. Drugs that are renally eliminated should be avoided (eg, gabapentin).

Pregnancy — Sleep disturbance increases during the course of pregnancy, affecting one-quarter of individuals in the first trimester to over two-thirds by the end of the third trimester [80]. Common causes of insomnia also change over time, from nausea, urinary frequency, and backache early in pregnancy to fetal movements, heartburn, leg cramps, RLS, OSA, and physical limitations in achieving a comfortable position by the end of pregnancy. Although many pregnant individuals have disturbed sleep, most do not identify it as a disorder, possibly because they have prepared for it or recognize that it is time limited. Nevertheless, some patients do have more severe insomnia or are more severely disturbed by nighttime awakenings or associated daytime dysfunction.

RLS is important to recognize as it occurs with increased frequency as pregnancy progresses and often interferes with initiation of sleep. Often, the simple prompt "Do your legs bother you when you are lying down at night?" will facilitate diagnosis during pregnancy. Treatment options include oral or intravenous iron supplementation, nonpharmacologic therapies, and medications in selected patients. (See "Restless legs syndrome during pregnancy and lactation".)

There are few controlled data on treatment of insomnia in pregnancy [81]. Nonpharmacologic therapies are the safest option and are preferred over pharmacotherapy by most patients [82-84]. Several randomized trials support the efficacy of CBT-I in pregnancy using in-person or digital delivery methods [85-89].

Over-the-counter sedating antihistamines such as doxylamine or diphenhydramine can be used in patients who desire medication and have no alternative causes of sleeplessness that can be addressed more effectively (eg, RLS, gastroesophageal reflux) [90,91]. Other pharmacologic options, including BZRAs and sedating antidepressants, should generally be avoided as potential risks likely outweigh benefits. (See "Prenatal care: Patient education, health promotion, and safety of commonly used drugs", section on 'Difficulty sleeping'.)

Menopause — Females in the menopausal transition commonly report insomnia. Factors associated with insomnia include vasomotor symptoms (hot flashes and night sweats), reproductive hormonal changes themselves, an increase in obstructive sleep apnea prevalence, and increased risk of comorbid mood and medical disorders. (See "Clinical manifestations and diagnosis of menopause", section on 'Sleep disturbance'.)

Similar to the general population, CBT-I is an effective treatment for menopausal insomnia, both in patients with and without nocturnal hot flashes. In one trial, six-session individual CBT-I delivered by telephone was effective for both overall insomnia severity and difficulty falling and staying asleep (but not for total sleep time) in menopausal insomnia with hot flashes [92]. In another trial, face-to-face multimodality CBT-I and single-modality sleep-restriction therapy (SRT) were each more effective than sleep hygiene education among 150 postmenopausal females with menopause-related chronic insomnia [93,94]. The degree of improvement on sleep outcome measures as well as depression and maladaptive thinking tended to be greatest with CBT-I, and there was trend towards greater insomnia remission rates with CBT-I over SRT.

For patients with hot flashes and sleep disturbances, improvements in sleep have also been observed with a range of pharmacotherapies including menopausal hormone therapy, venlafaxine, gabapentin, fluoxetine, eszopiclone, escitalopram, and suvorexant [95-97]. (See "Menopausal hot flashes", section on 'Management'.)

Yoga and exercise may be beneficial for sleep during the menopausal transition, although the effect size is likely to be small [98,99].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Insomnia in adults".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Insomnia (The Basics)")

Beyond the Basics topics (see "Patient education: Insomnia (Beyond the Basics)" and "Patient education: Insomnia treatments (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Pretreatment assessment – Insomnia often has multiple etiologies (table 2). All patients should be evaluated for predisposing and precipitating factors (eg, depression, anxiety, pain), concomitant medications that may be interfering with sleep, and maladaptive cognitive and behavioral responses to sleeplessness (table 1 and table 3). (See 'Initial assessment and counseling' above.)

Short-term insomnia – Short-term insomnia (<3 months) usually results from psychologic or physiologic stress and is often self-limited, lasting days to weeks.

