Select a psychoactive drug that is of pharmacological interest to you, but not one you will review as part of your Critical Review. For this paper, you may choose drugs of abuse; however, the paper mu

Clinical Medicine Reviews in Therapeutics 2013:5 11–23 doi: 10.4137/CMRT.S1026 5 This article is available from http://www.la-press.co m. © Libertas Academica Ltd.

Clinical Medicine Reviews in Therapeutic s Review Clinical Medicine Reviews in Therapeutics 2013:5 11 Current and Emerging Pharmacotherapies in the Management of Insomnia in Adults Pamela Song 1 and Sooyeon Suh 2,3 1Department of Neurology, ilsan Paik Hospital, inje University, Goyang, Korea. 2Korea University Ansan Hospital, institute of Human Genomic Studies, Danwon-gu, Ansan-si, South Korea. 3Stanford University, Department of Psychiatry, Redwood City, CA, USA. Corresponding author email: [email protected] u Abstract: This review primarily focuses on the pharmacotherapy for insomnia, including current and emerging pharmacotherapy for insomnia. Currently, there are 10 FDA-approved drugs for the treatment of insomnia. The mainstay treatments are benzodiazepine receptor agonists, which are now available in controlled release formulation to support sleep maintenance. Melatonin receptor agonists are usually used for sleep initiation insomnia. Doxepin an antidepressant is approved for treatment of insomnia. Anticonvulsants and antipsychotics are used ‘off-label’ to treat insomnia, despite limited data to support its use. 5-HT2A serotonin receptor inverse agonists and orexin receptor antagonists are in Phase II and III clinical trials \ for insomnia with different mechanisms of action.

When treating insomnia patients, it is important to individualize treatment by considering various comorbid conditions. Further research is needed to evaluate the efficacy and safety of newly emerging medications, as well as medications that are frequently used without estab lished clinical evidence. Keywords: insomnia, pharmacotherapy, treatment Song and Suh 12 Clinical Medicine Reviews in Therapeutics 2013:5 Background Insomnia is one of the most common sleep disorders, with population-based studies indicating prevalence in the U.S. to be between 6%–10%. 1,2 Up to 30% of Americans report experiencing at least one insomnia symptom. 3 Insomnia is characterized by a combina - tion of nocturnal symptoms, including difficulties initiating sleep, maintaining sleep and early morn - ing awakenings, and daytime symptoms that impair occupational, social, or other areas of functioning.

Insomnia can exist alone or in conjunction with comorbid medical and/or psychiatric conditions. Insomnia is recognized as an independent disorder in all diagnostic classification systems. The Interna - tional Classification of Sleep Disorders, 2nd edition (ICSD) and the Diagnostic and Statistical Manual for Mental Disorders 4th edition (DSM-IV) include diagnostic criteria of insomnia as having both noctur - nal symptoms (ie, difficulty initiating or maintaining sleep, early morning awakenings, or nonrestorative sleep) and daytime symptoms characterized by impairment of daytime functioning (ie, difficulties with memory or concentration that are related to sleep difficulty) accompanied by marked distress due to sleep disturbance. There are pharmacological and non-pharmacolog - ical treatment options available for insomnia. This review primarily focuses on the pharmacotherapy for insomnia, including current and emerging pharmaco - therapy for insomnia. Non-pharmacological options include cognitive-behavioral therapy for insomnia (CBTI), which is an effective treatment of insom - nia, alone and in combination with pharmacotherapy.

CBTI is a short-term treatment that, on average, includes 4–8 sessions and adopts a multicomponent approach, which typically includes sleep restriction, stimulus control, cognitive therapy, sleep hygiene and relaxation training, and can help patients learn how to fall asleep quicker and improve sleep quality. While a full review of non-pharmacological treatments is beyond the scope of this article, see Siebern et al for a review. 4 Pharmacological Treatment for Insomnia Pharmacotherapy has been the treatment of choice for insomnia for more than 100 years, beginning with bar - biturates in the early 1900s. Treatment is recommended when insomnia has a significant impact on a patient’s sleep quality, health, daytime functioning or comorbid conditions. 5 Pharmacological treatment for insomnia has its basis in neurotransmitter systems affecting sleep and wake promoting systems. Wake-promoting neu - rotransmitters include Norepinephrine (NE), Serotonin (5-HT), Acetylcholine (Ach), Histamine (HA) and Hypocretin/Orexin (HOX). Sleep-promoting inhibi - tory neurotransmitters consist of Adenosine (AD), Gamma-aminobutyric acid (GABA), Galanin, and Melatonin (MT). A mutually inhibitory relationship exists between the sleep-promoting systems and the wake-promoting systems. 6 The treatment agents used to treat insomnia antagonize wake-promoting systems or enhance sleep-promoting systems. Pharmacological agents commonly used to treat insomnia include benzodiazepine-receptor agonists (BzRAs; ben - zodiazepines and nonbenzodiazepines), melatonin receptor agonists, antidepressants, antipsychotics, anticonvulsants, antihistamines, 5-HT2A serotonin inverse agonists, and orexin receptor antagonists.

The Food and Drug Administration (FDA) currently approve 10 medications for the treatment of insomnia (See Table 1). Benzodiazepine receptor agonists Benzodiazepine receptor agonists (BzRAs) are the allosteric modulators of gamma-aminobutyric acid type receptor (GABA) benzodiazepine recep - tor (BzR). 7 When BzRAs bind to benzodiazepine receptors (BzR), these compounds enhance inhibi - tory action of gamma-aminobutyric acid (GABA) by opening the chloride channel, which then hyperpolar - izes the neuron and leads to the inhibition of neuronal action potential firing. 7 GABA is the primary inhibi - tory neurotransmitter in the brain and exerts its action by binding at two distinct types of GABA receptors (A and B). 8 The GABA A receptors have a 5-protein transmembrane channel constructing a pentameric structure. The α, β, and γ subunits occur in different combinations within particular locations of the brain, resulting in substantial diverse effects of GABA on brain function. 9 The sleep-enhancing effects of both benzodiazepines and non-benzodiazepine result from their binding to the GABA A complex at a site on the α subunits. 9 These agents differ in their affinity for different alpha subunits ( α1– α6); of these, α1, α2, and α3 are most abundant in the brain. 10–12 Binding to Pharmacotherapies of insomnia Clinical Medicine Reviews in Therapeutics 2013:5 13 Tab le 1.

Characteristics of FDA-approved insomnia pharmacological agents. Generic name Trade name Type Elimination half-life (hours) Tmax (hours) Major me tab olic pathway Available tab lets (mg) Recommended dose for adults (mg) Habit forming Zaleplon Sonata Pyrazolopyrimidine 0.9–1.1 1.1 Aldehyde oxidase, CYP3A4 5, 15 10 Yes Zolpidem tartrate Ambien Ambien CR edluar intermezzo Zolpimist imidazopyrimidine 2.0–5.5 1.7–2.5 CYP3A4, CYP1A2, CYP2C9 5, 10 6.25, 12.5 10 Schedule iv drug eszopliclone Lunesta Cyclopyrrolone 6–7 1.3–1.6 CYP3A1, CYP2e1 1, 2 2–3 Schedule iv drug Triazolam Halcion Benzodiazepine 2–5.5 1–3 CYP3A4, glucuronide conjugation 0.125, 0.25 0.25 Schedule iv drug Temazepam Restoril Benzodiazepine 8–20 1–3 Glucuronide conjugation 7, 5, 15, 30 30 Schedule iv drug estazolam ProSom Benzodiazepine 10–24 1.5–2.0 CYP3A4 1, 2 2 Schedule iv drug Flurazepam Dalmane Benzodiazepine 40–250 0.5–1.5 CYP2C19, CYP3A4 15, 30 30 Schedule iv drug Quazepam Doral Benzodiazepine -20–120 2 CYP3A4, CYP2C19 7.5, 15 7.5–15 Schedule iv drug Ramelteon Rozerem Melatonin receptor agonist 0.8–2 0.7–0.95 CYP1A2, CYP2C, CYP3A4 8 8 No Doxepin Silenor Tricyclic antidepressant 0–50 1.5–4 CYP3A4, CYP2C19, CYP2D6, CYP2C9, CYP1A2 3, 6 3–6 No Notes:

