Abstract
Circadian rhythm sleep‐wake disorders (CRSWDs) are characterized by persistent or recurrent patterns of sleep disturbance related primarily to alterations of the circadian rhythm system or the misalignment between the endogenous circadian rhythm and exogenous factors that affect the timing or duration of sleep. These disorders collectively represent a significant unmet medical need, with a total prevalence in the millions, a substantial negative impact on quality of life, and a lack of studied treatments for most of these disorders. Activation of the endogenous melatonin receptors appears to play an important role in setting the circadian clock in the suprachiasmatic nucleus of the hypothalamus. Therefore, melatonin agonists, which may be able to shift and/or stabilize the circadian phase, have been identified as potential therapeutic candidates for the treatment of CRSWDs. Currently, only one melatonin receptor agonist, tasimelteon, is approved for the treatment of a CRSWD: non–24‐hour sleep‐wake disorder (or non‐24). However, three additional commercially available melatonin receptor agonists—agomelatine, prolonged‐release melatonin, and ramelteon—have been investigated for potential use for treatment of CRSWDs. Data indicate that these melatonin receptor agonists have distinct pharmacologic profiles that may help clarify their clinical use in CRSWDs. We review the pharmacokinetic and pharmacodynamic properties of these melatonin agonists and summarize their efficacy profiles when used for the treatment of CRSWDs. Further studies are needed to determine the therapeutic potential of these melatonin agonists for most CRSWDs.
Keywords: melatonin agonists, sleep drug effects, circadian rhythm, drug effects
Misalignment between the circadian system and the external environment can have a pronounced impact on physiologic functioning, overall health, and disease susceptibility, including increased risk for metabolic disturbances, coronary disease, cancer, psychiatric disorders, and other chronic diseases.1 Disruption of the sleep‐wake cycle is a salient consequence of this misalignment and represents the primary clinical feature of circadian rhythm sleep‐wake disorders (CRSWDs). These disorders collectively represent a significant unmet medical need, with a total prevalence in the millions, a substantial negative impact on quality of life, and a lack of studied treatments for most of these disorders.2, 3, 4 The ability of melatonin receptor activation to phase‐shift the circadian clock during discrete time windows5 provides a compelling rationale for the therapeutic potential of this system in the treatment of CRSWDs. Currently, four melatonin receptor agonists are commercially available, but only one, tasimelteon, is specifically approved for the treatment of a CRSWD: non–24‐hour sleep‐wake disorder (non‐24). Additional studies have assessed the use of other melatonin agonists for the treatment of various CRSWDs, although, to our knowledge, a systematic review has not been recently published. The objectives of this review are to compare the pharmacokinetic and pharmacodynamic properties of these compounds, review the existing data describing their use in CRSWDs, and examine the potential impact of their distinct pharmacologic properties on clinical efficacy in the treatment of CRSWDs.
The Circadian Timing System
The term “circadian” is derived from the Latin circa dies, which means “around a day.” Circadian rhythms are endogenously generated oscillations in behavior or physiology that occur within a period of ~24 hours. In humans, the average circadian period is ~24.2 hours, with a wide interindividual range of 23.7–25.3 hours.6 Many processes critical for overall health are strongly influenced by the circadian system including sleep‐wake rhythms.7, 8 Central to this regulation is a circadian timing system (CTS), consisting of input signals, a central “pacemaker,” and output signals that relay timing information to the “subordinate” oscillators throughout the brain and body. Because the endogenous circadian system runs slightly longer than 24 hours, the CTS must be reset by external zeitgebers, or “time givers,” on a daily basis to maintain alignment with the 24‐hour day. The light‐dark cycle is the primary environmental zeitgeber, with an ability to alter the timing of the circadian system and ensure that the multitude of downstream rhythms is aligned to the 24‐hour day.9 In mammals, specialized photoreceptors in the retina are responsible for capturing the environmental light information used by the CTS. These intrinsically photosensitive retinal ganglion cells (ipRGCs) are distinct from the rods and cones responsible for vision, and they are the principal photoreceptors of the circadian system.10 The ipRGCs convey light information directly to the central pacemaker located in the suprachiasmatic nucleus (SCN) of the anterior hypothalamus.11 In turn, the SCN relays this information via output signals that maintain harmony among the numerous physiologic processes that are locally organized at the level of individual tissues.12
The timing of the sleep‐wake cycle, which is appropriately synchronized to nighttime in most individuals,13 is regulated by the CTS. The SCN promotes the timing of sleep by sending direct and indirect projections to brain regions involved with sleep and arousal.14 As a result, circadian rhythms in alertness, drowsiness, mood, and other behaviors (process C) interact with the homeostatic sleep pressure (process S) that builds as a function of time spent awake.15 Ideally, these two processes are coordinated so that the human body is primed for activity that coincides with light onset and is maintained throughout the day and evening, whereas sleep pressure and reduced circadian arousal are synchronized to facilitate nighttime sleep onset and maintenance that ultimately facilitates repair and restoration during the night phase.16 As discussed in the next section, circadian disruption or environmental misalignment can compromise the timing of the sleep‐wake cycle, which forms the clinical basis of all CRSWDs.
CRSWD Classification
The International Classification of Sleep Disorders, Third Edition, defines CRSWDs as persistent or recurrent patterns of sleep disturbance due primarily to one of the following: alterations of the circadian rhythm system or misalignment between the endogenous circadian rhythm and exogenous factors that affect the timing or duration of sleep.17 The circadian‐related sleep disruption leads to insomnia, excessive daytime sleepiness, or both. These sleep disturbances are associated with impairment of social, occupational, and/or other areas of functioning.17 CRSWDs can be broadly divided into exogenous and endogenous subgroups. Exogenous CRSWDs include jet lag disorder and shift work disorder that result from externally mediated (i.e., behavioral) disruptions to an otherwise properly functioning circadian system.3 Conversely, endogenous CRSWDs involve abnormalities within the circadian system itself that impair proper alignment to the external environment. Whereas endogenous disorders typically involve genetic or biological disruption to the system, behavioral or environmental factors, such as maladaptive exposure to light cues, may contribute to the development of these disorders.2 This subgroup includes advanced sleep‐wake phase disorder (ASWPD), delayed sleep‐wake phase disorder (DSWPD), non‐24, and irregular sleep‐wake rhythm disorder (ISWRD).17 Table 1 provides a brief summary of CRSWDs.17 More extensive descriptions of the etiology, symptoms, and prevalence of these disorders are beyond the scope of this review but are available elsewhere.2, 3, 18, 19
Table 1.