For patients with mild or manageable levels of distress, we provide education on sleep hygiene (table 3), reassurance, and a plan for clinical follow-up. If insomnia persists at follow-up, we reassess level of distress and proceed as for chronic insomnia below. (See 'Approach to acute insomnia' above.)

When acute insomnia is severe or associated with substantial distress, we offer short-term use of an insomnia medication to help address immediate interference with daytime function and to control escalating anxiety about sleep (algorithm 1). Medication selection is reviewed separately. (See "Pharmacotherapy for insomnia in adults", section on 'Drug selection'.)

Chronic insomnia – Patients with chronic insomnia (≥3 months) have persistent sleep difficulties that last months to years and often follow a waxing and waning course (table 10).

In most patients with chronic insomnia, we suggest cognitive behavioral therapy for insomnia (CBT-I) as first-line therapy, rather than medication (Grade 2B). CBT-I is more efficacious, avoids medication risks, and provides patients with lifelong skills. (See 'Overview of cognitive behavioral therapy' above.)

In patients with severe distress (eg, deterioration in daytime function or excessive anxiety regarding sleeplessness), use of a medication in combination with CBT-I is reasonable, as this may enhance the patient's ability to follow sleep restriction and stimulus control aspects of CBT-I. (See 'Choice of initial therapy' above.)

When medications are used for chronic insomnia, selection should be individualized based on patient age and comorbidities, the type of insomnia complaint, side effect profiles, cost, and clinician and patient preference (algorithm 1 and table 8A-C). Safe prescribing practices may help mitigate common risks of insomnia medications (table 11). (See "Pharmacotherapy for insomnia in adults", section on 'Drug selection'.)

Follow-up and monitoring

Patients on long-term pharmacotherapy should be seen at least every six months to reassess need for medication and reinforce optimal sleep habits. (See 'Response assessment' above.)

Attempts to discontinue sedative medications are a valuable but difficult task, as patients are anxious about the return of sleeplessness. To discontinue long-term therapy, taper slowly (eg, over many months), ideally in conjunction with CBT-I. (See 'Tapering medications' above.)

Special populations – Special populations in whom additional treatment considerations arise include older adults, patients with comorbid psychiatric, neurologic, and sleep disorders, and those with medical comorbidities and conditions. (See 'Treatment considerations for specific populations' above.)

