Pharmacology of ramelteon, a selective MT1/MT2 receptor agonist: a novel therapeutic drug for sleep disorders

Masaomi Miyamoto, Masaomi Miyamoto

Abstract

An estimated one-third of the general population is affected by insomnia, and this number is increasing due to more stressful working conditions and the progressive aging of society. However, current treatment of insomnia with hypnotics, gamma-aminobutyric acid A (GABA(A)) receptor modulators, induces various side effects, including cognitive impairment, motor disturbance, dependence, tolerance, hangover, and rebound insomnia. Ramelteon (Rozerem; Takeda Pharmaceutical Company Limited, Osaka, Japan) is an orally active, highly selective melatonin MT(1)/MT(2) receptor agonist. Unlike the sedative hypnotics that target GABA(A) receptor complexes, ramelteon is a chronohypnotic that acts on the melatonin MT(1) and MT(2) receptors, which are primarily located in the suprachiasmatic nucleus, the body's "master clock." As such, ramelteon possesses the first new therapeutic mechanism of action for a prescription insomnia medication in over three decades. Ramelteon has demonstrated sleep-promoting effects in clinical trials, and coupled with its favorable safety profile and lack of abuse potential or dependence, this chronohypnotic provides an important treatment option for insomnia.

Conflict of interest statement

The authors have no conflicts of interest.

Figures

Figure 1
Figure 1
Comparison of chemical structures of melatonin and ramelteon, (S)N‐[2‐(1, 6, 7, 8‐tetrahyrdo‐2H‐indeno[4, 5‐b]furan‐8‐yl)ethyl]propionamide.
Figure 2
Figure 2
(A) Effects of ramelteon on sleep and wakefulness in freely moving cats. Each value shows the mean (with standard error) percentage of time spent in the stages of wakefulness, SWS, or REM sleep during each block of 2 h after drug administration. Eight of 14 cats were randomly used in each dose group. *P≤ 0.05, **P≤ 0.01, compared with the vehicle‐treated control (ANOVA). #P≤ 0.05, ##P≤ 0.01, compared with the vehicle‐treated control (paired t‐test with Holm's correction) [49]. (B) Effects of melatonin on sleep and wakefulness in freely moving cats. Each value shows the mean (with standard error) percentage of time spent in the stages of wakefulness, SWS, or REM sleep during each block of 2 h after drug administration. Eight of 14 cats were randomly used in each dose group. *P≤ 0.05, **P≤ 0.01, compared with the vehicle‐treated control (ANOVA). #P≤ 0.05, ##P≤ 0.01, compared with the vehicle‐treated control (paired t‐test with Holm's correction) [49].
Figure 2
Figure 2
(A) Effects of ramelteon on sleep and wakefulness in freely moving cats. Each value shows the mean (with standard error) percentage of time spent in the stages of wakefulness, SWS, or REM sleep during each block of 2 h after drug administration. Eight of 14 cats were randomly used in each dose group. *P≤ 0.05, **P≤ 0.01, compared with the vehicle‐treated control (ANOVA). #P≤ 0.05, ##P≤ 0.01, compared with the vehicle‐treated control (paired t‐test with Holm's correction) [49]. (B) Effects of melatonin on sleep and wakefulness in freely moving cats. Each value shows the mean (with standard error) percentage of time spent in the stages of wakefulness, SWS, or REM sleep during each block of 2 h after drug administration. Eight of 14 cats were randomly used in each dose group. *P≤ 0.05, **P≤ 0.01, compared with the vehicle‐treated control (ANOVA). #P≤ 0.05, ##P≤ 0.01, compared with the vehicle‐treated control (paired t‐test with Holm's correction) [49].
Figure 3
Figure 3
Effects of ramelteon on latency to sleep onset in freely moving monkeys. Each value shows the mean latency ± SE to each light sleep (LS) and SWS at doses of 0.003 (A), 0.03 (B), and 0.3 mg/kg, p.o. (C). Six monkeys were used in each group. *P ≤ 0.05, **P ≤ 0.01, compared with vehicle‐treated controls [paired t test with Holm correction, 103].