Disorder | Brief description |
---|---|
Exogenous | |
Jet lag disorder | Endogenous sleep‐wake cycle is temporarily misaligned with the customary destination sleep‐and‐wake pattern following rapid travel across multiple time zones |
Shift work disorder | Symptoms of insomnia or excessive sleepiness that occur in association with work hours that overlap with the typical sleep period |
Endogenous | |
Advanced sleep‐wake phase disorder | Sleep‐and‐wake timing is advanced (i.e., earlier), usually by ≥ 2 hr, than required or desired times |
Delayed sleep‐wake phase disorder | Sleep‐and‐wake timing is delayed, usually by ≥ 2 hr, relative to what is typically considered normal |
Irregular sleep‐wake rhythm disorder | No clearly defined circadian rhythm; sleep‐wake pattern varies from day to day |
Non–24‐hr sleep‐wake disorder | The intrinsic circadian pacemaker is not entrained to a 24‐hr light‐dark cycle, and the endogenous sleep‐wake timing oscillates in and out of phase with the typical 24‐hr sleep‐wake pattern |
Treatment of CRSWDs
Nonpharmacologic options for the treatment of CRSWDs include light therapy and behavioral strategies that may be used alone or in conjunction with pharmacotherapy. Review articles covering these topics are available elsewhere.19, 20 In this review, we evaluate the therapeutic potential of pharmacologic treatment options, specifically melatonin receptor agonists, for CRSWDs.
The Endogenous Melatonin System
Targeting the melatonin receptors for the purpose of shifting and stabilizing the circadian phase represents a compelling pharmacologic approach for the treatment of CRSWDs. Melatonin secretion by the pineal gland is considered to be a reliable and predictable marker of the central pacemaker's circadian phase,21 which is controlled through a multisynaptic pathway originating from the SCN.22 In healthy individuals, the duration of melatonin secretion is a faithful representation of the period of darkness23 and is thought to influence the circadian clock reciprocally through feedback to melatonin receptors in the SCN.24 Although the exact role of endogenous melatonin in humans is unclear,25 pharmacologic administration of supraphysiologic doses of melatonin during discrete time windows has been shown to phase‐shift the circadian clock and potentially promote sleep.26 These effects are mediated through two melatonin receptors, MT1 and MT2, which are expressed in the SCN of mammalian species including humans.27 Although the exact role of each receptor is an area of ongoing research, preclinical studies suggest that the MT2 receptor is critical for phase‐shifting the clock,28 whereas MT1 activation inhibits SCN activity,29 potentially affecting the downstream sleep circuits.30 Activation of MT1 versus MT2 results in unique, G‐protein–coupled second‐messenger cascades that provide a molecular basis for the different functions of these two receptors.31, 32
Dietary Supplements Containing Melatonin
Melatonin, marketed as a dietary food supplement in the United States, is available in multiple formulations and doses. Because dietary supplements containing melatonin are a heterogeneous group of products supported by limited efficacy and safety data, drawing conclusions about the use of melatonin to treat conditions or disorders is challenging. As a dietary supplement, it has not been evaluated or approved by the U.S. Food and Drug Administration (FDA) to prevent or treat any disease or condition. Based on their review of the available literature, the American Academy of Sleep Medicine (AASM) clinical guidelines have determined an array of recommendations for the use of melatonin in CRSWDs.33 They specifically indicate “weak” for use (vs no therapy) of dietary melatonin for DSWPD, non‐24 in blind adults, and children with ISWRD. In addition, they list “no recommendation” for ASWPD, non‐24 in sighted patients, and “weak against” for elderly patients with ISWRD and dementia. A previous version of these guidelines recommended use in shift work disorder and jet lag disorder.34 As a caveat, the AASM guidelines note concerns regarding the substantial variability in potency, purity, dissolvability, and safety between lots and brands of dietary supplements containing melatonin, with some brands determined to have no available melatonin present.35, 36, 37In addition, to our knowledge, no well‐controlled, large‐scale, long‐term clinical trials assessing clinical efficacy and safety of these dietary supplements have been published.38
Melatonin Receptor Agonists
The pharmaceutical development of melatonin receptor agonists has yielded a collection of compounds that, by virtue of the approval process, have the benefit of well‐characterized pharmacologic profiles and extensive safety data (Table 2). Two of these compounds, ramelteon and tasimelteon, are approved by the FDA and available commercially in the United States, and three compounds—agomelatine, prolonged‐release melatonin, and tasimelteon—are approved by the European Medicines Agency and are commercially available in Europe. These melatonin receptor agonist compounds have distinct pharmacokinetic and pharmacodynamic characteristics that differentially modulate the melatonin system.39, 40, 41, 42, 43 Although only tasimelteon is indicated for the treatment of a CRSWD (non‐24 in adults), all four have been investigated for potential use in the management of CRSWDs. We review the differences and commonalities of these compounds with emphasis on their respective data for the treatment of CRSWDs.
Table 2.
Agonist | Location | Indication | Dosage | Common adverse eventsa |
---|---|---|---|---|
Agomelatine (Valdoxan or Thymanax; Servier Pharmaceuticals, Neuilly‐sur‐Seine, France)40 | Europe | Treatment of major depressive episodes in adults < 75 yrs of ageb | 25 mg/day at bedtime; dose may be doubled if symptoms do not improve after 2 wks | Nausea, dizziness, headache, somnolence, insomnia, migraine, diarrhea, constipation, abdominal pain, vomiting, hyperhidrosis, back pain, fatigue, anxiety, increases in AST and ALT levelsc |
Prolonged‐release melatonin (Circadin; Neurim Pharmaceuticals, Tel Aviv, Israel)39 |
Europe South Africa South Korea |
Short‐term (daily for up to 13 wks) treatment of primary insomnia characterized by poor sleep quality in patients > 55 yrs of age | 2 mg/day, 1–2 hr before going to bed and after food | Headache, nasopharyngitis, back pain, arthralgiad |
Ramelteon (Rozerem; Takeda Pharmaceuticals, Deerfield, IL)43 |
United States Japan |
Treatment of insomnia characterized by difficulty with sleep onsete | 8 mg/day within 30 min of going to bed and without food | Somnolence, dizziness, fatigue, nausea, exacerbated insomniaf |
Tasimelteon (Hetlioz; Vanda Pharmaceuticals, Washington, DC)41, 42 |
United States Europe |
Treatment of non–24‐hr sleep‐wake disorder in adults | 20 mg/day just prior to going to bed and without food | Headache, increased ALT level, nightmares or unusual dreams, upper respiratory infection, urinary tract infectiong |
ALT = alanine aminotransferase; AST = aspartate aminotransferase.
As reported in the product prescribing information or summary of product characteristics.
Treatment may continue for at least 6 months following signs of improvement.
Incidence of ≥ 1/100 to < 1/10, not corrected for placebo.
Common according to the Medical Dictionary for Regulatory Activities definition;not corrected for placebo.
Patients should be reevaluated for comorbid diagnoses if insomnia symptoms do not remit after 7–10 days of treatment.
Incidence ≥ 3% and more common than with placebo.