  1. Singareddy R, Vgontzas AN, Fernandez-Mendoza J, et al. Risk factors for incident chronic insomnia: a general population prospective study. Sleep Med 2012; 13:346.
  2. Kalmbach DA, Pillai V, Arnedt JT, et al. Sleep system sensitization: evidence for changing roles of etiological factors in insomnia. Sleep Med 2016; 21:63.
  3. Do D. Trends in the use of medications with insomnia side effects and the implications for insomnia among US adults. J Sleep Res 2020; 29:e13075.
  4. Szmyd B, Rogut M, Białasiewicz P, Gabryelska A. The impact of glucocorticoids and statins on sleep quality. Sleep Med Rev 2021; 55:101380.
  5. American Academy of Sleep Medicine. International Classification of Sleep Disorders, 3rd ed, text revision, American Academy of Sleep Medicine, 2023.
  6. Sateia MJ, Buysse DJ, Krystal AD, et al. Clinical Practice Guideline for the Pharmacologic Treatment of Chronic Insomnia in Adults: An American Academy of Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med 2017; 13:307.
  7. Morin CM, Colecchi C, Stone J, et al. Behavioral and pharmacological therapies for late-life insomnia: a randomized controlled trial. JAMA 1999; 281:991.
  8. Jacobs GD, Pace-Schott EF, Stickgold R, Otto MW. Cognitive behavior therapy and pharmacotherapy for insomnia: a randomized controlled trial and direct comparison. Arch Intern Med 2004; 164:1888.
  9. Morin CM, Vallières A, Guay B, et al. Cognitive behavioral therapy, singly and combined with medication, for persistent insomnia: a randomized controlled trial. JAMA 2009; 301:2005.
  10. Beaulieu-Bonneau S, Ivers H, Guay B, Morin CM. Long-Term Maintenance of Therapeutic Gains Associated With Cognitive-Behavioral Therapy for Insomnia Delivered Alone or Combined With Zolpidem. Sleep 2017; 40.
  11. Qaseem A, Kansagara D, Forciea MA, et al. Management of Chronic Insomnia Disorder in Adults: A Clinical Practice Guideline From the American College of Physicians. Ann Intern Med 2016; 165:125.
  12. Schutte-Rodin S, Broch L, Buysse D, et al. Clinical guideline for the evaluation and management of chronic insomnia in adults. J Clin Sleep Med 2008; 4:487.
  13. Mitchell LJ, Bisdounis L, Ballesio A, et al. The impact of cognitive behavioural therapy for insomnia on objective sleep parameters: A meta-analysis and systematic review. Sleep Med Rev 2019; 47:90.
  14. Wilson SJ, Nutt DJ, Alford C, et al. British Association for Psychopharmacology consensus statement on evidence-based treatment of insomnia, parasomnias and circadian rhythm disorders. J Psychopharmacol 2010; 24:1577.
  15. Brasure M, Fuchs E, MacDonald R, et al. Psychological and Behavioral Interventions for Managing Insomnia Disorder: An Evidence Report for a Clinical Practice Guideline by the American College of Physicians. Ann Intern Med 2016; 165:113.
  16. Riemann D, Espie CA, Altena E, et al. The European Insomnia Guideline: An update on the diagnosis and treatment of insomnia 2023. J Sleep Res 2023; 32:e14035.
  17. Matthews EE, Arnedt JT, McCarthy MS, et al. Adherence to cognitive behavioral therapy for insomnia: a systematic review. Sleep Med Rev 2013; 17:453.
  18. Troxel WM, Conrad TS, Germain A, Buysse DJ. Predictors of treatment response to brief behavioral treatment of insomnia (BBTI) in older adults. J Clin Sleep Med 2013; 9:1281.
  19. Bathgate CJ, Edinger JD, Krystal AD. Insomnia Patients with Objective Short Sleep Duration have a Blunted Response to Cognitive Behavioral Therapy for Insomnia. Sleep 2016.
  20. Huedo-Medina TB, Kirsch I, Middlemass J, et al. Effectiveness of non-benzodiazepine hypnotics in treatment of adult insomnia: meta-analysis of data submitted to the Food and Drug Administration. BMJ 2012; 345:e8343.