Figure 4
Figure 4
Effects of melatonin on latency to sleep onset in freely moving monkeys. Each value shows the mean latency ± SE to light sleep (LS) and SWS at doses of 0.3 (A), 1 (B), and 3 mg/kg (C). Six or seven monkeys were used in each group. **P ≤ 0.01 compared with the control treated vehicle [paired t test with Holm correction, 103].
Figure 5
Figure 5
Effects of zolpidem on latency to sleep onset in freely moving monkeys. Each value shows the mean latency ± standard error to light sleep (LS) and SWS at doses of 1 (A), 3 (B), and 10 or 30 mg/kg, p.o. (C). Four monkeys were used in each group.
Figure 6
Figure 6
Typical electroencephalographic (EEG) spectra and fast Fourier transform (FFT) analysis in freely moving monkey when treated with ramelteon (0.3 mg/kg, p.o.), melatonin (1 mg/kg, p.o.), and zolpidem (30 mg/kg, p.o.). Typical samples were selected during non‐REM sleep. EEG, electro‐oculogram (EOG) and electromyographic (EMG) activities were recorded [103].
Figure 7
Figure 7
Effects of ramelteon on reentrainment of running‐wheel activity rhythm following an 8‐hour phase advance. Running‐wheel activity in the subjective night as a percentage of total activity in 24 hours is shown with standard error. Ramelteon or vehicle was administered 5–30 min before lights out of the new light‐dark cycle for 14 days starting the day of the phase‐shift. Arrows and black bars show the phase‐shift and the vehicle or ramelteon treatment periods [36].
Figure 8
Figure 8
Effects of ramelteon, melatonin, diazepam, and triazolam on the Morris water maze task in rats. Each value shows the mean time to find the platform submerged in the water (A, B, C, D). *P≤ 0.025, compared with the respective vehicle control group [one‐tailed Williams test, 36].
Figure 9
Figure 9
Percentage of mice failing the rota‐rod performance test after administration of ramelteon (A), melatonin (B), N‐acetyl‐5‐HT (C), or diazepam (D) at the indicated doses. Twelve mice were used in each group. **P≤ 0.01, compared with the control group treated with vehicle (chi‐square test with Holm's correction) [81].
Figure 10
Figure 10
Percentage of mice failing the rota‐rod performance test after administration of diazepam alone or diazepam in combination with ramelteon (A), melatonin (B), N‐acetyl‐5‐HT (C) at the indicated doses. Twelve mice were used in each group. *P≤ 0.05 **P≤ 0.01, compared with the control group treated with vehicle (chi‐square test with Holm's correction) [53].
Figure 11
Figure 11
Effect of ramelteon on place preference in rats. Increase in time spent in the compound‐associated compartment in the conditioned place‐preference tests in rats in the experiment 1 (A and C) and experiment 2 (B). The numbers of rats used are shown in parentheses. Vehicle groups in panels A and B are identical. *P≤ 0.05, compared with the value at the preconditioning phase (paired t‐test) [79].
Figure 12
Figure 12
Metabolic pathway of ramelteon in humans. Ramelteon is extensively metabolized, primarily by carboxylation and stereoselective hydroxylation. The major metabolite of ramelteon in serum is the monohydroxylated metabolite, M‐II.
Figure 13
Figure 13
Effects of ramelteon on sleep latency in patients with chronic insomnia under placebo‐controlled, double‐blind, randomized, 5‐period crossover study. All data are shown as the least square means. ***P≤ 0.001, compared with placebo [78].