Incidence ≥ 5% and at least twice as high as with placebo.
Aligning the circadian system to the external environment requires the circadian clock to be reinforced or shifted in different directions, depending on the disease subtype. Therefore, the optimal use of a melatonin receptor agonist as a therapeutic agent is not necessarily achieved by replicating the amplitude or duration of endogenous melatonin but may be better reached by phase‐shifting the SCN activity during discrete time windows of maximum effectiveness, such as dawn and dusk.44 Sensitivity of the clock to melatonin administration changes dramatically during the day, with peak sensitivities occurring just before bedtime and wake time.45 In addition, the direction of the response from melatonin receptor activation (i.e., phase advance vs phase delay) changes across the phase‐response curve, and the same melatonin receptor activation can have opposing effects on phase changes at different times of day or night.45 In both preclinical and human studies, timed pulsatile melatonin administration was more efficacious than prolonged administration for phase‐shifting the circadian clock.46, 47 Because the response of the endogenous clock is time dependent,45 the timing of administration as well as the pharmacokinetic parameters (Table 3)39, 48, 49, 50, 51, 52 of a given melatonin agonist will likely play a key role in its overall effects on the circadian system.
Table 3.
Parameter | Agomelatine51 | Prolonged‐release melatonin52 | Ramelteon49, 50 | Tasimelteon48 |
---|---|---|---|---|
Absolute bioavailability | 3.3 ± 1.1% | 15% (range 10–56%)a | 1.8% (range 0.5–12%) | 38.3% (range 26.94–54.33%) |
T1/2, mean ± SD or range, hrs | 0.9 ± 0.4 | 3.5–4.0 | 1.36 ± 0.49 | 1.3 ± 0.4 |
Tmax, range, hrs | 1.0–2.0 | 0.75–3.0 | 0.5–1.5 | 0.5–3.0 |
Metabolic pathways |
CYP1A1 CYP1A2 CYP2C9 |
CYP1A1 CYP1A2 CYPC19 (possible) |
CYP1A2 (primary) CYP2C CYP3A4 (secondary metabolism produces glucuronide conjugates) |
CYP1A2 CYP3A4 |
Protein binding | 95% | In vitro: ~60%, mainly to albumin, α1‐acid glycoprotein, and high‐density lipoprotein | 82%, mostly to serum albumin | 89.1% |
Volume of distribution (L) | 35 | Not reported | 73.6 | 56–126 |
CYP = cytochrome P450; T1/2 = half‐life; Tmax = time to reach maximum concentration;
This calculation was not derived from studies of the approved compound itself but was taken from various reports on dietary supplement melatonin.
Pharmacokinetics
As with any approved and regulated compound, consistent absolute bioavailability is considered a critical pharmacokinetic parameter for the development of melatonin agonists.53 One of the primary drawbacks of dietary supplement melatonin is its documented poor and highly variable bioavailability from person to person due to high first‐pass metabolism in the liver, poor gut absorption, or possibly a combination of both factors.54, 55 The four approved melatonin receptor agonists exhibit a wide range of absolute bioavailability parameters, with an average of 3.3% for agomelatine,51 15% for prolonged‐release melatonin,52, 55, 56 1.8% for ramelteon,49 and 38.3% for tasimelteon.48 It is notable that, in the absence of bioavailability data for the approved prolonged‐release melatonin formulation, the estimate provided for the approved product is based on immediate‐release formulations.39 The applicability of immediate‐release melatonin data to prolonged‐release melatonin is unclear, given the slower absorption rate of the prolonged‐release formulation. The absolute bioavailability information provided for agomelatine is also unclear. The estimate provided was calculated from pooled population data from a phase I clinical study comparing oral versus intravenous administration, rather than by the more conventional method of comparing area under the curve values after oral and intravenous administration in the same subject.51 Interindividual and intraindividual variability was high (on the order of 160% and 104%, respectively).51
The sensitivity and direction of the response of the circadian clock to melatonin receptor activation is time dependent.44, 45 Parameters affecting duration of exposure, including half‐life (T1/2), should be taken into consideration because treatment strategies will involve shifting the clock in different directions depending on the disorder subtype. Prolonged‐release melatonin has the longest T1/2 of the melatonin agonist compounds, with a range of 3.5–4 hours.39 The remaining melatonin agonists have T1/2 values within a comparable range (0.9–1.36 hours). The active metabolite of ramelteon, M‐II, has a mean T1/2 of 2.27 hours that is believed to contribute to the circadian response to this compound.57
Pharmacodynamics
Given the proposed functional specificity of the two melatonin receptors (MT1 and MT2),24 the precise binding characteristics of a given agonist will likely play a role in determining its therapeutic action. Whereas all of these compounds bind to both melatonin receptors, their differential binding affinities to MT1 versus MT2 are distinctive. Table 4 summarizes the pharmacodynamic characteristics of these agonists.48, 52, 58, 59
Table 4.
Parameter | Agomelatine51 | Prolonged‐release melatonin52 | Ramelteon59 | Tasimelteon48, 58 |
---|---|---|---|---|
Mechanism of action | MT1 and MT2 agonist; serotonin 5‐HT2C antagonist | MT1, MT2, and MT3 agonist | MT1, MT2, and selective MT3 agonist | MT1 and MT2 agonist |
Receptor binding affinity (Ki values [nM]) |
MT1: 0.10 MT2: 0.12 5‐HT2C: 6.15 |
MT1: 0.081 MT2: 0.383 |
MT1: 0.014 MT2: 0.112 |
MT1: 0.304 MT2: 0.0692 |
Metabolite activity | Primary metabolites: (hydroxylated and demethylated agomelatine) inactive | Principal metabolite (6‐sulphatox‐y melatonin [aMT6s]) inactive | Four metabolites (M‐II, M‐IV, M‐I, and M‐III [in order of prevalence in human serum]); MT‐II active at MT1 (10% affinity of parent) and MT2 (20% affinity of parent compound), with systemic exposure 20‐ to 100‐fold higher than parent compound and mean terminal T1/2 of 2–5 hrs | Primary metabolites: M3, M9, M11, M12, M13, and M14; low binding affinity for MT1 and MT2 (< 1/10 of the binding affinity of the parent compound); low activity at melatonin receptors (at least 13‐fold less than parent compound); mean terminal elimination T1/2 of the main metabolites ranges from 1.3 ± 0.5–3.7 ± 2.2 hrs |
T1/2 = half‐life.