  21. Edinger JD, Arnedt JT, Bertisch SM, et al. Behavioral and psychological treatments for chronic insomnia disorder in adults: an American Academy of Sleep Medicine clinical practice guideline. J Clin Sleep Med 2021; 17:255.
  22. Morin CM, Inoue Y, Kushida C, et al. Endorsement of European guideline for the diagnosis and treatment of insomnia by the World Sleep Society. Sleep Med 2021; 81:124.
  23. Perlis ML, Grandner MA, Chakravorty S, et al. Suicide and sleep: Is it a bad thing to be awake when reason sleeps? Sleep Med Rev 2016; 29:101.
  24. Avidan AY, Fries BE, James ML, et al. 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 2005; 53:955.
  25. Zhao JL, Cross N, Yao CW, et al. Insomnia disorder increases the risk of subjective memory decline in middle-aged and older adults: a longitudinal analysis of the Canadian Longitudinal Study on Aging. Sleep 2022; 45.
  26. Roth T, Jaeger S, Jin R, et al. Sleep problems, comorbid mental disorders, and role functioning in the national comorbidity survey replication. Biol Psychiatry 2006; 60:1364.
  27. Bélanger L, Belleville G, Morin C. Management of Hypnotic Discontinuation in Chronic Insomnia. Sleep Med Clin 2009; 4:583.
  28. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5), American Psychiatric Association, Arlington, VA 2013.
  29. Fava M, McCall WV, Krystal A, et al. Eszopiclone co-administered with fluoxetine in patients with insomnia coexisting with major depressive disorder. Biol Psychiatry 2006; 59:1052.
  30. Pollack M, Kinrys G, Krystal A, et al. Eszopiclone coadministered with escitalopram in patients with insomnia and comorbid generalized anxiety disorder. Arch Gen Psychiatry 2008; 65:551.
  31. McClintock SM, Husain MM, Wisniewski SR, et al. Residual symptoms in depressed outpatients who respond by 50% but do not remit to antidepressant medication. J Clin Psychopharmacol 2011; 31:180.
  32. Sylvia LG, Dupuy JM, Ostacher MJ, et al. Sleep disturbance in euthymic bipolar patients. J Psychopharmacol 2012; 26:1108.
  33. Alberti S, Chiesa A, Andrisano C, Serretti A. Insomnia and somnolence associated with second-generation antidepressants during the treatment of major depression: a meta-analysis. J Clin Psychopharmacol 2015; 35:296.
  34. Manber R, Edinger JD, Gress JL, et al. Cognitive behavioral therapy for insomnia enhances depression outcome in patients with comorbid major depressive disorder and insomnia. Sleep 2008; 31:489.
  35. Harvey AG, Soehner AM, Kaplan KA, et al. Treating insomnia improves mood state, sleep, and functioning in bipolar disorder: a pilot randomized controlled trial. J Consult Clin Psychol 2015; 83:564.
  36. Manber R, Buysse DJ, Edinger J, et al. Efficacy of Cognitive-Behavioral Therapy for Insomnia Combined With Antidepressant Pharmacotherapy in Patients With Comorbid Depression and Insomnia: A Randomized Controlled Trial. J Clin Psychiatry 2016; 77:e1316.
  37. Sadler P, McLaren S, Klein B, et al. Cognitive behavior therapy for older adults with insomnia and depression: a randomized controlled trial in community mental health services. Sleep 2018; 41.
  38. Irwin MR, Olmstead R. The Direct Effect of Cognitive Behavioral Therapy for Insomnia on Depression Prevention and the Mediation Effect via Insomnia Remission-Reply. JAMA Psychiatry 2022; 79:515.
  39. Belleville G, Guay S, Marchand A. Persistence of sleep disturbances following cognitive-behavior therapy for posttraumatic stress disorder. J Psychosom Res 2011; 70:318.
  40. Talbot LS, Maguen S, Metzler TJ, et al. Cognitive behavioral therapy for insomnia in posttraumatic stress disorder: a randomized controlled trial. Sleep 2014; 37:327.
  41. Freeman D, Waite F, Startup H, et al. Efficacy of cognitive behavioural therapy for sleep improvement in patients with persistent delusions and hallucinations (BEST): a prospective, assessor-blind, randomised controlled pilot trial. Lancet Psychiatry 2015; 2:975.