Figure 14
Figure 14
Latency to persistent sleep and total sleep time, as measured by PSG, with ramelteon 8 mg, ramelteon 16 mg, and placebo at weeks 1, 3, and 5 (A and B). Subjective sleep latency and total sleep time, as measured by the post‐sleep questionnaire (C and D). For comparisons between ramelteon dose and placebo, *P≤ 0.05, **P≤ 0.01, and ***P≤ 0.001 [68].
Figure 15
Figure 15
Effects of ramelteon on patient‐reported sleep latency in older adults with chronic insomnia. Patients include older adults (≥65 years; n = 829) with chronic insomnia. Placebo, ramelteon 4 mg, or ramelteon 8 mg was taken nightly for 5 weeks, and patient‐reported sleep data were the collected sleep diaries. *P≤ 0.05, **P≤ 0.01, compared with the placebo control [87].

References

    1. Silber MH. Chronic insomnia. N Engl J Med 2005;353:803–810.
    1. Lamberg L. Several sleep disorders reflect gender differences. Psychiatr News 2007;42:40.
    1. Cohn MA. Hypnotics and the control of breathing: review. Br J Pharmacol 1983;16:245S–250S.
    1. Mitler MM. Nonselective and selective benzodiazepine receptor agonists—where are we today? Sleep 2000;23:S39–S47.
    1. Roache JD, Griffiths RR. Comparison of triazolam and pentobarbital: performance impairment, subjective effects and abuse ability. J Pharmacol Exp Ther 1985;234:120–133.
    1. Barbera J, Shapio C. Benefit‐risk assessment of zaleplon in the treatment of insomnia. Drug Saf 2005;28:301–308.
    1. Griffiths RR, Sannerud CA, Ator NA, Brady JV. Zolpidem behavioral pharmacology in baboons: Self‐injection, discrimination, tolerance and withdrawal. J Pharmacol Exp Ther 1992;260:1199–1208.
    1. Liappas IA, Malitas PN, Dimopoulos NP, Gitsa OE, Liappas AI, Nikolaou CK, Christodoulou GN. Zolpidem dependence case series: Possible neurobiological mechanisms and clinical management. J Psychopharmacol 2003;17:131–135.
    1. Preston GC, Ward CE, Broks P, Traub M, Stahl SM. Effects of lorazepam on memory, attention and sedation in man: Antagonism by Ro 15–1788. Psychopharmacology 1989;97:222–227.
    1. Sanger DJ, Benavides J, Perrault G, Morel E, Cohen C, Joly D, Zivkovic B. Recent developments in the behavioral pharmacology of benzodiazepine (omega) receptors: Evidence for the functional significance of receptor subtypes. Neurosci Biobehav Rev 1994;18:355–372.
    1. 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;15:141–152.
    1. Terzano MG, Rossi M, Palomba V, Smerieri A, Parrino L. New drugs for insomnia: Comparative tolerability of zopiclone, zolpidem and zaleplon. Drug Saf 2003;26:261–282.
    1. Troy SM, Lucki I, Unruh MA, Cevallos WH, Leister CA, Martin PT, Furlan PM, Mangano R. Comparison of the effects of zaleplon, zolpidem, and triazolam on memory, learning, and psychomotor performance. J Clin Psychopharmacol 2000;20:328–337.
    1. Roth T, Roehrs T, Vogel G. Zolpidem in the treatment of transient insomnia: A double‐blind, randomized comparison with placebo. Sleep 1995;18:246–251.
    1. Krystal AD, Wash JK, Laska E, Caron J, Amato DA, Wessel TC, Roth T. 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;26:793–799.
    1. Roth T, Soubrane C, Titeux L, Wash JK. Efficacy and safety of zolpidem‐MR: a double‐blind, placebo‐controlled study in adults with primary insomnia. Sleep Med 2006;7:397–406.
    1. Farber RH, Burke PJ. Post‐bedtime dosing with indiplon in adults and the elderly: Results from two placebo‐controlled, active comparator crossover studies in healthy volunteers. Curr Med Res Opin 2008;24:837–846.
