MedPath

Ramelteon Advanced Drug Monograph

Published:Aug 7, 2025

Generic Name

Ramelteon

Brand Names

Rozerem

Drug Type

Small Molecule

Chemical Formula

C16H21NO2

CAS Number

196597-26-9

Associated Conditions

Insomnia

A Comprehensive Pharmacological and Clinical Review of Ramelteon (DB00980)

Section 1: Executive Summary

1.1. Overview

Ramelteon represents a significant departure from traditional hypnotic agents, establishing a novel therapeutic class as a selective melatonin receptor agonist. Marketed under the brand name Rozerem, it is indicated for the treatment of insomnia characterized by difficulty with sleep onset.[1] Its development and approval marked a paradigm shift in the pharmacological management of sleep disorders, moving away from the mechanism of generalized central nervous system (CNS) depression employed by benzodiazepines and related drugs. Instead, Ramelteon leverages a chronobiotic mechanism, targeting the physiological pathways that regulate the body's intrinsic sleep-wake cycle.[3]

1.2. Key Differentiators

The defining characteristic of Ramelteon is its unique mechanism of action. It acts as a potent and selective agonist at the melatonin MT1 and MT2 receptors located within the suprachiasmatic nucleus (SCN) of the hypothalamus, the body's master circadian pacemaker.[5] By mimicking the effects of endogenous melatonin, it promotes the natural signaling cascade that initiates sleep.[4] This targeted action stands in stark contrast to older hypnotics that modulate the gamma-aminobutyric acid (GABA) receptor complex.[3] A direct consequence of this mechanistic specificity is Ramelteon's exceptional safety profile. Extensive clinical evaluation has demonstrated a lack of abuse potential, physical dependence, or withdrawal phenomena upon discontinuation.[1] This has led to its unique status among prescription hypnotics as a non-controlled substance in the United States, a critical differentiator for patients with a history of or predisposition to substance use disorders.[3]

1.3. Efficacy and Limitations

The clinical efficacy of Ramelteon in its approved indication is statistically significant but modest in magnitude. Pivotal trials have consistently shown that it reduces the latency to persistent sleep compared to placebo.[9] However, the absolute reduction in sleep onset time is often small, on the order of several minutes, which has led to debate regarding its clinical relevance and contributed to its non-approval by the European Medicines Agency.[1] Its effects on sleep maintenance and total sleep time are less consistent.[1] The therapeutic utility of Ramelteon is further constrained by its pharmacokinetic profile, which is characterized by extremely low oral bioavailability (1.8%) due to extensive first-pass metabolism.[11] This metabolic vulnerability makes it highly susceptible to clinically significant drug-drug interactions, most notably a contraindication with the potent CYP1A2 inhibitor fluvoxamine.[13]

1.4. Place in Therapy

Within the contemporary therapeutic armamentarium for insomnia, Ramelteon occupies a distinct niche defined by safety. It is a primary therapeutic option for patient populations in whom the risks associated with traditional hypnotics are unacceptable, such as individuals with a history of substance use disorder, the elderly who are at high risk for falls and cognitive impairment, or patients seeking to avoid agents with dependence liability.[14] Beyond its approved indication, a compelling body of evidence is emerging for its off-label use in the prevention of delirium in hospitalized patients, a condition for which there are no approved pharmacological treatments. Meta-analyses suggest a significant prophylactic effect, positioning this as a promising and potentially high-impact future application for the drug.[1]

Section 2: Chemical Identity and Physicochemical Properties

2.1. Nomenclature and Identifiers

The unambiguous identification of a pharmaceutical agent is foundational to all scientific and clinical discourse. Ramelteon is the established generic name for this small molecule drug.[1] It was developed by Takeda Pharmaceuticals, Inc. under the developmental code name TAK-375 and is marketed in the United States under the brand name Rozerem.[2]

Its formal chemical identity is defined by the International Union of Pure and Applied Chemistry (IUPAC) name: (S)-N-(2-(1,6,7,8-tetrahydro-2H-indeno[5,4-b]furan-8-yl)ethyl)propionamide.[12] To ensure global uniqueness and facilitate database cross-referencing, it is assigned several standard identifiers. The Chemical Abstracts Service (CAS) has assigned it the number 196597-26-9.[1] In major biomedical databases, it is cataloged as DrugBank ID DB00980, PubChem Compound ID 208902, and IUPHAR/BPS ID 1356.[1] These identifiers are essential for retrieving consistent and accurate information across diverse chemical, pharmacological, and clinical data repositories.