  42. Tek C, Palmese LB, Krystal AD, et al. The impact of eszopiclone on sleep and cognition in patients with schizophrenia and insomnia: a double-blind, randomized, placebo-controlled trial. Schizophr Res 2014; 160:180.
  43. Hasler BP, Martin CS, Wood DS, et al. A longitudinal study of insomnia and other sleep complaints in adolescents with and without alcohol use disorders. Alcohol Clin Exp Res 2014; 38:2225.
  44. Drummond SP, Gillin JC, Smith TL, DeModena A. The sleep of abstinent pure primary alcoholic patients: natural course and relationship to relapse. Alcohol Clin Exp Res 1998; 22:1796.
  45. Currie SR, Clark S, Hodgins DC, El-Guebaly N. Randomized controlled trial of brief cognitive-behavioural interventions for insomnia in recovering alcoholics. Addiction 2004; 99:1121.
  46. Arnedt JT, Conroy DA, Armitage R, Brower KJ. Cognitive-behavioral therapy for insomnia in alcohol dependent patients: a randomized controlled pilot trial. Behav Res Ther 2011; 49:227.
  47. Roth T, Coulouvrat C, Hajak G, et al. Prevalence and perceived health associated with insomnia based on DSM-IV-TR; International Statistical Classification of Diseases and Related Health Problems, Tenth Revision; and Research Diagnostic Criteria/International Classification of Sleep Disorders, Second Edition criteria: results from the America Insomnia Survey. Biol Psychiatry 2011; 69:592.
  48. Alessi C, Vitiello MV. Insomnia (primary) in older people: non-drug treatments. BMJ Clin Evid 2015; 2015.
  49. Irwin MR, Carrillo C, Sadeghi N, et al. Prevention of Incident and Recurrent Major Depression in Older Adults With Insomnia: A Randomized Clinical Trial. JAMA Psychiatry 2022; 79:33.
  50. Kyle SD, Miller CB, Rogers Z, et al. Sleep restriction therapy for insomnia is associated with reduced objective total sleep time, increased daytime somnolence, and objectively impaired vigilance: implications for the clinical management of insomnia disorder. Sleep 2014; 37:229.
  51. Siu PM, Yu AP, Tam BT, et al. Effects of Tai Chi or Exercise on Sleep in Older Adults With Insomnia: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e2037199.
  52. Irwin MR, Olmstead R, Motivala SJ. Improving sleep quality in older adults with moderate sleep complaints: A randomized controlled trial of Tai Chi Chih. Sleep 2008; 31:1001.
  53. Irwin MR, Olmstead R, Carrillo C, et al. Cognitive behavioral therapy vs. Tai Chi for late life insomnia and inflammatory risk: a randomized controlled comparative efficacy trial. Sleep 2014; 37:1543.
  54. Yang PY, Ho KH, Chen HC, Chien MY. Exercise training improves sleep quality in middle-aged and older adults with sleep problems: a systematic review. J Physiother 2012; 58:157.
  55. Hein M, Lanquart JP, Loas G, et al. Prevalence and risk factors of moderate to severe obstructive sleep apnea syndrome in insomnia sufferers: a study on 1311 subjects. Respir Res 2017; 18:135.
  56. Fung CH, Martin JL, Dzierzewski JM, et al. Prevalence and symptoms of occult sleep disordered breathing among older veterans with insomnia. J Clin Sleep Med 2013; 9:1173.
  57. Sweetman A, Lack L, Crawford M, Wallace DM. Comorbid Insomnia and Sleep Apnea: Assessment and Management Approaches. Sleep Med Clin 2022; 17:597.
  58. Lechat B, Appleton S, Melaku YA, et al. Comorbid insomnia and sleep apnoea is associated with all-cause mortality. Eur Respir J 2022; 60.
  59. Mason M, Cates CJ, Smith I. Effects of opioid, hypnotic and sedating medications on sleep-disordered breathing in adults with obstructive sleep apnoea. Cochrane Database Syst Rev 2015; :CD011090.
  60. Carter SG, Carberry JC, Cho G, et al. Effect of 1 month of zopiclone on obstructive sleep apnoea severity and symptoms: a randomised controlled trial. Eur Respir J 2018; 52.