    1. Rush CR, Frey JM, Griffith RR. Zaleplon and triazolam in humans: Acute behavioral effects and abuse potential. Psychopharmacology (Berl) 1999;145:39–51.
    1. Vermeeren A. Residual effects of hypnotics: Epidemiology and clinical implications. CNS Drugs 2004;18:297–328.
    1. Wagner J, Wagner ML. Non‐benzodiazepines for the treatment of insomnia. Sleep Med Rev 2000;4:551–581.
    1. Evans SM, Funderburk FR, Griffith RR. Zolpidem and triazolam in humans: behavioral and subjective effects and abuse liability. J Pharmacol Exp Ther 1990;255:1246–1255.
    1. Griffiths RR, Johnson MW. Relative abuse ability of hypnotic drugs: A conceptual framework and algorithm for differentiating among compounds. J Clin Psychiatry 2005;66:31–41.
    1. 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;98:1371–1378.
    1. Borbély AA, Mattmann P, Loepfe M, Strauch I, Lehmann D. Effect of benzodiazepine hypnotics on all‐night sleep EEG spectra. Hum Neurobiol 1985;4:189–194.
    1. Brunner DP, Dijk DJ, Munch M, Borbély AA. Effect of zolpidem on sleep and sleep EEG spectra in healthy young men. Psychopharmacology (Berl) 1991;104:1–5.
    1. Feige B, Volderholzer U, Riemann D, Hohagen F, Berger M. Independent sleep EEG slow‐wave sleep and spindle band dynamics associated with 4 weeks of continuous application of short‐half‐life hypnotics in healthy subjects. Clin Neurophysiol 1999;110:1965–1974.
    1. Rosenburg R, Caron J, Roth T, Amato D. An assessment of the efficacy and safety of eszopiclone in the treatment of transient insomnia in healthy adults. Seep Med 2005;6:15–22.
    1. Drake CL, Roehrs TA, Mangano RM, Roth T. Dose‐response effects of zaleplon as compared with triazolam (0.25 mg) and placebo in chronic primary insomnia. Hum Psychopharmacol 2000;15:595–604.
    1. Hemmeter U, Müller M, Bischof R, Annen B, Holsboer‐Trachsler E. Effect of zopiclone and temazepam on sleep EEG parameters, psychomotor and memory functions in healthy elderly volunteers. Psychopharmacology 2000;147:384–396.
    1. Feinberg I, Maloney T, Cambell IG. Effects of hypnotics on the sleep EEG of healthy young adults: New data and psychopharmacologic implications. J Psychiatr Res 2000;34:423–438.
    1. Lerner AB, Case JD, Takahashi Y, Lee TH, Mori W. Isolation of melatonin, the pineal gland factor that lightens melanocytes. J Am Chem Soc 1958;80:2587.
    1. Dubocovich ML. Selective MT2 melatonin receptor antagonists block melatonin‐mediated phase advances of circadian rhythms. FASEB J 1998;12:1211–1220.
    1. Liu C, Weaver DR, Jin X, Shearman LP, Pieschl RL, Gribkoff VK, Reppert SM. Molecular dissection of two distinct actions of melatonin on the suprachiasmatic circadian clock. Neuron 1997;19:91–102.
    1. Wehr TA, Aeschbach D Jr., Duncan WC. Evidence for a biological dawn and dusk in the human circadian timing system. J Physiol 2001;535:937–951.
    1. Akerstedt T, Froberg JE, Friberg Y, Wetterberg L. Melatonin excretion, body temperature and subjective arousal during 64 hours of sleep deprivation. Psychoneuroendocrinology 1979;4:219–225.
    1. Zhdanova IV, Wurtman RJ, Morabito C, Piotrovska VR, Lynch HJ. Effects of low oral doses of melatonin, given 2–4 hours before habitual bedtime, on sleep in normal young humans. Sleep 1996;19:423–431.