Agomelatine binds to MT1 and MT2 receptors with approximately equal affinity and is distinctive in that it has additional antagonist properties at the serotonin receptor subtype (5‐HT2c).40 Ramelteon exhibits a 10‐fold greater affinity for MT1 than MT2,59 and prolonged‐release melatonin exhibits an 8‐fold greater affinity for MT1 than MT2.39 Tasimelteon is the only one of these four compounds that shows a greater affinity for MT2 than MT1, with the difference being 4‐fold.58
The primary metabolites of both agomelatine and prolonged‐release melatonin are biochemically inactive; therefore, the timing and duration of the activity of these drugs fully depend on the properties of the parent compounds. The primary metabolite of ramelteon, M‐II, is active and exhibits significantly higher systemic exposure than the parent compound.45, 47 M‐II exhibits approximately a tenth and a fifth the binding affinity of the parent molecule for the human MT1 and MT2 receptors, respectively. Although the binding affinity for M‐II is lower than that of the parent compound, its overall mean systemic exposure is 20‐ to 100‐fold higher than that for ramelteon. As a result of these properties, the active metabolites of ramelteon may be relevant at a clinical level,47 specifically when being studied in circadian rhythm disorders.57 For tasimelteon, the primary metabolites (M9, M11, M12, M13, M14, and M3, a glucuronidated metabolite) have binding affinity to the melatonin receptors that is at least 13‐fold lower than that of the parent compound.42, 48 This weak affinity suggests that such metabolites are unlikely to contribute significantly to the observed clinical effects.
These distinctive pharmacodynamic properties, coupled with the theorized function of the MT1 and MT2 receptors, are consistent with the approved clinical application for each melatonin agonist. The additional binding and antagonism of the 5‐HT2C receptor with the melatonin receptor binding of agomelatine is hypothesized to contribute to its efficacy in treating major depressive disorder.40 Prolonged‐release melatonin and ramelteon, each indicated for insomnia, show a much higher MT1 binding affinity that may contribute to the reported soporific effects of these drugs.42, 43 In addition, the increased systemic exposure associated with the active metabolite of ramelteon, MT‐II,49 theoretically provides a functional similarity to prolonged‐release melatonin in terms of exposure duration. Tasimelteon, indicated for non‐24, preferentially binds the MT2 receptor, believed to be most critical for phase‐shifting the clock.28, 60
Evidence for the Use of Melatonin Receptor Agonists in Treating CRSWDs
Melatonin agonists represent compelling pharmacologic treatments for CRSWDs. In an effort to provide a concise summary of available data describing the use of these agents for CRSWDs, we performed a systematic review of published studies.
Literature Search
The assessment of available literature for the treatment of CRSWDs with approved melatonin receptor agonists was conducted in April 2015 and again in December 2015 to include the most recent literature. We conducted a search of the PubMed database using the following keywords: [(DSPD OR delayed sleep phase disorder) OR (ASPD OR advanced sleep phase disorder) OR (ISWR OR irregular sleep‐wake rhythm disorder) OR (non‐24 OR non–24‐hour sleep‐wake rhythm disorder OR free‐running disorder) OR (jet lag OR jet lag disorder) OR (shift work OR shift work disorder) AND (treatment OR therapy OR medication) OR (melatonin agonist) OR (prolonged‐release melatonin OR Circadian) OR (ramelteon OR Rozerem) OR (tasimelteon OR Hetlioz) OR (agomelatine OR Valdoxan)].
Evidence was evaluated according to the evidence grading scale used by the Oxford system for evidence‐based medicine (Table 5).34 The scoring criteria reflect the nature of the experimental design, rather than the specific clinical outcome measures obtained.
Table 5.
Evidence level | Criteria |
---|---|
1 | High‐quality randomized, controlled trial of well‐characterized subjects or patients |
2 | Cohort study or flawed clinical trial (e.g., small sample size, blinding not specified, incompletely validated reference standardsa, possible nonrandom assignment to treatment) |
3 | Case‐control study |
4 | Case series (or poor‐quality cohort and case‐control studies) |
Reference standards: polysomnography, sleep logs, actigraphy, phase markers, validated self‐reports.
Adapted from reference 34 with permission.
These search criteria yielded a total of eight articles, ranging from a single‐patient case study to double‐blind, placebo‐controlled, parallel‐group multicenter trials. Table 6 summarizes the eight studies.57, 61, 62, 63, 64, 65, 66, 67 Most of the results (not including small case studies) examined treatment of exogenous CRSWDs including jet lag and shift work disorders.57, 61, 62, 63 These disorders are the most prevalent CRSWDs in the general population.17 The only results focused on endogenous CRSWDs were studies aimed at treating non‐24.64, 65, 66, 67 Overall, the lack of evidence for studies intended to treat any other endogenous CRSWD reflects a significant need for additional research that uses the therapeutic potential of the melatonin system.
Table 6.
Evidence levelb | Compound | Study design | Population | Treatment regimen | Key findings | |
---|---|---|---|---|---|---|
Jet lag disorder | ||||||
2 | Tasimelteon | Two randomized, double‐blind, placebo‐controlled, parallel‐group studies (1 phase II study and 1 phase III study)61 | 451 healthy volunteers with experimentally induced jet lagc: phase II study (n = 39) and phase III study (n = 412) |
Phase II study: tasimelteon 10, 20, 50, or 100 mg/day given 30 min before bedtime for 3 days Phase III study: tasimelteon 20, 50, or 100 mg/day given 30 min before bedtime for 3 days |
Phase II study:
Phase III study:
|
|
2 | Ramelteon | Randomized, double‐blind, multicenter, placebo‐controlled study57 | 75 healthy volunteers with experimentally induced jet lagc | Ramelteon 1, 2, 4, or 8 mg/day given 30 min before bedtime for 4 days following a 5‐hr phase shift |
|
|
1 | Ramelteon | Randomized, double‐blind, placebo‐controlled, parallel‐group study62 | 110 participants with a history of jet lag–induced sleep difficulty | Ramelteon 1, 4, or 8 mg/day for 5 days given 5 min before lights out, based on each participant's habitual bedtime |
|
|
Shift work disorder | ||||||
2 | Ramelteon | Placebo‐controlled, crossover study63 | 10 healthy volunteers | Ramelteon 8‐mg single dose given prior to a 2‐hr afternoon nap opportunity followed by a simulated night shift |
|
|
Non–24‐hr sleep‐wake disorder | ||||||
1 | Tasimelteon |
Randomized, double‐masked, placebo‐controlled, parallel‐group, multicenter study64
Study 1: tasimelteon 20 mg vs placebo for 26 wks Study 2: 11‐wk, open‐label run‐in period followed by 8‐wk randomized treatment period (withdrawal to placebo or continued treatment) |
84 (study 1) and 20 (study 2) totally blind patients with a confirmed diagnosis of non‐24 |
Study 1: tasimelteon 20 mg/day given 1 hr before target bedtime for up to 26 wks Study 2: tasimelteon 20 mg/day given 1 hr before target bedtime for 8 wks |
Study 1:
Study 2:
|
|
4 | Agomelatine | Case report65 | 61‐yr‐old patient with traumatic brain injury and suspected non‐24 based on sleep‐wake patterns (i.e., no subjective measures or biological assays) | Agomelatine 25 mg/day at 10 p.m. for 60 days before sleep assessment and maintained for up to 18 mo |
|
|
2 | Prolonged‐release melatonin | Randomized, double‐blind, placebo‐controlled study66: 2‐wk, placebo run‐in period; 6‐wk double‐blind treatment; 2‐wk placebo run‐out period | 13 totally blind subjects reporting periodic sleep difficulties with sleep diary evidence of a phase delay for ≥ 6 wks | Prolonged‐release melatonin 2 mg/night for 6 wks |
|
|
4 | Ramelteon | Case series67 | 2 sighted patients with non‐24 based on clinical interviews and sleep diary assessments |
Patient 1: ramelteon 8 mg/night, with triazolam 0.125 mg/night added after 25 days Patient 2: ramelteon 8 mg/night + methylcobalamin (vitamin B12) 500 μg 3 times/day with meals |
Patient 1:
Patient 2:
|
CGI‐C = Clinical Global Impression of Change.