  61. Fung CH, Martin JL, Josephson K, et al. Efficacy of Cognitive Behavioral Therapy for Insomnia in Older Adults With Occult Sleep-Disordered Breathing. Psychosom Med 2016; 78:629.
  62. Wallace DM, Sawyer AM, Shafazand S. Comorbid insomnia symptoms predict lower 6-month adherence to CPAP in US veterans with obstructive sleep apnea. Sleep Breath 2018; 22:5.
  63. Wickwire EM, Smith MT, Birnbaum S, Collop NA. Sleep maintenance insomnia complaints predict poor CPAP adherence: A clinical case series. Sleep Med 2010; 11:772.
  64. Ong JC, Crawford MR, Dawson SC, et al. A randomized controlled trial of CBT-I and PAP for obstructive sleep apnea and comorbid insomnia: main outcomes from the MATRICS study. Sleep 2020; 43:zsaa041.
  65. Alessi CA, Fung CH, Dzierzewski JM, et al. Randomized controlled trial of an integrated approach to treating insomnia and improving the use of positive airway pressure therapy in veterans with comorbid insomnia disorder and obstructive sleep apnea. Sleep 2021; 44:zsaa235.
  66. Sweetman A, Lack L, Catcheside PG, et al. Cognitive and behavioral therapy for insomnia increases the use of continuous positive airway pressure therapy in obstructive sleep apnea participants with comorbid insomnia: a randomized clinical trial. Sleep 2019; 42.
  67. Sweetman A, Farrell S, Wallace DM, Crawford M. The effect of cognitive behavioural therapy for insomnia in people with comorbid insomnia and sleep apnoea: A systematic review and meta-analysis. J Sleep Res 2023; 32:e13847.
  68. Lettieri CJ, Shah AA, Holley AB, et al. Effects of a short course of eszopiclone on continuous positive airway pressure adherence: a randomized trial. Ann Intern Med 2009; 151:696.
  69. Lettieri CJ, Quast TN, Eliasson AH, Andrada T. Eszopiclone improves overnight polysomnography and continuous positive airway pressure titration: a prospective, randomized, placebo-controlled trial. Sleep 2008; 31:1310.
  70. Allen RP, Chen C, Garcia-Borreguero D, et al. Comparison of pregabalin with pramipexole for restless legs syndrome. N Engl J Med 2014; 370:621.
  71. Koyanagi A, Garin N, Olaya B, et al. Chronic conditions and sleep problems among adults aged 50 years or over in nine countries: a multi-country study. PLoS One 2014; 9:e114742.
  72. Linnet K, Gudmundsson LS, Birgisdottir FG, et al. Multimorbidity and use of hypnotic and anxiolytic drugs: cross-sectional and follow-up study in primary healthcare in Iceland. BMC Fam Pract 2016; 17:69.
  73. McCurry SM, Zhu W, Von Korff M, et al. Effect of Telephone Cognitive Behavioral Therapy for Insomnia in Older Adults With Osteoarthritis Pain: A Randomized Clinical Trial. JAMA Intern Med 2021; 181:530.
  74. Chang JR, Cheung YK, Sharma S, et al. Comparative effectiveness of non-pharmacological interventions on sleep in individuals with chronic musculoskeletal pain: A systematic review with network meta-analysis. Sleep Med Rev 2024; 73:101867.
  75. Budhiraja R, Parthasarathy S, Budhiraja P, et al. Insomnia in patients with COPD. Sleep 2012; 35:369.
  76. Owens RL, Macrea MM, Teodorescu M. The overlaps of asthma or COPD with OSA: A focused review. Respirology 2017; 22:1073.
  77. Lu XM, Zhu JP, Zhou XM. The effect of benzodiazepines on insomnia in patients with chronic obstructive pulmonary disease: a meta-analysis of treatment efficacy and safety. Int J Chron Obstruct Pulmon Dis 2016; 11:675.
  78. Scherer JS, Combs SA, Brennan F. Sleep Disorders, Restless Legs Syndrome, and Uremic Pruritus: Diagnosis and Treatment of Common Symptoms in Dialysis Patients. Am J Kidney Dis 2017; 69:117.
  79. Russcher M, Koch BC, Nagtegaal JE, et al. Long-term effects of melatonin on quality of life and sleep in haemodialysis patients (Melody study): a randomized controlled trial. Br J Clin Pharmacol 2013; 76:668.