    1. Tzischinsky O, Shlitner A, Lavie P. The association between the nocturnal sleep gate and nocturnal onset of urinary 6‐sulfatoxymelatonin. J Biol Rhythms 1993;8:199–209.
    1. Dubocovich ML. Melatonin receptors: Are there multiple subtypes? Trends Pharmacol Sci 1995;16:50–56.
    1. Reppert SM, Weaver DR, Ebisawa T. Cloning and characterization of a mammalian melatonin receptor that mediates reproductive and circadian responses. Neuron 1994;13:1177–1785.
    1. 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;92:8734–8738.
    1. Reppert SM, Weaver DR, Godson C. Melatonin receptors step into the light: Cloning and classification of subtypes. Trends Pharmacol Sci 1996;17:100–102.
    1. Slaugenhaupt SA, Roca AL, Liebert CB, Altherr MR, Gusella JF, Reppert SM. Mapping of the gene for the Mel1a‐melatonin receptor to human chromosome 4 (MTNR1A) and mouse chromosome 8 (Mtnr1a). Genomics 1995;27:355–357.
    1. Dubocovich ML. Molecular pharmacology, regulation and function of mammalian melatonin receptors. Front Biosci 2003;8:d1093–d1108.
    1. Jin X, Von Gall C, Pieschl RL, Gribkoff VK, Stehle JH, Reppert SM, Weaver DR. Targeted disruption of the mouse Mel(1b) melatonin receptor. Mol Cell Biol 2003;23:1054–1060.
    1. Von Gall C, Stehle JH, Weaver DR. Mammalian melatonin receptors: molecular biology and signal transduction. Cell Tissue Res 2002;309:151–62.
    1. Paul P, Lahaye C, Delagrange P, Nicolas JP, Canet E, Boutin JA. Characterization of 2‐[125I]iodomelatonin binding sites in Syrian hamster peripheral organs. J Pharmacol Exp Ther 1999;290:334–340.
    1. Nosjean O, Ferro M, Coge F, Beauverger P, Henlin JM, Lefoulon F, Fauchere JL, Delagrange P, Canet E, Boutin JA. Identification of the melatonin‐binding site MT3 as the quinone reductase 2. J Biol Chem 2000;275:31311–31317.
    1. 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;61:1369–1379.
    1. Miyamoto M, Nishikawa H, Doken Y, Hirai K, Uchikawa O, Ohkawa S. The sleep‐promoting action of ramelteon (TAK‐375) in freely moving cats. Sleep 2004;27:1319–1325.
    1. Hughes RJ, Sack RL, Lewy AJ. The role of melatonin and circadian phase in age‐related sleep‐maintenance insomnia: Assessment in a clinical trial of melatonin replacement. Sleep 1998;21:52–68.
    1. Zhdanova IV, Wurtman RJ, Lynch HJ, Ives JR, Dollins AB, Morabito C, Matheson JK, Schomer DL. Sleep‐inducing effects of low doses of melatonin ingested in the evening. Clin Pharmacol Ther 1995;57:552–558.
    1. Zhdanova IV, Wurtman RJ, Regan MM, Taylor JA, Shi JP, Leclair OU. Melatonin treatment for age‐related insomnia. J Clin Endocrinol Metab 2001;86:4727–4730.
    1. Zhdanova IV, Wurtman RJ. Efficacy of melatonin as a sleep‐promoting agent. J Biol Rhythms 1997;12:644–650.
    1. Arendt J. Importance and relevance of melatonin to human biological rhythms. J Neuroendocrinol 2003;15:427–431.
    1. Arendt J, Deacon S, English J, Hampton S, Morgan L. Melatonin and adjustment to phase shift. J Sleep Res 1995;4:74–79.
    1. Sharkey KM, Fogg LF, Eastman CI. Effects of melatonin administration on daytime sleep after simulated night shift work. J Sleep Res 2001;10:181–192.