No studies were found for advanced sleep phase disorder, delayed sleep phase disorder, or irregular sleep‐wake rhythm disorder.
Using the Oxford system for evidence‐based medicine.
Jet lag was experimentally induced with a transient 5‐hr phase advance in the light‐dark schedule designed to represent eastward travel across five time zones.
Subjective measures included sleep latency, total sleep time, number of awakenings, ease of falling asleep, sleep quality, and the restorative nature of sleep.
Exogenous CRSWD Treatment
Four studies evaluated approved melatonin receptor agonists as potential treatments for exogenous CRSWDs: three focused on jet lag disorder57, 61, 62 and one on shift work disorder.63
Of the three jet lag disorder studies, only one, a placebo‐controlled study of ramelteon 1, 4, or 8 mg, was conducted in 110 subjects with a history of jet lag–induced sleep difficulty.62 Given the chronic disturbance in these patients, this study is most representative of a clinical jet lag disorder population and represents the highest level of evidence (level 1) based on the established criteria. Compared with placebo, of the three dose levels tested, only ramelteon 1 mg was associated with a significant reduction in their primary end point, mean latency to persistent sleep.
The two additional jet lag studies used laboratory‐based acute protocols involving phase shifts in the light‐dark cycle in healthy volunteers, thus providing less reliable evidence (level 2).57, 61 The ramelteon study was conducted in 75 healthy volunteers with experimentally induced jet lag.57 The three lower doses of ramelteon tested (1, 2, and 4 mg) were associated with the primary outcome, namely shifts in the endogenous melatonin rhythm compared with placebo; however, none of the doses tested (1, 2, 4, and 8 mg) significantly affected sleep architecture parameters or subjective measures of sleep relative to placebo. The tasimelteon report included data from two randomized, double‐blind, placebo‐controlled studies conducted in 451 healthy volunteers with experimentally induced jet lag.61 The first was a phase II study of tasimelteon 10, 20, 50, or 100 mg conducted in 39 individuals, and the second was a phase III study of tasimelteon 20, 50, or 100 mg conducted in 412 individuals. In the phase II study, tasimelteon 50 and 100 mg significantly increased sleep efficiency, and all doses significantly reduced sleep latency compared with placebo. Subjects in the 20‐mg, 50‐mg, and 100‐mg treatment groups also demonstrated significantly greater total sleep time compared with placebo. The 100‐mg dose also shifted the dim light melatonin onset significantly earlier than placebo. In the phase III study, all tasimelteon doses (20, 50, and 100 mg) significantly improved sleep efficiency, total sleep time, and sleep latency relative to placebo. The 20‐mg and 50‐mg doses also significantly improved sleep maintenance (decreased wake after sleep onset) compared with placebo.
A single study assessed the effects of an approved melatonin receptor agonist (ramelteon 8 mg) in shift work disorder.63 The placebo‐controlled crossover study used a simulated night shift in 10 healthy subjects and thus provided an intermediate level of evidence (level 2). Participants received ramelteon or placebo followed by a 2‐hour nap opportunity and then a simulated night shift. Sleep efficiency and neurobehavioral tests were performed the following morning to determine if ramelteon 8 mg, taken before an afternoon nap, would improve sleep and neurobehavioral symptoms following a night shift. No statistically significant differences were seen in sleep efficiency between subjects receiving ramelteon and those receiving placebo during the nap prior to the night shift. Immediately following the nap and during the simulated night shift, subjects scored significantly lower on neurobehavioral performance assessments in the ramelteon arm.
Endogenous CRSWD Treatment
Four articles described the use of approved melatonin receptor agonists for non‐24: two were randomized, double‐blind, placebo‐controlled studies conducted in blind patients with non‐24,64, 66 one was a case report detailing use in two sighted patients with non‐24,67 and another was a case report regarding use in a single patient with traumatic brain injury.65 Diagnostic criteria for non‐24 were not consistent across studies, thus complicating interpretation.
The only publication providing the highest level of evidence (level 1) was a combined report of two clinical efficacy trials for the use of tasimelteon for non‐24.64 These randomized, double‐masked, placebo‐controlled, parallel‐group multicenter trials were completed in totally blind adults with a confirmed diagnosis of non‐24. Study 1 assessed the efficacy of tasimelteon 20 mg to entrain the circadian clock (measured by melatonin timing) and improve sleep parameters (changes in nighttime and daytime sleep) compared with placebo over a 26‐week period, and study 2 assessed the maintenance effect of tasimelteon 20 mg over 8 weeks following randomized withdrawal to placebo or continued treatment. Circadian period was estimated from rhythms in the urinary melatonin metabolite 6‐sulfatoxymelatonin (aMT6s). This metabolite is a faithful representation of circulating melatonin and therefore provides a biomarker that is a reliable proxy of circadian amplitude and phase. In study 1, tasimelteon treatment was associated with higher rates of circadian entrainment (defined by aMT6s acrophase) and improved sleep parameters (defined by significantly reduced daytime sleep and significantly increased nighttime sleep). In study 2, patients receiving placebo during the randomized withdrawal remained entrained at a significantly lower rate and had significantly decreased nighttime sleep and significantly increased daytime sleep compared with those remaining on tasimelteon.