2.2. Chemical Structure and Properties

Ramelteon is a tricyclic indan derivative, classified as a member of the indanes chemical family.[2] Its empirical formula is

C16​H21​NO2​, corresponding to a molecular weight of approximately 259.34 g/mol.[17] The molecule possesses a single chiral center and is produced exclusively as the (S)-enantiomer, which is the pharmacologically active form.[2]

The chemical architecture of Ramelteon is a product of deliberate medicinal chemistry aimed at optimizing the properties of the natural hormone melatonin. The structure represents a key modification of the native melatonin molecule, where the nitrogen atom of the indole ring system has been replaced with a carbon atom to form a more stable indane ring. Furthermore, the flexible 5-methoxyl group of melatonin has been incorporated into a rigid dihydrofuran ring fused to the indane core.[2] These specific structural alterations are not incidental; they are the direct source of Ramelteon's enhanced pharmacodynamic and pharmacokinetic properties relative to its parent compound, conferring higher receptor affinity, greater selectivity, and a longer biological half-life.[2] This process of modifying a natural ligand to create a therapeutically superior drug is a successful example of rational drug design, retaining the desired mechanism of melatonin agonism while engineering a profile suitable for a modern pharmaceutical.

In its solid state, Ramelteon appears as a white to almost white crystalline solid or powder.[2] Its solubility profile presents a significant formulation challenge. It is freely soluble in organic solvents such as methanol, ethanol, and dimethyl sulfoxide (DMSO), but it is characterized as very slightly soluble or practically insoluble in water and across a wide range of aqueous buffer pH values.[12] The measured aqueous solubility is very low, approximately 11 mg/L at 25°C.[21] This poor aqueous solubility dictates its formulation as a solid oral tablet and is a key factor influencing its dissolution and absorption characteristics. Despite this, in vitro studies have demonstrated that the molecule is stable in simulated gastric and intestinal fluids, indicating that it is not chemically degraded in the gastrointestinal tract prior to absorption.[23] The melting point of the crystalline solid is in the range of 114.0 to 118.0 °C.[21]

Table 1: Ramelteon - Key Identifiers and Physicochemical Properties

PropertyValueSource(s)
Generic NameRamelteon1
Brand NameRozerem1
Developmental CodeTAK-37517
DrugBank IDDB009801
CAS Number196597-26-91
IUPAC Name(S)-N-(2-(1,6,7,8-tetrahydro-2H-indeno[5,4-b]furan-8-yl)ethyl)propionamide12
Chemical FormulaC16​H21​NO2​17
Molecular Weight259.34 g/mol17
AppearanceWhite to almost white crystalline solid/powder2
Aqueous SolubilityVery slightly soluble (~11 mg/L at 25°C)12
Organic SolubilityFreely soluble in methanol, ethanol, DMSO18
Melting Point114.0 - 118.0 °C21
OriginatorTakeda Pharmaceuticals, Inc.2

Section 3: Comprehensive Pharmacological Profile

3.1. Pharmacodynamics: A Novel Chronobiotic Mechanism of Action

The pharmacological activity of Ramelteon is fundamentally different from that of traditional sedative-hypnotic agents. Rather than inducing sleep through generalized CNS depression, Ramelteon acts as a chronobiotic, a substance that adjusts the timing of the internal biological clock. Its mechanism is a precise mimicry of the natural physiological process that governs the sleep-wake cycle.[3]

3.1.1. Selective Melatonin Receptor Agonism

Ramelteon is a potent and selective agonist of the melatonin MT1 and MT2 receptors.[1] These G protein-coupled receptors are densely expressed in the suprachiasmatic nucleus (SCN) of the hypothalamus, a small region of the brain that functions as the body's "master clock," regulating circadian rhythms over a roughly 24-hour cycle.[4] In vitro studies confirm that Ramelteon demonstrates full agonist activity at both human MT1 and MT2 receptors, with binding affinities in the low picomolar range, indicating high potency.[1] For example, reported binding affinity constants (

Ki​) are as low as 14 pM for human MT1 and 112 pM for human MT2 receptors.[22] By binding to and activating these receptors, Ramelteon mimics the action of endogenous melatonin, the hormone naturally produced by the pineal gland in response to darkness. This activation serves as a powerful signal to the SCN, promoting the transition from wakefulness to sleep in alignment with the biological night.[4]