    1. Hardeland R, Poeggeler B, Srinivasan V, Trankht I, Pandi‐Perumal SR, Cardinali DP. Melatonergic drugs in clinical practice. Arzneimittelforsung 2008;58:1–10.
    1. Yeleswaram K, McLaughlin LG, Knipe JO, Schabdach D. Pharmacokinetics and oral bioavailability of exogenous melatonin in preclinical animal models and clinical implications. J Pineal Res 1997;22:45–51.
    1. Garfinkel D, Laudon M, Zisapel N. Improvement of sleep quality by controlled‐release melatonin in benzodiazepine‐treated elderly insomniacs. Arch Gerontol Geriatr 1997;24:223–231.
    1. Haimov I, Lavie P, Laudon M, Herer P, Vigder C, Zisapel N. Melatonin replacement therapy of elderly insomiacs. Sleep 1995;18:598–603.
    1. Faust R, Garratt PJ, Trujillo Perez MA, Piccio VJ, Madsen C, Stenstrom A, Frolund B, Davidson K, Teh MT, Sugden D. 7‐substituted‐melatonin and 7‐substitued‐1‐methylmelatonin analogues: Effect of substituents on potency and binding affinity. Bioorg Med Chem 2007;15:4543–4551.
    1. Pandi‐Perumal SR, Srinivasan V, Poeggeler B, Hardeland R, Cardinali DP. Drug insight: The use of melatonergic agonists for the treatment of insomnia‐focus on ramelteon. Nat Clin Pract Neurol 2007;3:221–228.
    1. Rivara S, Lodola A, Mor M, Bedini A, Spandoni G, Lucini V, Pannacci M, Fraschini F, Scaglione F, Sanchez RO, et al N‐(substituted‐anilinoethyl)amides: Design, synthesis, and pharmacological characterization of a new class of melatonin receptor ligands. J Med Chem 2007;50:6618–6626.
    1. Uchikawa O, Fukatsu K, Tokunoh R, Kawada M, Matsumoto K, Imai Y, Hinuma S, Kato K, Nishikawa H, Hirai K, et al Synthesis of a novel series of tricyclic indan derivatives as melatonin receptor agonists. J Med Chem 2002;45:4222–4239.
    1. 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;46:140–148.
    1. Hibberd M, Stevenson SJ. A phase‐I open‐label study of the absorption, metabolism, and excretion of (14C)‐ramelteon (TAK‐375) following a single oral dose in healthy male subjects. Sleep 2004;27:A54.
    1. Johnson MW, Suess PE, Griffiths RR. Ramelteon: A novel hypnotic lacking abuse liability and sedative side effects. Arch Gen Psychiatry 2006;63:1149–1157.
    1. Zammit G, Schwartz H, Roth T, Wright L, Sainati S, Zhang J. Phase III study of ramelteon in a first‐night‐effect model of transient insomnia. Sleep Med 2005;6:S50–S51.
    1. Zammit G, Erman M, Weigand S, Sainati S, Zhang J, Roth T. Evaluation of the efficacy and safety of ramelteon in subjects with chronic insomnia. J Clin Sleep Med 2007;3:495–504.
    1. Kato K, Hirai K, Nishiyama K, Uchikawa O, Fukatsu K, Ohkawa S, Kawamata Y, Hinuma S, Miyamoto M. Neurochemical properties of ramelteon (TAK‐375), a selective MT1/MT2 receptor agonist. Neuropharmacology 2005;48:301–310.
    1. Niles LP, Pickering DS, Sayer BG. HPLC‐purified 2‐[125I]iodomelatonin labels multiple binding sites in hamster brain. Biochem Biophys Res Commun 1987;147:49–56.
    1. Pickering DS, Niles LP. Pharmacological characterization of melatonin binding sites in Syrian hamster hypothalamus. Eur J Pharmacol 1990;175:71–77.
    1. Morgan PJ, Lawson W, Davidson G, Howell HE. Melatonin inhibits cyclic AMP in cultured ovine pars tuberalis cells. J Mol Endocrinol 1989;5:R5–R8.