The second randomized, double‐blind, placebo‐controlled study evaluating prolonged‐release melatonin use in an endogenous CRSWD was conducted in 13 totally blind subjects who reported periodic sleep difficulties and who had sleep diary evidence of a phase delay for at least 6 weeks.66 Because no formal diagnosis of non‐24 was required, the study provided a lower level of evidence (level 2). The study included 2 weeks of a placebo run‐in period, a 6‐week randomized (1:1) treatment period with patients given nightly prolonged‐release melatonin 2 mg or placebo, and finally a 2‐week placebo run‐out period. Outcome measures included a daily voice‐recorded sleep diary, Clinical Global Impression of Change (CGI‐C) score, WHO‐Five Well‐being Index (WHO‐5) score, and safety measures. Efficacy was defined as a change in sleep duration of more than 20 minutes. This predefined primary clinical end point was achieved; however, the improvement in the melatonin group was not statistically significantly different from that observed in the placebo group. The authors alluded to the fact that none of these end points were powered to demonstrate a statistically significant effect given the sample size of this study. Sleep latency, sleep onset and offset times, number and duration of naps, CGI‐C scores, and WHO‐5 scores were also not significantly different between groups.
The two case reports provide the lowest level of evidence (level 4). One described nightly administration of agomelatine 25 mg to a 61‐year‐old man with traumatic brain injury and non‐24.65 The authors postulated that the patient's circadian clock was damaged as a result of a large subarachnoid hemorrhage. Inspection of the patient's sleep pattern over ~1 year indicated a free‐running period of about 25 hours with no discernible pattern found for weekly or monthly analyses. The authors found a significant increase in sleep efficiency after 60 days of agomelatine 25 mg taken at 10 p.m., and a significant improvement in behavioral outcomes (as assessed by the Overt Aggression Scale Modified for Neuro‐Rehabilitation) assessed after 18 months of the nightly agomelatine dose.
The second report described the efficacy of ramelteon as part of a combination regimen for the treatment of non‐24 in two sighted patients.67 These individuals were determined to have non‐24 based on clinical interviews and sleep diary assessments that led the authors to believe a free‐running pattern had persisted for a number of months prior to their entering the clinic. Patient 1 was given nightly ramelteon 8 mg, and after 25 days was additionally given a 0.125‐mg dose of the hypnotic triazolam. The patient exhibited a free‐running sleep onset over the first 10 days of ramelteon treatment alone and then switched to a consistent sleep onset at 2 a.m. Persistent daytime sleepiness occurred throughout ramelteon monotherapy. Ramelteon and triazolam combination therapy normalized the sleep pattern by day 34. Patient 2 was given ramelteon 8 mg at night and methylcobalamin (vitamin B12) 500 μg 3 times/day with meals. The patient's sleep onset stabilized in the first day and sleep offset stabilized a few days later, which continued for the duration of his treatment. The authors listed a number of limitations for their non‐24 diagnoses in these sighted individuals, including no biomarker or actigraphy‐based analysis and a loss of the patients’ sleep diaries without providing further description. The results of these case reports should be interpreted with caution, given the diagnostic limitations of these data for patients.
Conclusion
The currently approved melatonin receptor agonists exhibit distinct pharmacologic profiles that may underlie their clinical characteristics, approved indications, and potential for use in treating CRSWDs. Effective treatment for these disorders will adjust the circadian system and align endogenous rhythms to the external environment. This requires the circadian clock to be shifted in different directions, depending on the disease subtype. Given that the sensitivity and direction of the circadian response changes based on time of day, timing of administration and duration of systemic exposure should be taken into consideration with the treatment of these disorders. When reviewing the available data for the use of melatonin receptor agonists for the treatment of CRSWDs, a relative paucity of evidence clearly remains regarding these disorders, including the most prevalent endogenous disorder, the delayed sleep phase type. Intervention for the exogenous disorders—jet lag disorder and shift work disorder—have only been assessed in a handful of cases, despite accounting for the vast majority of individuals with CRSWDs, with an estimated prevalence in the millions worldwide.46 In addition to the relatively limited number of studies for the treatment of CRSWDs, there are no approved therapies for almost all of these disorders. Accordingly, there is a tremendous opportunity for additional research that uses the therapeutic potential of the melatonin system and takes into consideration the distinctive properties of these diverse melatonergic compounds to address this unmet medical need.
Acknowledgments
The authors would like to thank their colleagues at Vanda Pharmaceuticals including Joseph Medicis, Pharm.D., FCP, FASHP; Aaron Sheppard, Ph.D.; Melissa Olivadoti, Ph.D.; and especially Paolo Baroldi, M.D., Ph.D., for their valuable insight and editorial comments on previous versions of this manuscript.
Technical editorial assistance in preparing this manuscript for publication from the authors’ original creation was provided by Synchrony Medical Communications, LLC, West Chester, Pennsylvania.
Disclosures: Wilbur Williams III and Dewey McLin III are employees of Vanda Pharmaceuticals, Inc., Washington, D.C. Marlene Dressman is a former employee of Vanda Pharmaceuticals, Inc. David Neubauer has not received financial compensation for his contribution to this manuscript. He has served in the past as a consultant for Vanda Pharmaceuticals.
Social media summary: Circadian rhythm sleep‐wake disorders (CRSWDs) substantially impact millions of lives. Although melatonin receptors represent compelling therapeutic targets, only one drug currently has a CRSWD indication. This article compares the pharmacology and clinical data of melatonin agonists for CRSWD treatment.
Funding: Funding for this support was provided by Vanda Pharmaceuticals, Inc., Washington, D.C.