  80. Schweiger MS. Sleep disturbance in pregnancy. A subjective survey. Am J Obstet Gynecol 1972; 114:879.
  81. Bacaro V, Benz F, Pappaccogli A, et al. Interventions for sleep problems during pregnancy: A systematic review. Sleep Med Rev 2020; 50:101234.
  82. Okun ML, Ebert R, Saini B. A review of sleep-promoting medications used in pregnancy. Am J Obstet Gynecol 2015; 212:428.
  83. Sedov I, Madsen JW, Goodman SH, Tomfohr-Madsen LM. Couples' treatment preferences for insomnia experienced during pregnancy. Fam Syst Health 2019; 37:46.
  84. Sedov ID, Goodman SH, Tomfohr-Madsen LM. Insomnia Treatment Preferences During Pregnancy. J Obstet Gynecol Neonatal Nurs 2017; 46:e95.
  85. Manber R, Bei B, Simpson N, et al. Cognitive Behavioral Therapy for Prenatal Insomnia: A Randomized Controlled Trial. Obstet Gynecol 2019; 133:911.
  86. Tomfohr-Madsen LM, Clayborne ZM, Rouleau CR, Campbell TS. Sleeping for Two: An Open-Pilot Study of Cognitive Behavioral Therapy for Insomnia in Pregnancy. Behav Sleep Med 2017; 15:377.
  87. Felder JN, Epel ES, Neuhaus J, et al. Efficacy of Digital Cognitive Behavioral Therapy for the Treatment of Insomnia Symptoms Among Pregnant Women: A Randomized Clinical Trial. JAMA Psychiatry 2020; 77:484.
  88. Kalmbach DA, Cheng P, O'Brien LM, et al. A randomized controlled trial of digital cognitive behavioral therapy for insomnia in pregnant women. Sleep Med 2020; 72:82.
  89. Bei B, Pinnington DM, Quin N, et al. Improving perinatal sleep via a scalable cognitive behavioural intervention: findings from a randomised controlled trial from pregnancy to 2 years postpartum. Psychol Med 2023; 53:513.
  90. Miller MA, Mehta N, Clark-Bilodeau C, Bourjeily G. Sleep Pharmacotherapy for Common Sleep Disorders in Pregnancy and Lactation. Chest 2020; 157:184.
  91. Khazaie H, Ghadami MR, Knight DC, et al. Insomnia treatment in the third trimester of pregnancy reduces postpartum depression symptoms: a randomized clinical trial. Psychiatry Res 2013; 210:901.
  92. McCurry SM, Guthrie KA, Morin CM, et al. Telephone-Based Cognitive Behavioral Therapy for Insomnia in Perimenopausal and Postmenopausal Women With Vasomotor Symptoms: A MsFLASH Randomized Clinical Trial. JAMA Intern Med 2016; 176:913.
  93. Drake CL, Kalmbach DA, Arnedt JT, et al. Treating chronic insomnia in postmenopausal women: a randomized clinical trial comparing cognitive-behavioral therapy for insomnia, sleep restriction therapy, and sleep hygiene education. Sleep 2019; 42.
  94. Kalmbach DA, Cheng P, Arnedt JT, et al. Treating insomnia improves depression, maladaptive thinking, and hyperarousal in postmenopausal women: comparing cognitive-behavioral therapy for insomnia (CBTI), sleep restriction therapy, and sleep hygiene education. Sleep Med 2019; 55:124.
  95. Attarian H, Hachul H, Guttuso T, Phillips B. Treatment of chronic insomnia disorder in menopause: evaluation of literature. Menopause 2015; 22:674.
  96. Cheng YS, Tseng PT, Wu MK, et al. Pharmacologic and hormonal treatments for menopausal sleep disturbances: A network meta-analysis of 43 randomized controlled trials and 32,271 menopausal women. Sleep Med Rev 2021; 57:101469.
  97. Rahman SA, Nathan MD, Wiley A, et al. A double-blind, randomized, placebo-controlled trial of suvorexant for the treatment of vasomotor symptom-associated insomnia disorder in midlife women. Sleep 2022; 45:zsac007.
  98. Newton KM, Reed SD, Guthrie KA, et al. Efficacy of yoga for vasomotor symptoms: a randomized controlled trial. Menopause 2014; 21:339.
  99. Sternfeld B, Guthrie KA, Ensrud KE, et al. Efficacy of exercise for menopausal symptoms: a randomized controlled trial. Menopause 2014; 21:330.
Topic 97867 Version 31.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