*Schedule iv drug: According to the Controlled Substances Act, the medication has a low potential for abuse compared to Schedule iii substances, but abuse may lead to limited physical dependence or psychological dependence; This risk may be greater with higher dosages of the medication. Song and Suh 14 Clinical Medicine Reviews in Therapeutics 2013:5 the α1 subunit appears to have the potential for anti - convulsant, amnestic and ataxic effects, in addition to sleep-enhancing effects. Binding to the α2 or α3 sub - units may have anxiolytic and myorelaxant effects, while binding to the α5 subunits may lead to myo - relaxation, or cognitive impairment. 10–14 The benzo - diazepine non-selectively binds to α1– α5 subunits. 7 Non-benzodiazepine BzRAs have a more selective profile, binding preferentially to α1 subunits for sedating effects. 11 The mechanism for the predominant treatment of insomnia is enhancing the inhibitory effects of GABA to promote sleep. These include benzodiaz - epine and non-benzodiazepine BzRAs. There are currently 5 benzodiazepines that are FDA-approved for the treatment of insomnia. These are estazolam (ProSom), flurazepam (Dalmane), quazepam (Doral), temazepam (Restoril), and triazolam (Halcion). There are individual differences in the pharmacokinetic and pharmacodynamic actions of BzRAs; thus, the clinical efficacy and safety profile may vary among individuals. Despite the variation in rapidity of onset and duration of hypnotic action, benzodiazepines have been shown to be efficacious for the treatment of insomnia. Meta-analyses of benzodiazepines have been reported to improve sleep and wake complaints, daytime functioning or distress, and PSG measures of reduction in the sleep latency (SL), number of arous - als, and wake after sleep onset (WASO), as well as increase total sleep time (TST). 15,16 Although meta- analyses are helpful in describing general effects of these medications because they combine studies of various agents at multiple doses, they are less helpful in guiding specific treatment decisions. Non-benzodiazepine BzRAs that are currently FDA-approved include zolpidem tartrate, zaleplon, and eszopiclone for the short-term treatment of insomnia. These drugs demonstrate hypnotic efficacy similar to that of benzodiazepines along with better safety profiles. Non-benzodiazepine BzRAs gener - ally cause less disruption of normal sleep architecture than benzodiazepines. 17 Psychomotor and memory impairment may be less problematic, especially when compared to longer-acting benzodiazepines. Zolpidem tartrate preferably binds to α1 subunits of GABA A receptor, and the selective binding pro - duces sedation without interfering with other benzo - diazepine properties. Zolpidem tartrate is currently sold in four different forms based on indication and delivery method; zolpidem tartrate immedi - ate release (Ambien ®), zolpidem tartrate controlled release (Ambien CR ®), zolpidem tartrate oral solution (Zolpimist ®), and zolpidem tartrate sublingual tab let (Intermezzo ®, Edluar ®). Zolpidem tartrate was approved by the FDA (U.S. Food and Drug Administration) for the short-term treatment of insomnia. Its bioavailability after oral administration is 70%, it has a half-life of 1.5–2.4 hours and its effects can last up to 8 hours. 18 It is excreted as inactive me tab olites mainly by the kidney (56%) and enterally (37%). The excretion is reduced in the elderly, and in patients with hepatic and renal impairment.

The effectiveness of zolpidem, compare to placebo, has been shown to improve SL, sleep duration, and efficacy, with persistent improvements throughout 35 days 19 and up to 12 months of follow up. 20 Head - aches are the most commonly reported adverse effect, which appears to be dose-dependent. A small percent - age of patients experience CNS-related effects such as drowsiness, incoordination, dizziness, hallucina - tions and ataxia. Sleep-related events such as pre - paring and eating food, compulsive house cleaning, sleep driving, house painting, and sleepwalking have been reported. 21–23 These reports indicate that patients typically do not have a memory of the sleep-related event, and in most cases, the behavior resolved when zolpidem was discontinued. Sudden withdrawal from zolpidem may precipitate an epileptic seizure. Most of the case reports indicated that zolpidem withdrawal seizures occur after withdrawal of supratherapeutic doses ranging from 130 mg to 600 mg per day. 24–27 The controlled release formulation of zolpidem tar - trate is sold under the trade name Ambien-CR ®, which is a 2-layer tab let that provides a biphasic release zolpidem: an immediate release phase followed by a prolonged release phase. This allows immedi - ate release of 60% of the dose, with the remainder being released at a slower rate. 28 Zolpidem controlled release (12.5 mg) produces plasma concentrations that are higher in the middle of the typical sleep cycle than with 10 mg zolpidem immediate release. This, in turn, produces a benzodiazepine agonist effects in the 3–6 hour postdosage. 18 There is one double-blind pla - cebo controlled study of zolpidem controlled release in chronic primary insomnia for 6 months. This study revealed that zolpidem controlled release for - Pharmacotherapies of insomnia Clinical Medicine Reviews in Therapeutics 2013:5 15 mulation provided sustained improvements in sleep onset and maintenance compare to placebo. Thus, it is FDA approved for sleep onset and/or sleep main - tenance insomnia. 29 Zolpimist ® is an oral solution of zolpidem tartrate, currently marketed as Ambien ®, as an oral spray. 30 It is approved by the U.S. FDA with indication for the short-term treatment of insom - nia characterized by difficulty with sleep initiation.

Zolpimist is considered bioequivalent to the imme - diate release tab let formulation, without having to ingest with water. 31 Zolpimist has slightly more rapid and peak serum concentration, which leads to faster onset of action. Each spray delivers 5 mg of zolpidem tartrate in 100 mcL. Clinically recommended doses are 10 mg for adults and 5 mg for the elderly once daily, immediately before bedtime. 31 Intermezzo ® is a low-dose sublingual tab let of zolpidem tartrate, and is FDA approved for sleep maintenance insom - nia, especially for middle-of-the-night awakening. 30 This formulation contains a lower dose of the drug (1.75 mg and 3.5 mg tab let), and is recommended only if there is at least 4 more hours before the patient’s planned awakening. Recommended dose is 1.75 mg for women, and 3.5 mg for men, once per night if needed. Edluar ® is a sublingual tab let of zolpidem tartrate. It has been reported to initiate sleep signifi - cantly earlier as compared to an equivalent dose of oral zolpidem, 32 and also has more convenient admin - istration. It is FDA-approved for short-term treatment of insomnia characterized by difficulties with sleep initiation. It is supplied as a 5 mg or 10 mg sublingual tab let. The recommended doses are 5 mg for elderly and 10 mg for adults, once daily immediately before getting into bed. Zaleplon (Sonata ®) was approved by the FDA for the short-term treatment of insomnia. Like zolpidem, it acts as a selective agonist on the GABA A receptor, but with low affinity. 33 It has a short half-life of about 0.9–1.1 hours and effects usually last for up to 4 hours.

Considering the short half-life of Zaleplon, it was pri - marily indicated for the short-treatment of insomnia.

Clinical trials suggest that Zaleplon is also useful for patients who have frequent nocturnal awakenings (sleep maintenance insomnia) because it can be taken for middle-of-the-night insomnia, as long as it is taken 4 hours or more before expected wake up time, without residual sedating effects. 34 A double-blind, placebo controlled, crossover dosing study revealed that zolpidem (10 mg) and zaleplon (10 mg) effec - tively shortened SL and increased TST compared to placebo. Residual sedation was not detected as little as 4 hours after zaleplon 10 mg, whereas residual seda - tion was detected with zolpidem. 35 Rebound insomnia after discontinuing nightly treatment with zaleplon has been investigated in a double-blind design study for up to 5 weeks, and rebound insomnia has not been found in these trials. 36,37 Similarly, no withdrawal syndrome was identified following discontinuation of nightly zaleplon use. 36 Eszopiclone (Lunesta ®), another non-benzodi - azepine BzRA, has high relative affinity for α2, and α3 subunits. 38 It is me tab olized by cytochrome P450-3A (CYP3A). Eszopiclone is rapidly absorbed and has a half-life of 6–7 hours, longer than the other nonbenzodiazepine BzRAs. 39 Eszopiclone consistently improves sleep maintenance relative to placebo, based on measures of shortened wake time after sleep onset, and prolonged TST. 40–42 However, eszopiclone may also produce residual sedation and impairment of driv - ing performance in the initial waking hours. It should therefore be prescribed to patients who expect to spend 8 hours or more in bed after ingestion. Eszopiclone has also been associated with dysgeusia (bitter taste), although this is not considered an adverse effect. FDA-approved benzodiazepines are generally recommended for short-term use, as long-term use may result in adverse events, such as residual next day sedation, impaired motor and cognitive func - tion, frequent falls for elderly, amnesia and rebound daytime anxiety. 43,44 Residual sedating effects occur more commonly with long acting benzodiazepines.