    1. Vanecek J, Vollrath L. Melatonin inhibits cyclic AMP and cyclic GMP accumulation in the rat pituitary. Brain Res 1989;505:157–159.
    1. Yukuhiro N, Kimura H, Nishikawa H, Ohkawa S, Yoshikubo S, Miyamoto M. Effects of ramelteon (TAK‐375) on nocturnal sleep in freely moving monkeys. Brain Res 2004;1027:59–66.
    1. Daley JT, Turner RS, Freeman A, Bliwise DL, Rye DB. Prolonged assessment of sleep and daytime sleepiness in unrestrained Macaca mulatta . Sleep 2006;29:221–231.
    1. Buscemi N, Vandermeer B, Friesen C, Bialy L, Tubman M, Ospina M, Klassen TP, Witmans M. The efficacy and safety of drug treatments for chronic insomnia in adults: A meta‐analysis of RCTs. J Gen Intern Med 2007;22:1335–1350.
    1. 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.
    1. Hirai K, Kita M, Ohta H, Nishikawa H, Fujiwara Y, Ohkawa S, Miyamoto M. Ramelteon (TAK‐375) accelerates reentrainment of circadian rhythm after a phase advance of the light‐dark cycle in rats. J Biol Rhythms 2005;20:27–37.
    1. Inoue S, Shimazoe T, Moriya T, Shinohara K, Miyamoto M, Watanabe S. Effects of TAK‐375, a novel ML1‐selective melatonin receptor agonist, on entrainment function in circadian rhythm. J Pharmacol Sci 2003;91:90.
    1. Miyamoto M. Effect of ramelteon (TAK‐375), a selective MT1/MT2 receptor agonist, on motor performance in mice. Neurosci Lett 2006;402:201–204.
    1. Spyraki C, Fibiger HC. A role for the mesolimbic dopamine system in the reinforcing properties of diazepam. Psychopharmacology (Berl) 1988;94:133–137.
    1. France CP, Weltman RH, Koek H, Cruz CM, McMahon LR. Acute and chronic effects of ramelteon in rhesus monkeys (Macaca mulatta): Dependence liability studies. Behav Neurosci 2006;120:535–541.
    1. Nishida N, Sasaki M, Wakasa Y, Awasaki Y, Yamamoto M, Yanagita T. Reinforcing effect of ramelteon assessed by intravenous self‐administration experiments in rhesus monkeys. Sleep 2005;28:A5.
    1. 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;28:303–307.
    1. Roth T, Seiden D, Weigand S, Zhang J. A 2‐night, 3‐period, crossover study of ramelteon's efficacy and safety in older adults with chronic insomnia. Curr Med Res Opinion 2007;23:1005–1014.
    1. 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 Medicine 2006;7:312–318.
    1. DeMicco M, Wang‐Weigand S, Zhang J. Long‐term therapeutic effects of ramelteon treatment in adults with chronic insomnia: A 1‐year study. Sleep 2006;29:A234.
    1. Richardson G, Wang‐Weigand S, Zhang J, DeMicco M. Long‐term safety of ramelteon treatment in adults with chronic insomnia: A 1‐year study. Sleep 2006;29:A233.
    1. Partinen M. Epidemiology of sleep disorders In: Kryger MH, Roth T, Dement WC, editors. Principles and Practice of Sleep Medicine, 2nd ed Philadelphia , PA : WB Saunders, 1994; 437–452.
    1. Sack RL, Lewy AJ, Blood ML, Keith LD, Nakagawa H. Circadian rhythm abnormalities in totally blind people: Incidence and clinical significance. J Clin Endocrinol Metab 1992;75:127–134.
    1. Dahlitz M, Alvarez B, Vignau J, English J, Arendt J, Parkes JD. Delayed sleep phase response to melatonin. Lancet 1991;337:1121–1124.