3.1.2. Receptor Subtype Selectivity and Functional Roles

The two primary melatonin receptor subtypes, MT1 and MT2, are thought to mediate distinct but complementary functions. Activation of the MT1 receptor is believed to be primarily responsible for attenuating the wakefulness-promoting signals from the SCN, thereby directly promoting sleepiness and facilitating the onset of sleep.[5] In contrast, activation of the MT2 receptor is thought to be more involved in the phase-shifting effects of melatonin on the circadian rhythm, helping to entrain or reset the biological clock to the external light-dark cycle.[5]

Ramelteon exhibits a distinct binding profile, with an approximately 8-fold higher affinity for the MT1 receptor compared to the MT2 receptor.[1] This preferential binding to MT1 suggests that its pharmacological action may be more pronounced in initiating sleep (an MT1-mediated function) rather than in producing robust phase shifts of the circadian clock (an MT2-mediated function). This pharmacological nuance aligns perfectly with its approved clinical indication for the treatment of sleep-onset insomnia, as opposed to primary circadian rhythm sleep disorders.[1] Furthermore, Ramelteon demonstrates very low affinity for the MT3 receptor, which has since been identified as the enzyme quinone reductase 2 and is not believed to be involved in sleep regulation.[1]

3.1.3. Lack of Affinity for Other CNS Receptors

A defining pharmacodynamic feature of Ramelteon, and the cornerstone of its safety profile, is its remarkable selectivity. Extensive receptor binding assays have confirmed that Ramelteon has no appreciable affinity for a wide array of other CNS targets.[1] This includes the GABA receptor complex, which is the primary target of benzodiazepines and Z-drugs, as well as receptors for benzodiazepines, opiates, serotonin, dopamine, norepinephrine, acetylcholine, and various other neuropeptides and cytokines.[1] It also does not interfere with the activity of numerous enzymes in standard screening panels.[21]

This "clean" receptor binding profile is the direct pharmacological basis for Ramelteon's favorable safety and tolerability. Unlike GABAergic agents, which achieve hypnosis through widespread CNS depression, Ramelteon's targeted action avoids the receptor systems responsible for the common side effects of older hypnotics, such as anxiolysis, euphoria, significant motor impairment, anterograde amnesia, and respiratory depression. This mechanistic purity is the fundamental reason why Ramelteon has been shown to lack abuse potential, does not produce physical dependence, and is associated with a lower incidence of next-day residual effects.[3]

3.1.4. Role of the Active Metabolite (M-II)

Following administration, Ramelteon is extensively metabolized into several compounds, with one major active metabolite known as M-II.[20] This metabolite is also an agonist at the MT1 and MT2 receptors, but it is significantly less potent than the parent drug. In vitro functional assays show M-II to be 17 to 25 times less potent than Ramelteon, and its binding affinity for the MT1 and MT2 receptors is approximately one-tenth and one-fifth that of the parent molecule, respectively.[12]

Despite its lower potency, the M-II metabolite circulates in the bloodstream at concentrations that are 20- to 100-fold higher than the parent drug, resulting in a much greater overall systemic exposure.[12] This discrepancy arises from the extensive first-pass metabolism of Ramelteon. The pharmacokinetic profile of M-II is also different, with a longer elimination half-life (2–5 hours) compared to the parent drug (1–2.6 hours).[1] The combination of its sustained, high systemic exposure and its longer duration of action suggests that M-II likely contributes significantly to the overall therapeutic effect of the medication. While the rapid peak of the highly potent parent drug is primarily responsible for the initial sleep-onset effect, the prolonged presence of the M-II metabolite may provide a continuing, albeit weaker, melatonergic signal throughout the night, potentially contributing to the maintenance of the drug's effect with long-term use.

3.2. Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)

The clinical behavior of Ramelteon is profoundly influenced by its pharmacokinetic properties. Its journey through the body—from absorption to elimination—is characterized by rapid absorption, extensive distribution, and near-complete metabolic clearance, which collectively dictate its dosing, administration, and potential for drug interactions.