Tolerance is defined as a reduction of a drug’s effect with repeated administration of a s tab le dose, or the need to increase the dose to maintain effects for repeated use. Several randomized double-blind stud - ies assessed the effects of continuous sedative hyp - notic use: zolpidem for 5 weeks, 19 zaleplon for 4 33 and 5 weeks, 37 and eszopiclone for 6 18 and 12 months. 41,45 In long-term use of benzodiazepines (2 years), no no tab le dose escalation were seen. Subgroups with a higher risk of dose escalation included antidepressant recipients and patients who filled duplicate prescrip - tions at different pharmacies. 46 Rebound insomnia is a common discontinua - tion effect of BZRA hypnotics, which is defined as a worsening of sleep relative to the patient’s status Song and Suh 16 Clinical Medicine Reviews in Therapeutics 2013:5 before starting treatment. It typically lasts for 1 ∼2 nights immediately after hypnotic discontinuation, and is more likely to occur after abrupt discontinua - tion of the shorter-acting agents. 47 Rebound insomnia is different from recrudescence, which is a return of symptoms to pretreatment level, and withdrawal syn - drome, which is the appearance of new symptoms. Abuse liability has been a concern as chronic use of BZRA may lead to their abuse following behav - ioral and/or physical dependence. Patients who use chronic hypnotics rarely self-escalate the dose and use it for nontherapeutic (ex. daytime) purposes. There is one study with a follow-up period of 12 months of nightly zolpidem use, which reported no evi - dence of dose escalation. 20 In contrast, the placebo group increased their placebo dose. This suggests that hypnotic self-administration can be explained by therapy-seeking behavior, and does not reflect behavioral dependence. While abuse potential of BZRA (zolpidem, and eszopiclone) seem to be very low, individuals with a history of substance abuse or dependence, or psychiatric comorbidities are at risk. 48 Melatonin receptor ligands Melatonin is a hormone secreted by the pineal gland in the brain, synthesized from serotonin. It is involved in circadian system regulation and has sleep-enhancing effects. There are 3 different melatonin (MT) recep - tors (MT1, MT2, and MT3). Agonists of MT1 are thought to induce sleepiness, whereas MT2 receptors are responsible for regulation of circadian rhythms.

These receptors are primarily located in the suprachi - asmatic nucleus (SCN) and binding to MT1 receptors could attenuate SCN stimulatory output (alerting sig - nal), subsequently promoting a hypnotic effect. 49,50 Melatonin is an over-the-counter (OTC) medi - cation, sold as a food supplement. It has a time to maximum concentration (T max ) of 0.5 hours and an elimination half-life of roughly 1 hour. 51 A dose-response relationship does not appear to exist for melatonin, and the means to determine optimal dosing are lacking. 17 There is controversy regarding the sleep-promoting effects of melatonin because of widely varying inclusion and exclusion criteria, melatonin dosages, and timing of administration. 52 In one study, no significant differences in SL or TST were seen in 10 primary insomnia patients compared to placebo, when given 0.3 mg or 1 mg of melatonin, or placebo 1 hour before bedtime in a double-blind cross-over design. 53 Similarly, a study on 10 patients with persistent insomnia who were randomized to 1 or 5 mg of melatonin revealed a lack of improvement in sleep SL or TST, and no effect on mood or alertness, but reported a subjective sense of improved sleep quality. 54 A much higher dose of melatonin (75 mg) improved SL, and daytime alertness compared to pla - cebo in a double-blind study of 13 insomnia patients. 55 A recent meta-analysis showed a reduction in SL by 4 minutes, increased sleep efficiency (SE) by 2.2% and increased TST by 12.8 minutes. 56 Melatonin doses as high as 75 mg have been used in clinical tri - als without significant toxicity. 55 Ramelteon is a melatonin receptor agonist, which is FDA-approved for the treatment of sleep initiation insomnia. It has a half-life of 0.8–2 hours, and T max of 0.7–0.95 hours. 57 It binds to MT1 and MT2 mela - tonin receptors within the SCN, and is me tab olized by CYP1A2, with CYP2C and CYP3A4. 58 Ramelt - eon, like melatonin, has no clear dose-response relationship; however, a therapeutic dosage of 8 mg and timing of dosing 30 minutes before bedtime has been recommended. 57 Ramelteon (16 mg or 64 mg), when given to normal adults of transient insomnia, improved SL by 10–15 min, and increased TST; how - ever, wake after sleep onset (WASO), time spent in each sleep stage and number of awakenings were not significantly different from placebo. 59 Similar studies revealed improvement in SL for primary insomnia patients with different doses of Ramelteon in adults (4 and 32 mg) and in elderly (4 and 8 mg), but no subjective improvements in sleep quality or TST. 44,60 Effects persisted even if treatment was extended to 5 weeks for both adults and elderly primary insomnia patients. 61,62 Adverse effects occur in more than 2% of patients, and the most commonly reported symp - toms are headache, somnolence, and sore throat. 60 Elevation of prolactin has been reported with Ramelt - eon compared with placebo in women. 63 There were no reports of rebound insomnia after medication discontinuation, and no significant effects of abuse potential or motor and cognitive impairment at up to 20 times the recommended therapeutic dose. 59,64 Smoking is an inducer of the CYP1A2 isozyme, decreasing ramelteon efficacy, and dose reduction is recommended when attempting smoking cessation. 65 Pharmacotherapies of insomnia Clinical Medicine Reviews in Therapeutics 2013:5 17 Agomelatine (AGO-178) is a chemical compound that is structurally related to melatonin. It is a MT1 and MT2 receptor agonist as well as a 5-HT2c antagonist. It was initially investigated as a chrono - biotic; however, with the discovery of 5-HT2c sero - toninergic receptor it is more focused on anxiolytic and antidepressant effects. The approval indication in the European Union (EU) is depression. There are currently no randomized studies with primary insom - nia patients, and most clinical studies are done in patients with major depressive disorder. Study results have shown it to positively influence disturbed cir - cadian rhythms in depressed patients by significantly improving all phases of disturbed sleep and the over - all quality of sleep, with a favorable impact on day - time alertness. 66 Tasimelteon (VEC-162) is a melatonin receptor agonist (MT1 and MT2). Tasimelteon was effective in reducing SL (in phase II and III clinical trials) and in resetting the circadian melatonin rhythm (in phase II trials), which indicated its potential sui tab ility as treat - ment for jet lag, shift work and circadian rhythm sleep disorders. 67 The drug is well tolerated without next- day functioning impairment. Further studies concern - ing efficacy and safety are still pending.