References
- 1. Baron KG, Reid KJ. Circadian misalignment and health. Int Rev Psychiatry 2014;26:139–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep disorders: part II, advanced sleep phase disorder, delayed sleep phase disorder, free‐running disorder, and irregular sleep‐wake rhythm. An American Academy of Sleep Medicine review. Sleep 2007;30:1484–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Sack RL, Auckley D, Auger RR, et al. Circadian rhythm sleep disorders: part I, basic principles, shift work and jet lag disorders. An American Academy of Sleep Medicine review. Sleep 2007;30:1460–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Drake CL, Roehrs T, Richardson G, Walsh JK, Roth T. Shift work sleep disorder: prevalence and consequences beyond that of symptomatic day workers. Sleep 2004;27:1453–62. [DOI] [PubMed] [Google Scholar]
- 5. Cajochen C, Krauchi K, Wirz‐Justice A. Role of melatonin in the regulation of human circadian rhythms and sleep. J Neuroendocrinol 2003;15:432–7. [DOI] [PubMed] [Google Scholar]
- 6. Dijk DJ, Lockley SW. Integration of human sleep‐wake regulation and circadian rhythmicity. J Appl Physiol 2002;92:852–62. [DOI] [PubMed] [Google Scholar]
- 7. Saper CB, Cano G, Scammell TE. Homeostatic, circadian, and emotional regulation of sleep. J Comp Neurol 2005;493:92–8. [DOI] [PubMed] [Google Scholar]
- 8. Buijs RM, Scheer FA, Kreier F, et al. Organization of circadian functions: interaction with the body. Prog Brain Res 2006;153:341–60. [DOI] [PubMed] [Google Scholar]
- 9. Czeisler CA, Allan JS, Strogatz SH, et al. Bright light resets the human circadian pacemaker independent of the timing of the sleep‐wake cycle. Science 1986;233:667–71. [DOI] [PubMed] [Google Scholar]
- 10. Ruby NF, Brennan TJ, Xie X, et al. Role of melanopsin in circadian responses to light. Science 2002;298:2211–3. [DOI] [PubMed] [Google Scholar]
- 11. Ibata Y, Okamura H, Tanaka M, et al. Functional morphology of the suprachiasmatic nucleus. Front Neuroendocrinol 1999;20:241–68. [DOI] [PubMed] [Google Scholar]
- 12. Kriegsfeld LJ, Silver R. The regulation of neuroendocrine function: timing is everything. Horm Behav 2006;49:557–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Roenneberg T, Kumar CJ, Merrow M. The human circadian clock entrains to sun time. Curr Biol 2007;17:R44–5. [DOI] [PubMed] [Google Scholar]
- 14. Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep and circadian rhythms. Nature 2005;437:1257–63. [DOI] [PubMed] [Google Scholar]
- 15. Dijk DJ, Czeisler CA. Contribution of the circadian pacemaker and the sleep homeostat to sleep propensity, sleep structure, electroencephalographic slow waves, and sleep spindle activity in humans. J Neurosci 1995;15:3526–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Czeisler CA, Gooley JJ. Sleep and circadian rhythms in humans. Cold Spring Harb Symp Quant Biol 2007;72:579–97. [DOI] [PubMed] [Google Scholar]
- 17. American Academy of Sleep Medicine . The international classification of sleep disorders, 3rd ed. Darien, IL: American Academy of Sleep Medicine; 2014. [Google Scholar]
- 18. Barion A, Zee PC. A clinical approach to circadian rhythm sleep disorders. Sleep Med 2007;8:566–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Dodson ER, Zee PC. Therapeutics for circadian rhythm sleep disorders. Sleep Med Clin 2010;5:701–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Gooley JJ. Treatment of circadian rhythm sleep disorders with light. Ann Acad Med Singapore 2008;37:669–76. [PubMed] [Google Scholar]
- 21. Korf HW, von GC. Mice, melatonin and the circadian system. Mol Cell Endocrinol 2006;252:57–68. [DOI] [PubMed] [Google Scholar]
- 22. Kalsbeek A, Buijs RM. Output pathways of the mammalian suprachiasmatic nucleus: coding circadian time by transmitter selection and specific targeting. Cell Tissue Res 2002;309:109–18. [DOI] [PubMed] [Google Scholar]
- 23. Brzezinski A. Melatonin in humans. N Engl J Med 1997;336:186–95. [DOI] [PubMed] [Google Scholar]
- 24. Dubocovich ML, Rivera‐Bermudez MA, Gerdin MJ, Masana MI. Molecular pharmacology, regulation and function of mammalian melatonin receptors. Front Biosci 2003;8:d1093–108. [DOI] [PubMed] [Google Scholar]
- 25. Skene DJ, Arendt J. Human circadian rhythms: physiological and therapeutic relevance of light and melatonin. Ann Clin Biochem 2006;43:344–53. [DOI] [PubMed] [Google Scholar]
- 26. Wyatt JK, Dijk DJ, Ritz‐De CA, Ronda JM, Czeisler CA. Sleep‐facilitating effect of exogenous melatonin in healthy young men and women is circadian‐phase dependent. Sleep 2006;29:609–18. [DOI] [PubMed] [Google Scholar]
- 27. Reppert SM. Melatonin receptors: molecular biology of a new family of G protein‐coupled receptors. J Biol Rhythms 1997;12:528–31. [DOI] [PubMed] [Google Scholar]
- 28. Dubocovich ML, Yun K, Al‐Ghoul WM, Benloucif S, Masana MI. Selective MT2 melatonin receptor antagonists block melatonin‐mediated phase advances of circadian rhythms. FASEB J 1998;12:1211–20. [DOI] [PubMed] [Google Scholar]
- 29. 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. [DOI] [PubMed] [Google Scholar]
- 30. Dubocovich ML, Markowska M. Functional MT1 and MT2 melatonin receptors in mammals. Endocrine 2005;27:101–10. [DOI] [PubMed] [Google Scholar]
- 31. Reppert SM, Godson C, Mahle CD, Weaver DR, Slaugenhaupt SA, Gusella JF. Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc Natl Acad Sci U S A 1995;92:8734–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Brydon L, Roka F, Petit L, et al. Dual signaling of human Mel1a melatonin receptors via G(i2), G(i3), and G(q/11) proteins. Mol Endocrinol 1999;13:2025–38. [DOI] [PubMed] [Google Scholar]
- 33. Auger RR, Burgess HJ, Emens JS, Deriy LV, Thomas SM, Sharkey KM. Clinical practice guideline for the treatment of intrinsic circadian rhythm sleep‐wake disorders: advanced sleep‐wake phase disorder (ASWPD), delayed sleep‐wake phase disorder (DSWPD), non‐24‐hour sleep‐wake rhythm disorder (N24SWD), and irregular sleep‐wake rhythm disorder (ISWRD). An update for 2015. An American Academy of Sleep Medicine clinical practice guideline. J Clin Sleep Med 2015;11:1199–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Morgenthaler TI, Lee‐Chiong T, Alessi C, et al. Standards of Practice Committee of the American Academy of Sleep Medicine. Practice parameters for the clinical evaluation and treatment of circadian rhythm sleep disorders. An American Academy of Sleep Medicine report. Sleep 2007;30:1445–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Aldhous M, Franey C, Wright J, Arendt J. Plasma concentrations of melatonin in man following oral absorption of different preparations. Br J Clin Pharmacol 1985;19:517–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Williamson BL, Tomlinson AJ, Mishra PK, Gleich GJ, Naylor S. Structural characterization of contaminants found in commercial preparations of melatonin: similarities to case‐related compounds from L‐tryptophan associated with eosinophilia‐myalgia syndrome. Chem Res Toxicol 1998;11:234–40. [DOI] [PubMed] [Google Scholar]
- 37. Naylor S, Johnson KL, Williamson BL, Klarskov K, Gleich GJ. Structural characterization of contaminants in commercial preparations of melatonin by on‐line HPLC‐electrospray ionization‐tandem mass spectrometry. Adv Exp Med Biol 1999;467:769–77. [DOI] [PubMed] [Google Scholar]
- 38. Harris IM. Regulatory and ethical issues with dietary supplements. Pharmacotherapy 2000;20:1295–302. [DOI] [PubMed] [Google Scholar]
- 39. Temmler Pharma GmbH & Co. KG . Circadin 2 mg prolonged‐release tablets [package insert]. Marburg, Germany: Temmler Pharma GmbH & Co. KG; 2016. [Google Scholar]
- 40. Les Laboratoires Servier Industrie . Valdoxan (agomelatine) 25 mg film‐coated tablets [package insert]. Giddy, France: Les Laboratoires Servier Industrie; 2016. [Google Scholar]
- 41. European Medicines Agency . Hetlioz Summary of product characteristics. 2015. Available from http://ec.europa.eu/health/documents/community-register/2015/20150703132093/anx_132093_en.pdf. Accessed May 1, 2016.