    1. Oldani A, Ferini‐Strambi L, Zucconi M, Stankov B, Fraschini F, Smirne S. Melatonin and delayed sleep phase syndrome: Ambulatory polygraphic evaluation. Neuroreport 1994;6:132–134.
    1. Nagtegaal JE, Kerkhof GA, Smits MG, Swart AC, Van Der Meer YG. Delayed sleep phase syndrome: A placebo‐controlled cross‐over study on the effects of melatonin administered five hours before the individual dim light melatonin onset. J Sleep Res 1998;7:135–143.
    1. Herxheimer A, Petrie KJ. Melatonin for prevention and treatment of jet lag. Cochrane Database Syst Rev 2001;1:CD001520.
    1. Crasson M, Kjiri S, Colin A, Kjiri K, L'Hermite‐Baleriaux M, Ansseau M, Legros JJ. Serum melatonin and urinary 6‐sulfatoxymelatonin in major depression. Psychoneuroendocrinology 2004;29:1–12.
    1. Rubin RT, Heist EK, McGeoy SS, Hanada K, Lesser IM. Neuroendocrine aspects of primary endogenous depression. XI. Serum melatonin measures in patients and matched control subjects. Arch Gen Psychiatry 1992;49:558–567.
    1. Loo H, Dalery J, Macher JP, Payen A. Pilot study comparing in blind therapeutic effect of two doses of agomelatine, melatonin and 5‐HT2c receptors antagonist, in the treatment of major depressive disorders. Encephale 2002;28:356–362.
    1. Montgomery SA, Kasper S. Severe depression and antidepressants: Focus on a pooled analysis of placebo‐controlled studies on agomelatine. Int Clin Psychopharmacol 2007;22:283–291.
    1. Kennedy SH, Kutcher SP, Ralevski E, Brown GM. Nocturnal melatonin and 24‐hour 6‐sulphatoxymelatonin levels in various phases of bipolar affective disorder. Psychiatry Res 1996;63:219–222.
    1. Srinivasan V, Smits M, Spence W, Lowe AD, Kayumov L, Pandi‐perumal SR, Parry B, Cardinali DP. Melatonin in mood disorders. World J Biol Psychiatry 2006;7:138–151.
    1. Nair NP, Hariharasubramanian N, Pilapil C, Isaac I, Thavundayil JX. Plasma melatonin—an index of brain aging in humans? Biol Psychiatry 1986;21:141–150.
    1. Mirmiran M, Swaab DF, Kok JH, Hofman MA, Witting W, Van Gool WA. Circadian rhythms and the suprachiasmatic nucleus in perinatal development, aging and Alzheimer's disease. Prog Brain Res 1992;93:151–162.
    1. Skene DJ, Vivien‐Roels B, Sparks DL, Hunsaker JC, Pevet P, Ravid D, Swaab DF. Daily variation in the concentration of melatonin and 5‐methoxytryptophol in the human pineal gland: Effect of age and Alzheimer's disease. Brain Res 1990;528:170–174.
    1. Wu YH, Swaab DF. The human pineal gland and melatonin in aging and Alzheimer's disease. J Pineal Res 2005;38:145–152.
    1. Asayama K, Yamadera H, Ito T, Suzuki H, Kudo Y, Endo S. Double blind study of melatonin effects on the sleep‐wake rhythm, cognitive and non‐cognitive functions in Alzheimer type dementia. J Nippon Med Sch 2003;70:334–341.
    1. Caldinali DP, Brusco LI, Liberczuk C, Furio AM. The use of melatonin in Alzheimer's disease. Neuroendocrinol Lett 2002;23(Suppl):20–23.
    1. Mahlberg R, Kunz D, Sutej I, Kuhl KP, Hellweg R. Melatonin treatment of day‐night rhythm disturbances and sundowning in Alzheimer disease: An open‐label pilot study using actigraphy. J Clin Psychopharmacol 2004;24:456–459.

Source: PubMed

3
Abonnere