Antidepressants Antidepressants are widely used for the treatment of insomnia, and its effects on sleep are based on the findings from clinical studies in patients with mental psychiatric problems. Their mechanisms for insom - nia treatment include blocking wake-promoting neurotransmitters (acetylcholine, histamine, norepi - nephrine, serotonin, and dopamine) for sleep enhanc - ing effects. 6,57 Among them, the main sedating effects are caused by the anticholinergic and antihistamine effects. 17 The most commonly prescribed antidepres - sants are tricyclic antidepressants (doxepine, ami - triptyline, trimipramine), tetracyclic antidepressant (mirtazapine) and trazodone. Except for doxepin, most of the antidepressants for insomnia are off-label use. However, antidepressants are commonly used as hypnotics in conjunction with single medication for insomnia and psychiatric disorders, or to avoid ben - zodiazepine side effects and dependence. Serotonin reuptake inhibitors (SSRI), and serotonin and norepi - nephrine reuptake inhibitors (SNRI) block serotonin reuptake, as well as noradrenaline reuptake. This may frequently be associated with insomnia, via rapid eye movement (REM) suppression and sleep disruption. 68 Doxepin is a tricyclic antidepressant, and FDA- approved for sleep maintenance insomnia. Typical anti - depressant doses are 25–150 mg, and hypnotic doses are 3–6 mg. In the high dose range, doxepin has anti- histamine, anticholinergic, anti-serotonergic and anti- adrenergic effects, but in hypnotic doses ( ,10 mg) it has a relatively pure anti-histamine effect. 69 In a large clinical trial, adults and elderly patients with primary insomnia were prescribed 1 mg or 3 mg of doxepin, or placebo for 12 weeks of nightly use. Doxepin improved the sleep parameters including WASO, TST, and overall SE, and treatment effects were maintained until the end of the study. 70 It is remarkable to note that participants did not report significant next-day resid - ual effects. In a similar study, adults with chronic pri - mary insomnia were followed for 35 days with 3 mg and 6 mg of doxepin, which resulted in improvement in sleep maintenance, and early morning awakenings without next-day residual effects. 71 In a double-blind, placebo-controlled trial in elderly patients with pri - mary insomnia, 6 mg of doxepin produced significant improvements in sleep maintenance, sleep duration, and sleep quality, compared to placebo, and treatment effects were maintained throughout the 4 weeks of trial. 72 Rebound insomnia or withdrawal effects upon discontinuation were not seen in these trials. Amitriptyline is a tricyclic antidepressant, with a long half-life of 10–100 hours and a T max of 2–5 hours. 57 A long T max needs to be considered when choosing the dosing hour needed in order to achieve optimal time to fall asleep. 17,57 There is no data on the effects of amitriptyline in patients with primary insomnia. Open label trials without control groups indicated some improvements in sleep quality in comorbid insomnia (anxiety and depression). 73 One study compared sleep effects of amitriptyline and escitalopram to placebo in healthy subjects. Amitriptyline reduced WASO and REM, and increased sleep continuity compare to pla - cebo. However, increased periodic limb movements (PLM), and PLM arousal index, as well as increase daytime sleepiness were observed the next day. 74 The side effects are orthostatic hypotension, weight gain, dry mouth, constipation, urinary retention, and car - diac dysrhythmias. Trazodone is a triazolopyridine antidepressant that antagonizes 5-HT1, 5-HT2 and also has a weak Song and Suh 18 Clinical Medicine Reviews in Therapeutics 2013:5 serotonin reuptake inhibition effect. The half-life is 7–15 hours, and T max is 1–2 hours. 57 It was the most commonly used medication for insomnia until 2002. 75 Despite its popular use, there are limited stud - ies focused on efficacy and safety of trazodone. In primary insomnia patients, trazodone increases slow wave sleep (SWS). 76 In one study, insomnia patients who were prescribed 50 mg of trazodone were com - pared to placebo controls, and those taking trazodone displayed fewer night time awakenings, fewer min - utes of stage 1 sleep and provided self-reports cit - ing difficulty sleeping. However, trazodone was also associated significant impairment in short-term memory, verbal learning, equilibrium, and arm mus - cle endurance across time points. 77 Trazodone has lead to demonstrated improvement of both depres - sion and insomnia. 78,79 The common side effects are residual morning sedation, orthostatic hypotension, and priapism. Mirtazapine is a tetracyclic piperazinoazepine with inhibition of 5-HT2, 5-HT3 and α2-adrenergic receptors. It has half-life of 20–40 hours and a T max of 0.25–2 hours. 57 There were no randomized clinical trials of mirtazapine in primary insomnia. In healthy adults, mirtazapine has been shown to reduce SL and increase SWS and SE. 80,81 Oral doses of 5 mg, 15 mg, and 30 mg of mirtazapine, placebo, and 10 mg diazepam were given the night before sur - gery, and self-reported assessment of sleep quality improved in mirtazapine without significant side effects. 82 Additionally, sleep quality improved in a dose-dependent manner. Mirtazapine in depressed patients without clinically diagnosed insomnia showed improvement in SL and TST, but these stud - ies were not controlled by a placebo group. 83,84 There were improvements in SL, TST, SE, and WASO by PSG measures when comparing 2 weeks of mirtazap - ine to fluoxetine. 85 Additionally, depression assess - ment based on the 21-item Hamilton Rating Scale for Depression (HAM-D) demonstrated improvement in depression symptoms, largely associated with the degree of subjective sleep complaints by means of the 3 sleep items. 85 Interestingly, studies have shown that mirtazapine improved sleep quality in peri- menopausal women with hot flashes. 86 Mirtazapine in combination with melatonin for women with peri-menopausal insomnia improved sleep qual - ity. 87 Mirtazapine causes significant weight gain, which may be attributed to mirtazapine’s high anti - histaminic (H1) receptor affinity. 88,89 The mirtazapine may also cause dry mouth and constipation.

Antipsychotics Antipsychotics are used to treat insomnia, which is best indicated for patients with psychotic disorders. 5 The antipsychotics antagonize dopamine, histamine, serotonin, cholinergic and adrenergic receptors, and thus induce sleep. 57 The most commonly used antip - sychotics are quetiapine and olanzapine. 75 Quetiapine has a half-life of 7 hours, and T max of 1 hour. In healthy subjects, quetiapine significantly improved sleep ini - tiation and continuity compared to a placebo con - trol when measured by polysomnography (PSG). 90 A study in primary insomnia demonstrated quetia - pine improved PSG variables of TST and SE, and subjective sleep measures indexed using the PSQI (Pittsburgh Sleep Quality Index). 91 A typical dose of quetiapine for insomnia is less than the recommended dose for psychotic disorders, but even with low doses of quetiapine, body weight increase has been reported as a side effect. 92 Olazapine has a half-life of 30 hours and a T max of 5 hours, which makes it relatively unlikely to be effective for sleep onset difficulty when taken near bed time; however, it likely has a prolonged sleep enhancing effect. 17,57 There is a scarcity of infor - mation regarding efficacy in insomnia and proper dosage. Additionally, the safety profile of increased risk of me tab olic syndrome largely limits its use with insomnia patients. Olanzapine is primarily studied in schizophrenia, with a reported increased risk of me ta- bolic syndrome. The overall rate of me tab olic syn - drome in patients who are treated with olanzapine is 41.6% with a hazard ratio of 1.62, and the incidence is 27.4% with a hazard ratio of 1.88. 93 Olanzapine cause significant weight gain of .7% of body weight changes in 23.6% ∼61.5% of patients. Anticonvulsants Anticonvulsants are also prescribed off-label for insomnia. It is recommended and beneficial for patients who suffer from both insomnia and an under - lying epileptic disorder. Gabapentin and pregabalin are a structural analogue of GABA, but are thought to exert their primary central nervous system effects by binding to the alpha-2-delta subunit of the N-type Pharmacotherapies of insomnia Clinical Medicine Reviews in Therapeutics 2013:5 19 voltage gated calcium channels, thereby diminish - ing the release of wake-promoting neurotransmitters such as glutamate and norepinephrine. 94 Tiagabine is a GABA reuptake inhibitor and a sleep-promoting inhibitory neurotransmitter. They are FDA-approved for partial seizures but not for insomnia, and there are limited studies on effective doses for insomnia. Gabapentin in normal adults have been shown to increase SWS. 95 There is one study on the treatment effects of gabapentin in primary insomnia. A total of 18 primary insomnia patients were treated with gaba - pentin for 4 weeks, and showed increase in SWS, along with improvement in SE and spontaneous arousals. 96 Gabapentin is also effective in insomnia during alcohol abstinences, and treating sleep distur - bance in pain related disorders, epilepsy, and restless legs syndrome. 97–100 The most common side effects of gabapentin are sedation, dizziness, and ataxia. Pregabalin increases SWS in normal adults. 101 Currently, there are no studies on the effects of pregab - alin in primary insomnia. However, pregabalin has been reported to improve sleep disturbance in fibro - myalgia, anxiety and pain-related syndrome. 102–104 The most common side effects of pregabalin are seda - tion, dizziness, dry mouth, cognitive impairment and increased appetite. Tiagabine is a GABA reuptake inhibitor. In primary insomnia patients, tiagabine 4, 8, 12, and 16 mg lead to a significant increase in SWS in a dose-dependent man - ner as compared to a placebo. However, no significant changes were seen in SL and TST, and improvement in WASO was only observed in the 16 mg dose. 105 One other study with primary insomnia patients reported no improvements in SL, TST, or WASO in the tiagabine treatment group compared to placebo, but increases in SWS were seen. 106 These studies reported drowsiness and nausea as the most common side effects of tiaga - bine. Although tiagabine is FDA-approved for treat - ing partial seizure, there have been reports regarding an increased risk of de novo seizures, which limited its use to treat seizures. 107,108 Antihistamine Antihistamines are primary indicated for the treat - ment of allergies. However, there are agents with potent anti-histamine efficacy including antidepres - sants of doxepin and mirtazapine, as well as antip - sychotics including olanzapine and quetiapine. 57 Diphenhydramine and Doxylamine are the two most commonly used antihistamines for insomnia that cross the brain blood barrier. Diphenhydramine is an over-the-counter insom - nia medication. It has a half-life of 5–11 hours, and a T max of 2–2.5 hours. 17 There is 1 recent randomized placebo control study in primary insomnia patients. In mild insomnia patients, there were increases in SE and TST reported in sleep diaries compared to placebo, but no objective significance measured by PSG. 109 Similar sleep maintenance effects were seen in groups of psy - chiatric patients, as well as in outpatients in a primary care practice. 110,111 The most common side effects are sedation, dizziness, psychomotor impairment, cogni - tive impairment, dry mouth, blurred vision, constipa - tion, urinary retention, and weight gain. Doxylamine is also available as over-the-counter medication. It has a half-life of 10–12 hours, and a T max of 1.5–2.5 hours. 17 No studies examining the treatment in insomnia with doxylamine have been reported.