- 42. Vanda Pharmaceuticals Inc . HETLIOZ (tasimelteon) capsules, for oral use [package insert]. Washington, DC: Vanda Pharmaceuticals Inc.; 2014. [Google Scholar]
- 43. Takeda Pharmaceuticals America, Inc . Rozerem (ramelteon) tablets [package insert]. Deerfield, IL: Takeda Pharmaceuticals America, Inc.; 2010. [Google Scholar]
- 44. Zaidan R, Geoffriau M, Brun J, et al. Melatonin is able to influence its secretion in humans: description of a phase‐response curve. Neuroendocrinology 1994;60:105–12. [DOI] [PubMed] [Google Scholar]
- 45. Lewy AJ, Bauer VK, Ahmed S, et al. The human phase response curve (PRC) to melatonin is about 12 hours out of phase with the PRC to light. Chronobiol Int 1998;15:71–83. [DOI] [PubMed] [Google Scholar]
- 46. Zisapel N. Circadian rhythm sleep disorders: pathophysiology and potential approaches to management. CNS Drugs 2001;15:311–28. [DOI] [PubMed] [Google Scholar]
- 47. Burgess HJ, Revell VL, Eastman CI. A three pulse phase response curve to three milligrams of melatonin in humans. J Physiol 2008;586:639–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Torres R, Dressman MA, Kramer WG, Baroldi P. Absolute bioavailability of tasimelteon. Am J Ther 2015;22:355–60. [DOI] [PubMed] [Google Scholar]
- 49. Karim A, Tolbert D, Cao C. Disposition kinetics and tolerance of escalating single doses of ramelteon, a high‐affinity MT1 and MT2 melatonin receptor agonist indicated for treatment of insomnia. J Clin Pharmacol 2006;46:140–8. [DOI] [PubMed] [Google Scholar]
- 50. Stevenson S, Bryson S, Amakye D, Hibberd M. Study to investigate the absolute bioavailability of a single oral dose of ramelteon (TAK‐375) in healthy male subjects. Clin Pharmacol Ther 2014;75:P22. [Google Scholar]
- 51. European Medicines Agency [eds]. Assessment report for for Valdoxan. Report No. EMEA/655251/2008. London, UK: European Medicines Agency; 2008. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/000915/WC500046226.pdf. Accessed May 2016. [Google Scholar]
- 52. European Medicines Agency . Assessment report for Circadin. Report No. EMEA/H/C/695. London, UK: European Medicines Agency; 2007. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Scientific_Discussion/human/000695/WC500026808.pdf. Accessed May 2016. [Google Scholar]
- 53. Turek FW, Gillette MU. Melatonin, sleep, and circadian rhythms: rationale for development of specific melatonin agonists. Sleep Med 2004;5:523–32. [DOI] [PubMed] [Google Scholar]
- 54. Waldhauser F, Waldhauser M, Lieberman HR, Deng MH, Lynch HJ, Wurtman RJ. Bioavailability of oral melatonin in humans. Neuroendocrinology 1984;39:307–13. [DOI] [PubMed] [Google Scholar]
- 55. DeMuro RL, Nafziger AN, Blask DE, Menhinick AM, Bertino JS Jr. The absolute bioavailability of oral melatonin. J Clin Pharmacol 2000;40:781–4. [DOI] [PubMed] [Google Scholar]
- 56. Di WL, Kadva A, Johnston A, Silman R. Variable bioavailability of oral melatonin. N Engl J Med 1997;336:1028–9. [DOI] [PubMed] [Google Scholar]
- 57. Richardson GS, Zee PC, Wang‐Weigand S, Rodriguez L, Peng X. Circadian phase‐shifting effects of repeated ramelteon administration in healthy adults. J Clin Sleep Med 2008;4:456–61. [PMC free article] [PubMed] [Google Scholar]
- 58. Lavedan C, Forsberg M, Gentile AJ. Tasimelteon: a selective and unique receptor binding profile. Neuropharmacology 2015;91:142–7. [DOI] [PubMed] [Google Scholar]
- 59. Pandi‐Perumal SR, Srinivasan V, Spence DW, et al. Ramelteon: a review of its therapeutic potential in sleep disorders. Adv Ther 2009;26:613–26. [DOI] [PubMed] [Google Scholar]
- 60. Hunt AE, Al‐Ghoul WM, Gillette MU, Dubocovich ML. Activation of MT(2) melatonin receptors in rat suprachiasmatic nucleus phase advances the circadian clock. Am J Physiol Cell Physiol 2001;280:C110–8. [DOI] [PubMed] [Google Scholar]
- 61. Rajaratnam SM, Polymeropoulos MH, Fisher DM, et al. Melatonin agonist tasimelteon (VEC‐162) for transient insomnia after sleep‐time shift: two randomised controlled multicentre trials. Lancet 2009;373:482–91. [DOI] [PubMed] [Google Scholar]
- 62. Zee PC, Wang‐Weigand S, Wright KP Jr, Peng X, Roth T. Effects of ramelteon on insomnia symptoms induced by rapid, eastward travel. Sleep Med 2010;11:525–33. [DOI] [PubMed] [Google Scholar]
- 63. Cohen DA, Wang W, Klerman EB, Rajaratnam SM. Ramelteon prior to a short evening nap impairs neurobehavioral performance for up to 12 hours after awakening. J Clin Sleep Med 2010;6:565–71. [PMC free article] [PubMed] [Google Scholar]
- 64. Lockley SW, Dressman MA, Licamele L, et al. Tasimelteon for non‐24‐hour sleep‐wake disorder in totally blind people (SET and RESET): two multicentre, randomised, double‐masked, placebo‐controlled phase 3 trials. Lancet 2015;386:1754–64. [DOI] [PubMed] [Google Scholar]
- 65. O'Neill B, Gardani M, Findlay G, Whyte T, Cullen T. Challenging behaviour and sleep cycle disorder following brain injury: a preliminary response to agomelatine treatment. Brain Inj 2014;28:378–81. [DOI] [PubMed] [Google Scholar]
- 66. Roth T, Nir T, Zisapel N. Prolonged release melatonin for improving sleep in totally blind subjects: a pilot placebo‐controlled multicenter trial. Nat Sci Sleep 2015;7:13–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Yanagihara M, Nakamura M, Usui A, et al. The melatonin receptor agonist is effective for free‐running type circadian rhythm sleep disorder: case report on two sighted patients. Tohoku J Exp Med 2014;234:123–8. [DOI] [PubMed] [Google Scholar]