3.2.1. Absorption

After oral administration in a fasted state, Ramelteon is absorbed rapidly from the gastrointestinal tract. Peak plasma concentrations (Cmax) are typically reached at a median time (Tmax) of approximately 0.75 hours, with a range of 0.5 to 1.5 hours.[11] Mass balance studies using radiolabeled drug indicate that the total absorption from the gut is high, with at least 84% of the administered dose being absorbed.[5]

However, this high absorption is contrasted by an extremely low absolute oral bioavailability of only 1.8%.[11] This significant discrepancy is due to extensive and rapid first-pass metabolism, primarily in the liver, where the vast majority of the absorbed drug is metabolized before it can reach systemic circulation. This pharmacokinetic characteristic makes the systemic exposure of Ramelteon highly sensitive to factors that affect hepatic metabolism.

The absorption of Ramelteon is significantly affected by the presence of food. Administration with or immediately after a high-fat meal delays the rate of absorption, increasing the Tmax by approximately 45 minutes, and decreases the peak concentration (Cmax) by approximately 22%.[11] This delay can blunt the drug's sleep-initiating effect, which is the basis for the clinical recommendation to take Ramelteon on an empty stomach within 30 minutes of going to bed.[11]

3.2.2. Distribution

Once in the systemic circulation, Ramelteon is approximately 82% bound to plasma proteins, primarily albumin (accounting for 70% of binding), and this binding is independent of the drug's concentration.[5] It is extensively distributed into tissues and does not selectively accumulate in red blood cells.[11]

3.2.3. Metabolism

Metabolism is the primary route of elimination for Ramelteon and the reason for its low bioavailability. It undergoes extensive and rapid first-pass metabolism, principally via oxidation (hydroxylation and carbonyl formation) followed by secondary metabolism to form glucuronide conjugates.[20] This process occurs mainly in the liver and is mediated by several cytochrome P450 (CYP) isoenzymes.

The primary enzyme responsible for Ramelteon's metabolism is CYP1A2, which is estimated to contribute to approximately 49% of its clearance.[11] The secondary major pathway is through CYP2C19, accounting for an estimated 42% of metabolism.[27] A third, minor pathway involves CYP3A4, which contributes about 8.6%.[27] This heavy reliance on specific CYP pathways, particularly CYP1A2, is the root cause of its most significant clinical risks and drug-drug interactions. Any factor that inhibits these enzymes can dramatically increase the systemic exposure of Ramelteon. For instance, shutting down the primary CYP1A2 clearance pathway with a potent inhibitor like fluvoxamine leads to a disproportionately massive increase in systemic drug levels, turning a therapeutic dose into a significant overdose.[13] Similarly, hepatic impairment, which reduces the liver's metabolic capacity, effectively removes the "brake" on the drug's entry into the systemic circulation, leading to dangerously elevated exposure.[12] The clinical safety of Ramelteon is therefore fundamentally tethered to the patient's metabolic capacity, necessitating careful consideration of liver function and concomitant medications.

3.2.4. Excretion

Elimination of Ramelteon from the body is almost entirely dependent on its metabolism. Following an oral dose, approximately 84% is recovered in the urine and about 4% in the feces, almost exclusively in the form of metabolites.[1] An exceedingly small fraction, less than 0.1% of the dose, is excreted as the unchanged parent drug, highlighting the efficiency of its metabolic clearance.[12] The elimination half-life of the parent Ramelteon molecule is short, ranging from 1 to 2.6 hours.[1] As previously noted, its major active metabolite, M-II, has a longer elimination half-life of 2 to 5 hours, which contributes to the overall duration of the drug's effect.[1]

3.2.5. Pharmacokinetics in Special Populations

The pharmacokinetic profile of Ramelteon is altered in certain populations, requiring clinical consideration:

  • Geriatric Patients: In elderly subjects (≥65 years), both the peak plasma concentration (Cmax) and total systemic exposure (AUC) of Ramelteon are substantially increased, by approximately 86% and 97%, respectively, compared to younger adults.[11] Exposure to the M-II metabolite is also increased, though to a lesser extent. Despite these significant pharmacokinetic differences, clinical studies have not identified a need for dose adjustment in the elderly, likely due to the drug's wide therapeutic window.[20]
  • Hepatic Impairment: Liver function has a profound impact on Ramelteon exposure. In patients with mild hepatic impairment (Child-Pugh Class A), the AUC is increased nearly 4-fold. In patients with moderate hepatic impairment (Child-Pugh Class B), the exposure is even more dramatically increased, with the AUC rising by more than 10-fold.[11] Consequently, Ramelteon should be used with caution in patients with moderate hepatic impairment and is not recommended for use in patients with severe hepatic impairment (Child-Pugh Class C).[12]
  • Renal Impairment: Renal impairment, including patients requiring chronic hemodialysis, does not appear to significantly alter the pharmacokinetic parameters of Ramelteon or its M-II metabolite.[11] No dosage adjustment is necessary in this population.