5-HT2A serotonin receptor inverse agonists 5-HT2A serotonin receptor inverse agonists are devel - oped as neuroleptics, which are also under clinical trials for treatment of insomnia. Inverse agonists bind to the same receptors as agonists but induce the oppo - site response to that agonist. For this to happen, the receptor must have an intrinsic basal activity level, and binding of inverse agonist blocks such activity.

Serotonin is a wake-promoting neurotransmitter.

Some atypical selective serotonin reuptake inhibi - tors have been shown to increase SWS, particularly those that bind to the serotonin 5-HT2A receptor. 112 A decrease in SWS leads to a lighter level of sleep, which may increase arousal, wakefulness, and sleep fragmentation. Currently, 5-HT2A serotonin recep - tor inverse agonists are under investigation for sleep maintenance therapy, but none have been approved for treatment of insomnia. Pimavanserin (ACP-103) is currently undergoing Phase III trials for psychosis and Phase II trials for insomnia. 113 In 45 healthy volunteers, Pimavanserin significantly increased SWS in a dose-dependent manner and decreased number of awakenings com - pared to placebo. Other PSG variables including TST, SL, and number of stage shifts did not change significantly. 114 Song and Suh 20 Clinical Medicine Reviews in Therapeutics 2013:5 Nelostanserin (APD-125) is a 5-HT2A receptor inverse agonist. In 173 adults with primary insom - nia, 10 and 40 mg of Nelostanserin were compared to placebo controls for 7-day treatment periods. 115 PSG measurement of WASO decreased in compari - son to placebo in the 10 mg dose (day 1/2 and 6/7), and the 40 mg dose (day 1/2), but not significantly by day 6/7 at the 40 mg dose. No serious adverse events were reported, and there was no significant next-day psychomotor impairment. These results were not rep - licated in a double-blinded, randomized placebo con - trolled trial in 675 patients with primary insomnia.

There were no improvements in primary (subjective number of awakenings after sleep onset) or secondary end points (WASO, TST, and SL), which lead to its discontinuation for development. 116 Eplivanserin (SR-46349) had completed 3 Phase III trials indicating reduced WASO and number of nocturnal awakenings compared to placebo, with - out residual effects or withdrawal symptoms after waking. 116 However, the pharmaceutical company withdrew its application for approval.

Orexin receptor antagonists Orexin ( = Hypocretin) is a wake-promoting neurotrans - mitter produced in the lateral hypothalamus. They promote wakefulness through orexin receptors (OX1, and OX2). A deficiency in orexin is known as narco - lepsy, characterized by symptoms of excessive daytime sleepiness, cataplexy, hypnogogic hallucination, sleep paralysis and disrupted nocturnal sleep. Compared to benzodiazepine receptor agonists, orexin receptor antag - onists lead to less confusion, amnesia, and unsteady gait and have been proposed to have lower abuse liability. 117 Orexin receptor antagonists are currently under clinical investigation for the treatment of insomnia; however, none have been approved by FDA. Suvorexant (MK-4305) is a nonselective (dual) OX1/OX2 receptor antagonist. A Phase I study in 103 volunteer subjects revealed improvement in SL, TST, and WASO measured by PSG, without next-morning residual cognitive effects. A Phase II study reported improvement in SE, and Phase III clinical trials reported improvement in SL and TST in the first month of a 12-month study comparing treatment to placebo.

During the following 2 months after 12 months of suvorexant treatment, there was worsening SL and TST after switching from suvorexant to placebo. There were no withdrawal or rebound insomnia after discontinuation. Merck recently announced that they have been accepted for standard review by the U.S.

Food and Drug administration, in November, 2012, and are now waiting for approval. Amorexant (ACT-078573) was withdrawn from clinical studies due to its side effect profile. However, studies have shown it to decrease wakefulness in a dose-dependent manner in healthy human subjects, without evidence of cataplexy. 118 In primary insom - nia, increases in SE, reductions in SL and reductions in WASO were reported. 44,116 Conclusion In a search for different options for the pharmaco - therapy of insomnia, we are now faced with an ample number of drug options that are under development.

These agents have mechanisms of action that are dif - ferent from benzodiazepine receptor agonists, which allows for a different pharmacological approach.

Further research evaluating the efficacy and safety of newly emerging medications are needed, along with research evaluating currently used medications without FDA approval, in order to support their wide - spread use. Therapeutic approaches should also con - sider non-pharmacological treatments of insomnia, such as cognitive-behavioral therapy for insomnia, as an adjunctive therapeutic option, and should consider different co-morbid conditions to tailor treatment for insomnia patients.

Author Contributions Analyzed the data: PS, SS. Wrote the first draft of the manuscript: PS. Contributed to the writing of the manuscript: PS, SS. Agree with manuscript results and conclusions: PS, SS. Jointly developed the struc - ture and arguments for the paper: PS, SS. Made critical revisions and approved final version: PS, SS. All authors reviewed and approved of the final manuscript.

Funding This study was supported by a grant from the National Research Foundation of Korea Grant funded by the Korean Government. [NRF-2012-S1A5BA01].

Competing Interests Author(s) disclose no potential conflicts of interest. Pharmacotherapies of insomnia Clinical Medicine Reviews in Therapeutics 2013:5 21 Disclosures and Ethics As a requirement of publication the authors have pro - vided signed confirmation of their compliance with ethical and legal obligations including but not lim - ited to compliance with ICMJE authorship and com - peting interests guidelines, that the article is neither under consideration for publication nor published elsewhere, of their compliance with legal and ethi - cal guidelines concerning human and animal research participants (if applicable), and that permission has been obtained for reproduction of any copyrighted material. This article was subject to blind, indepen - dent, expert peer review. The reviewers reported no competing interests. Provenance: the authors were invited to submit this paper.

References 1. Ohayon MM. Epidemiology of insomnia: What we know and what we still need to learn. Sleep Med Rev . 2002;6:97–111. 2. Ford DE, Kamerow DB. Epidemiologic study of sleep disturbances and psychiatric disorders. An opportunity for prevention? JAMA . 1989;262: 1479–84. 3. Ancoli-Israel S, Roth T. Characteristics of insomnia in the United States: Results of the 1991 National Sleep Foundation Survey. I. Sleep . 1999; 22(Suppl 2):S347–53. 4. Siebern AT, Suh S, Nowakowski S. Non-pharmacological treatment of insomnia. Neurotherapeutics . 2012;9:717–27. 5. Schutte-Rodin S, Broch L, Buysse D, Dorsey C, Sateia M. Clinical guide - line for the evaluation and management of chronic insomnia in adults. J Clin Sleep Med . 2008;4:487–504. 6. Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep and circa - dian rhythms. Nature . 2005;437:1257–63. 7. Mohler H, Fritschy JM, Rudolph U. A new benzodiazepine pharmacology. J Pharmacol Exp Ther . 2002;300:2–8. 8. Olsen RW, Sieghart W. International Union of Pharmacology. LXX. Subtypes of gamma-aminobutyric acid(A) receptors: classification on the basis of subunit composition, pharmacology, and function. Update. Pharmacol Rev . 2008;60:243–60. 9. Sieghart W, Sperk G. Subunit composition, distribution and function of GABA(A) receptor subtypes. Curr Top Med Chem . 2002;2:795–816. 10. Smith AJ, Alder L, Silk J, et al. Effect of alpha subunit on allosteric modula - tion of ion channel function in s tably expressed human recombinant gam - ma-aminobutyric acid(a) receptors determined using (36)cl ion flux. Mol Pharmacol . 2001;59:1108–18. 11. Rudolph U, Crestani F, Benke D, et al. Benzodiazepine actions mediated by specific gamma-aminobutyric acid(A) receptor subtypes. Nature . 1999;401: 796–800. 12. Rudolph U, Mohler H. Analysis of GABAA receptor function and dissection of the pharmacology of benzodiazepines and general anesthetics through mouse genetics. Annu Rev Pharmacol Toxicol . 2004;44:475–98. 13. Crestani F, Assandri R, Tauber M, Martin JR, Rudolph U. Contribution of the alpha1-GABA(A) receptor subtype to the pharmacological actions of benzodiazepine site inverse agonists. Neuropharmacology . 2002;43: 679–84. 14. van Rijnsoever C, Tauber M, Choulli MK, et al. Requirement of alpha5- GABAA receptors for the development of tolerance to the sedative action of diazepam in mice. J Neurosci . 2004;24:6785–90. 15. Nowell PD, Mazumdar S, Buysse DJ, Dew MA, Reynolds CF 3rd, Kupfer DJ. Benzodiazepines and zolpidem for chronic insomnia: a meta-analysis of treatment efficacy. JAMA . 1997;278:2170–7. 16. Holbrook AM, Crowther R, Lotter A, Cheng C, King D. Meta-analysis of ben - zodiazepine use in the treatment of insomnia. CMAJ . 2000;162:225–33. 17. Kryger M, Roth T, Dement WC. Principles and Practice of Sleep Medicine , 5th ed. 2011; Philidelphia: Elsevier. 18. Greenblatt DJ, Legangneux E, Harmatz JS, et al. Dynamics and kinetics of a modified-release formulation of zolpidem: Comparison with immediate- release standard zolpidem and placebo. J Clin Pharmacol . 2006;46:1469–80. 19. Scharf MB, Roth T, Vogel GW, Walsh JK. A multicenter, placebo-controlled study evaluating zolpidem in the treatment of chronic insomnia. J Clin Psychiatry . 1994;55:192–9. 20. Roehrs TA, Randall S, Harris E, Maan R, Roth T. Twelve months of nightly zolpidem does not lead to dose escalation: A prospective placebo-controlled study. Sleep . 2011;34:207–12. 21. Morgenthaler TI, Silber MH. Amnestic sleep-related eating disorder associ - ated with zolpidem. Sleep Med . 2002;3:323–7. 22. Mendelson WB. Sleepwalking associated with zolpidem. J Clin Psychopharmacol . 1994;14:150. 23. Canaday BR. Amnesia possibly associated with zolpidem administration. Pharmacotherapy . 1996;16:687–9. 24. Gilbert DL, Staats PS. Seizure after withdrawal from supratherapeutic doses of zolpidem tartrate, a selective omega I benzodiazepine receptor agonist. J Pain Symptom Manage . 1997;14:118–20. 25. Cubala WJ, Landowski J. Seizure following sudden zolpidem withdrawal. Prog Neuropsychopharmacol Biol Psychiatry . 2007;31:539–40. 26. Sethi PK, Khandelwal DC. Zolpidem at supratherapeutic doses can cause drug abuse, dependence and withdrawal seizure. J Assoc Physicians India. 2005;53:139–40. 27. Aragona M. Abuse, dependence, and epileptic seizures after zolpidem with - drawal: Review and case report. Clin Neuropharmacol . 2000;23:281–3. 28. Weinling E, McDougall S, Andre F, Bianchetti G, Dubruc C. Pharmacokinetic profile of a new modified release formulation of zolpidem designed to improve sleep maintenance. Fundam Clin Pharmacol . 2006;20:397–403. 29. Krystal AD, Erman M, Zammit GK, Soubrane C, Roth T. Long-term effi - cacy 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 . 2008;31:79–90. 30. Truven Health Analytics. Micromedex 2.0. Available at: http://www.thom sonhc.com /. Accessed Apr 22, 2013. 31. Weinling E, McDougall S, Andre F, Bianchetti G, Dubruc C. Pharmacokinetic profile of a new modified release formulation of zolpidem designed to improve sleep maintenance. Fundam Clin Pharmacol. 2006;20:397–403. 32. Staner L, Eriksson M, Cornette F, et al. Sublingual zolpidem is more effec - tive than oral zolpidem in initiating early onset of sleep in the post-nap model of transient insomnia: A polysomnographic study. Sleep Med . 2009; 10:616–20. 33. Elie R, Ruther E, Farr I, Emilien G, Salinas E. Sleep latency is shortened during 4 weeks of treatment with zaleplon, a novel nonbenzodiazepine hyp - notic. Zaleplon Clinical Study Group. J Clin Psychiatry . 1999;60:536–44. 34. Walsh JK, Pollak CP, Scharf MB, Schweitzer PK, Vogel GW. Lack of resid - ual sedation following middle-of-the-night zaleplon administration in sleep maintenance insomnia. Clin Neuropharmacol . 2000;23:17–21. 35. Zammit GK, Corser B, Doghramji K, et al. Sleep and residual sedation after administration of zaleplon, zolpidem, and placebo during experimental middle-of-the-night awakening. J Clin Sleep Med . 2006;2:417–23. 36. Fry J, Scharf M, Mangano R, Fujimori M. Zaleplon improves sleep without producing rebound effects in outpatients with insomnia. Zaleplon Clinical Study Group. Int Clin Psychopharmacol . 2000;15:141–52. 37. Walsh JK, Vogel GW, Scharf M, et al. A five week, polysomnographic assessment of zaleplon 10 mg for the treatment of primary insomnia. Sleep Med . 2000;1:41–9. 38. Nutt DJ, Stahl SM. Searching for perfect sleep: the continuing evolution of gabaa receptor modulators as hypnotics. J Psychopharmacol . 2010;24: 1601–12. 39. Drover DR. Comparative pharmacokinetics and pharmacodynamics of short-acting hypnosedatives: zaleplon, zolpidem and zopiclone. Clin Pharmacokinet . 2004;43:227–38. Song and Suh 22 Clinical Medicine Reviews in Therapeutics 2013:5 40. Krystal AD, Walsh JK, Laska E, et al. Sustained efficacy of eszopiclone over 6 months of nightly treatment: Results of a randomized, double-blind, placebo-controlled study in adults with chronic insomnia. Sleep . 2003;26: 793–9. 41. Walsh JK, Krystal AD, Amato DA, et al. Nightly treatment of primary insomnia with eszopiclone for 6 months: Effect on sleep, quality of life, and work limitations. Sleep . 2007;30:959–68. 42. McCall WV, Erman M, Krystal AD, et al. A polysomnography study of eszopiclone in elderly patients with insomnia. Curr Med Res Opin . 2006;22: 1633–42. 43. Roth T, Roehrs TA. A review of the safety profiles of benzodiazepine hypnotics. J Clin Psychiatry . 1991;52(Suppl):38–41. 44. Ioachimescu OC, El-Solh AA. Pharmacotherapy of insomnia. Expert Opin Pharmacother . 2012;13:1243–60. 45. Roth T, Walsh JK, Krystal A, Wessel T, Roehrs TA. An evaluation of the efficacy and safety of eszopiclone over 12 months in patients with chronic primary insomnia. Sleep Med . 2005;6:487–95. 46. Soumerai SB, Simoni-Wastila L, Singer C, et al. Lack of relationship between long-term use of benzodiazepines and escalation to high dosages. Psychiatr Serv . 2003;54:1006–11. 47. Roehrs T, Vogel G, Roth T. Rebound insomnia: Its determinants and significance. Am J Med . 1990;88:39S–42. 48. Hajak G, Muller WE, Wittchen HU, Pittrow D, Kirch W. Abuse and dependence potential for the non-benzodiazepine hypnotics zolpidem and zopiclone: A review of case reports and epidemiological data. Addiction . 2003;98:1371–8. 49. Liu C, Weaver DR, Jin X, et al. Molecular dissection of two distinct actions of melatonin on the suprachiasmatic circadian clock. Neuron . 1997;19: 91–102. 50. Nosjean O, Nicolas JP, Klupsch F, Delagrange P, Canet E, Boutin JA. Comparative pharmacological studies of melatonin receptors: MT1, MT2 and MT3/QR2. Tissue distribution of MT3/QR2. Biochem Pharmacol . 2001;61:1369–79. 51. DeMuro RL, Nafziger AN, Blask DE, Menhinick AM, Bertino JS Jr. The absolute bioavailability of oral melatonin. J Clin Pharmacol . 2000;40: 781–4. 52. Sack RL, Hughes RJ, Edgar DM, Lewy AJ. Sleep-promoting effects of melatonin: At what dose, in whom, under what conditions, and by what mechanisms? Sleep . 1997;20:908–15. 53. Almeida Montes LG, Ontiveros Uribe MP, Cortes Sotres J, Heinze Martin G. Treatment of primary insomnia with melatonin: A double-blind, placebo-controlled, crossover study. J Psychiatry Neurosci . 2003;28:191–6. 54. James SP, Sack DA, Rosenthal NE, Mendelson WB. Melatonin administra - tion in insomnia. Neuropsychopharmacology . 1990;3:19–23. 55. MacFarlane JG, Cleghorn JM, Brown GM, Streiner DL. The effects of exog - enous melatonin on the total sleep time and daytime alertness of chronic insomniacs: a preliminary study. Biol Psychiatry . 1991;30:371–6. 56. Brzezinski A, Vangel MG, Wurtman RJ, et al. Effects of exogenous mela - tonin on sleep: A meta-analysis. Sleep Med Rev . 2005;9:41–50. 57. Krystal AD. A compendium of placebo-controlled trials of the risks/ benefits of pharmacological treatments for insomnia: the empirical basis for US Clinical practice. Sleep Med Rev . 2009;13:265–74. 58. Greenblatt DJ, Harmatz JS, Karim A. Age and gender effects on the pharma - cokinetics and pharmacodynamics of ramelteon, a hypnotic agent acting via melatonin receptors MT1 and MT2. J Clin Pharmacol . 2007;47:485–96. 59. Roth T, Stubbs C, Walsh JK. Ramelteon (TAK-375), a selective MT1/MT2- receptor agonist, reduces latency to persistent sleep in a model of transient insomnia related to a novel sleep environment. Sleep . 2005;28:303–7. 60. Erman M, Seiden D, Zammit G, Sainati S, Zhang J. An efficacy, safety, and dose-response study of ramelteon in patients with chronic primary insomnia. Sleep Med . 2006;7:17–24. 61. Roth T, Seiden D, Sainati S, Wang-Weigand S, Zhang J, Zee P. Effects of ramelteon on patient-reported sleep latency in older adults with chronic insomnia. Sleep Med . 2006;7:312–8. 62. Zammit G, Roth T, Erman M, Sainati S, Weigand S, Zhang J. Double blind, placebo-controlled polysomnography and outpatient trial to evaluate the efficacy and safety of ramelteon in adult patients with chronic insomnia. Sleep . 2005;28:A228–9. 63. Richardson G, Wang-Weigand S. Effects of long-term exposure to ramelt - eon, a melatonin receptor agonist, on endocrine function in adults with chronic insomnia. Hum Psychopharmacol . 2009;24:103–11. 64. Johnson MW, Suess PE, Griffiths RR. Ramelteon: a novel hypnotic lacking abuse liability and sedative adverse effects. Arch Gen Psychiatry . 2006;63: 1149–57. 65. Schaffer SD, Yoon S, Zadezensky I. A review of smoking cessation: potentially risky effects on prescribed medications. J Clin Nurs . 2009;18: 1533–40. 66. Demyttenaere K. Agomelatine: a narrative review. Eur Neuropsychophar - macol . 2011;21(Suppl 4):S703–9. 67. Hardeland R. Tasimelteon, a melatonin agonist for the treatment of insomnia and circadian rhythm sleep disorders. Curr Opin Investig Drugs . 2009;10: 691–701. 68. Mayers AG, Baldwin DS. Antidepressants and their effect on sleep. Hum Psychopharmacol . 2005;20:533–59. 69. Weber J, Siddiqui MA, Wagstaff AJ, McCormack PL. Low-dose doxepin: In the treatment of insomnia. CNS Drugs . 2010;24:713–20. 70. Krystal AD, Durrence HH, Scharf M, et al. 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 . 2010;33:1553–61. 71. Krystal AD, Lankford A, Durrence HH, et al. Efficacy and safety of doxepin 3 and 6 mg in a 35-day sleep laboratory trial in adults with chronic primary insomnia. Sleep . 2011;34:1433–42. 72. Lankford A, Rogowski R, Essink B, Ludington E, Heith Durrence H, Roth T. Efficacy and safety of doxepin 6 mg in a four-week outpatient trial of elderly adults with chronic primary insomnia. Sleep Med . 2012;13:133–8. 73. Feren S, Katyal A, Walsh JK. Efficacy of hypnotic medications and other medications used for insomnia. 2006;1:387–97. 74. Doerr JP, Spiegelhalder K, Petzold F, et al. Impact of escitalopram on nocturnal sleep, day-time sleepiness and performance compared to amitriptyline: A randomized, double-blind, placebo-controlled study in healthy male subjects. Pharmacopsychiatry . 2010;43:166–73. 75. Walsh JK. Drugs used to treat insomnia in 2002: Regulatory-based rather than evidence-based medicine. Sleep . 2004;27:1441–2. 76. Montgomery I, Oswald I, Morgan K, Adam K. Trazodone enhances sleep in subjective quality but not in objective duration. Br J Clin Pharmacol . 1983;16:139–44. 77. Roth AJ, McCall WV, Liguori A. Cognitive, psychomotor and polysomno - graphic effects of trazodone in primary insomniacs. J Sleep Res . 2011;20: 552–8. 78. Wichniak A, Wierzbicka A, Jernajczyk W. Patients with insomnia and subthreshold depression show marked worsening of insomnia after discon - tinuation of sleep promoting medication. Prog Neuropsychopharmacol Biol Psychiatry . 2011;35:1671–6. 79. Sheehan DV, Croft HA, Gossen ER, et al. Extended-release trazodone in major depressive disorder: A randomized, double-blind, placebo-controlled study. Psychiatry (Edgmont) . 2009;6:20–33. 80. Aslan S, Isik E, Cosar B. The effects of mirtazapine on sleep: a placebo controlled, double-blind study in young healthy volunteers. Sleep . 2002;25: 677–9. 81. McCall C, McCall WV. What is the role of sedating antidepressants, antipsychotics, and anticonvulsants in the management of insomnia? Curr Psychiatry Rep . 2012;14:494–502. 82. Sorensen M, Jorgensen J, Viby-Mogensen J, Bettum V, Dunbar GC, Steffensen K. A double-blind group comparative study using the new anti-depressant org 3770, placebo and diazepam in patients with expected insomnia and anxiety before elective gynaecological surgery. Acta Psychi - atr Scand . 1985;71:339–46. 83. Winokur A, Sateia MJ, Hayes JB, Bayles-Dazet W, MacDonald MM, Gary KA. Acute effects of mirtazapine on sleep continuity and sleep architecture in depressed patients: A pilot study. Biol Psychiatry . 2000;48: 75–8. 84. Radhakishun FS, van den Bos J, van der Heijden BC, Roes KC, O’Hanlon JF. Mirtazapine effects on alertness and sleep in patients as recorded by interac - tive telecommunication during treatment with different dosing regimens. J Clin Psychopharmacol . 2000;20:531–7. Pharmacotherapies of insomnia Clinical Medicine Reviews in Therapeutics 2013:5 23 85. Winokur A, DeMartinis NA 3rd, McNally DP, Gary EM, Cormier JL, Gary KA. Comparative effects of mirtazapine and fluoxetine on sleep physiology measures in patients with major depression and insomnia. J Clin Psychiatry . 2003;64:1224–9. 86. Perez DG, Loprinzi CL, Barton DL, Pockaj BA, Sloan J, Novotny PJ, et al. Pilot evaluation of mirtazapine for the treatment of hot flashes. J Support Oncol . 2004;2:50–6. 87. Dolev Z. Case series of perimenopausal women with insomnia treated with mirtazapine followed by prolonged-release melatonin add-on and monotherapy. Arch Womens Ment Health . 2011;14:269–73. 88. Fawcett J, Barkin RL. Review of the results from clinical studies on the efficacy, safety and tolerability of mirtazapine for the treatment of patients with major depression. J Affect Disord . 1998;51:267–85. 89. Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev . 2001;7:249–64. 90. Cohrs S, Rodenbeck A, Guan Z, et al. Sleep-promoting properties of que - tiapine in healthy subjects. Psychopharmacology (Berl) . 2004;174:421–9. 91. Wiegand MH, Landry F, Bruckner T, Pohl C, Vesely Z, Jahn T. Quetiapine in primary insomnia: A pilot study. Psychopharmacology (Berl) . 2008;196: 337–8. 92. Coe HV, Hong IS. Safety of low doses of quetiapine when used for insomnia. Ann Pharmacother . 2012;46:718–22. 93. L’Italien GJ, Casey DE, Kan HJ, Carson WH, Marcus RN. Comparison of metabolic syndrome incidence among schizophrenia patients treated with aripiprazole versus olanzapine or placebo. J Clin Psychiatry . 2007;68: 1510–6. 94. Sills GJ. The mechanisms of action of gabapentin and pregabalin. Curr Opin Pharmacol . 2006;6:108–13. 95. Foldvary-Schaefer N, De Leon Sanchez I, Karafa M, Mascha E, Dinner D, Morris HH. Gabapentin increases slow-wave sleep in normal adults. Epilepsia . 2002;43:1493–7. 96. Lo HS, Yang CM, Lo HG, Lee CY, Ting H, Tzang BS. Treatment effects of gabapentin for primary insomnia. Clin Neuropharmacol . 2010;33:84–90. 97. Furieri FA, Nakamura-Palacios EM. Gabapentin reduces alcohol con - sumption and craving: A randomized, double-blind, placebo-controlled trial. J Clin Psychiatry . 2007;68:1691–700. 98. Arnold LM, Goldenberg DL, Stanford SB, et al. Gabapentin in the treat - ment of fibromyalgia: A randomized, double-blind, placebo-controlled, multicenter trial. Arthritis Rheum . 2007;56:1336–44. 99. Garcia-Borreguero D, Larrosa O, de la Llave Y, Verger K, Masramon X, Hernandez G. Treatment of restless legs syndrome with gabapentin: A double-blind, cross-over study. Neurology . 2002;59:1573–9. 100. de Haas S, Otte A, de Weerd A, van Erp G, Cohen A, van Gerven J. Exploratory polysomnographic evaluation of pregabalin on sleep distur - bance in patients with epilepsy. J Clin Sleep Med . 2007;3:473–8. 101. Hindmarch I, Dawson J, Stanley N. A double-blind study in healthy volun - teers to assess the effects on sleep of pregabalin compared with alprazolam and placebo. Sleep . 2005;28:187–93. 102. Russell IJ, Crofford LJ, Leon T, et al. The effects of pregabalin on sleep disturbance symptoms among individuals with fibromyalgia syndrome. Sleep Med . 2009;10:604–10. 103. Pauer L, Winkelmann A, Arsenault P, et al. An international, random - ized, double-blind, placebo-controlled, phase iii trial of pregabalin mono - therapy in treatment of patients with fibromyalgia. J Rheumatol . 2011;38: 2643–52. 104. Montgomery SA, Herman BK, Schweizer E, Mandel FS. The efficacy of pregabalin and benzodiazepines in generalized anxiety disorder present - ing with high levels of insomnia. Int Clin Psychopharmacol . 2009;24 214–22. 105. Walsh JK, Zammit G, Schweitzer PK, Ondrasik J, Roth T. Tiagabine enhances slow wave sleep and sleep maintenance in primary insomnia. Sleep Med . 2006;7:155–61. 106. Walsh JK, Perlis M, Rosenthal M, Krystal A, Jiang J, Roth T. Tiagabine increases slow-wave sleep in a dose-dependent fashion without affecting traditional efficacy measures in adults with primary insomnia. J Clin Sleep Med . 2006;2:35–41. 107. Trinka E, Moroder T, Nagler M, Staffen W, Loscher W, Ladurner G. Clinical and EEG findings in complex partial status epilepticus with tiagabine. Seizure . 1999;8:41–4. 108. Fitzek S, Hegemann S, Sauner D, Bonsch D, Fitzek C. Drug-induced non - convulsive status epilepticus with low dose of tiagabine. Epileptic Disord . 2001;3:147–50. 109. Morin CM, Koetter U, Bastien C, Ware JC, Wooten V. Valerian-hops combination and diphenhydramine for treating insomnia: A randomized placebo-controlled clinical trial. Sleep . 2005;28:1465–71. 110. Kudo Y, Kurihara M. Clinical evaluation of diphenhydramine hydrochlo - ride for the treatment of insomnia in psychiatric patients: A double-blind study. J Clin Pharmacol . 1990;30:1041–8. 111. Rickels K, Morris RJ, Newman H, Rosenfeld H, Schiller H, Weinstock R. Diphenhydramine in insomniac family practice patients: a double-blind study. J Clin Pharmacol . 1983;23:234–2. 112. Idzikowski C, Mills FJ, Glennard R. 5-hydroxytryptamine-2 antagonist increases human slow wave sleep. Brain Res . 1986;378:164–8. 113. Abbas A, Roth BL. Pimavanserin tartrate: A 5-HT2A inverse agonist with potential for treating various neuropsychiatric disorders. Expert Opin Pharmacother . 2008;9:3251–9. 114. Ancoli-Israel S, Vanover KE, Weiner DM, Davis RE, van Kammen DP. Pimavanserin tartrate, a 5-HT(2A) receptor inverse agonist, increases slow wave sleep as measured by polysomnography in healthy adult volunteers. Sleep Med . 2011;12:134–41. 115. Rosenberg R, Seiden DJ, Hull SG, et al. Apd125, a selective serotonin 5-HT(2A) receptor inverse agonist, significantly improves sleep mainte - nance in primary insomnia. Sleep . 2008;31:1663–71. 116. Sullivan SS, Guilleminault C. Emerging drugs for insomnia: new frontiers for old and novel targets. Expert Opin Emerg Drugs . 2009;14:411–22. 117. Sullivan S. Update on emerging drugs for insomnia. Expert Opin Emerg Drugs . 2012;17:295–8. 118. Roecker AJ, Coleman PJ. Orexin receptor antagonists: medicinal chemis - try and therapeutic potential. Curr Top Med Chem . 2008;8:977–87. Copyright ofClinical Medicine ReviewsinTherapeutics isthe property ofLibertas Academica Ltd.anditscontent maynotbecopied oremailed tomultiple sitesorposted toa listserv without thecopyright holder'sexpresswrittenpermission. However,usersmayprint, download, oremail articles forindividual use.