Table 2: Summary of Ramelteon Pharmacokinetic Parameters

ParameterValue / DescriptionClinical ImplicationSource(s)
Absorption (Tmax)Rapid; ~0.75 hours (fasted)Provides rapid onset of action suitable for sleep-onset insomnia.11
Bioavailability1.8% (absolute oral)Extremely low due to extensive first-pass metabolism. Systemic levels are highly sensitive to changes in hepatic function or metabolizing enzyme activity.11
Effect of FoodHigh-fat meal delays Tmax (~45 min) and reduces Cmax (~22%)Should be taken on an empty stomach within 30 minutes of bedtime to ensure maximal and timely effect.11
Distribution~82% plasma protein bound (mainly albumin)Extensive tissue distribution. Standard protein binding with low potential for displacement interactions.5
MetabolismExtensive hepatic metabolism via CYP450 enzymes: CYP1A2 (~49%), CYP2C19 (~42%), CYP3A4 (~9%)High potential for significant drug-drug interactions with inhibitors or inducers of these enzymes, especially CYP1A2. This is the drug's "Achilles' heel."11
Elimination Half-LifeRamelteon: 1–2.6 hours; Active Metabolite (M-II): 2–5 hoursShort half-life of parent drug minimizes next-day hangover effects. Longer half-life of M-II may contribute to sustained effect.1
Excretion84% in urine, 4% in feces, almost entirely as metabolites (<0.1% unchanged)Elimination is dependent on hepatic metabolism, not renal function. No dose adjustment needed in renal impairment.1

Section 4: Clinical Applications and Efficacy

The clinical utility of Ramelteon has been evaluated across a range of sleep and psychiatric disorders. While its approval is narrow, its unique mechanism has prompted investigation into several off-label applications, with varying degrees of success.

4.1. Approved Indication: Treatment of Sleep-Onset Insomnia

Ramelteon is approved by the U.S. Food and Drug Administration (FDA) for the treatment of insomnia characterized by difficulty with sleep onset in adult patients.[1] It is one of the few prescription hypnotics approved for long-term use without limitation on the duration of therapy, a testament to its safety and lack of dependence liability.[1] The standard recommended dosage is an 8 mg tablet taken orally within 30 minutes of going to bed.[13]

The efficacy of Ramelteon for this indication has been established in numerous randomized, placebo-controlled clinical trials. Pivotal Phase 3 studies, such as NCT00671086 and NCT00756002, were designed to assess its effect on objectively measured sleep parameters using polysomnography (PSG).[30] A key study by Zammit et al. evaluated Ramelteon (8 mg and 16 mg) over five weeks in subjects with chronic insomnia. The primary endpoint, Latency to Persistent Sleep (LPS), was significantly reduced with the 8 mg dose to 32.2 minutes, compared to 47.9 minutes for placebo, during the first week of treatment (a reduction of over 15 minutes, p<0.001). This statistically significant improvement in sleep onset was maintained at subsequent assessments at weeks 3 and 5.[9] Subjective measures, such as patient-reported sleep latency (sSL), also showed significant reductions compared to placebo.[9]

While the statistical significance of these findings is robust, the clinical magnitude of the effect is considered modest. Various meta-analyses have quantified the average reduction in subjective sleep latency to be in the range of 4 to 9 minutes compared to placebo.[1] A 2009 pooled analysis reported a more substantial 13-minute reduction.[1] The American Academy of Sleep Medicine (AASM) 2017 guidelines estimated the benefit at a 9-minute reduction in sleep latency.[1] The evidence for improvement in other sleep parameters, such as Total Sleep Time (TST) and wake after sleep onset (WASO), is mixed and less consistent.[1]

This discrepancy between statistical significance and perceived clinical magnitude was central to the decision by the European Medicines Agency (EMA) not to approve Ramelteon for use in the European Union. The EMA's Committee for Medicinal Products for Human Use (CHMP) concluded that the efficacy was not convincingly demonstrated across all pivotal trials and that the observed improvement in sleep onset was too small to be considered clinically meaningful.[1] Therefore, Ramelteon's place in treating insomnia is largely defined by a trade-off: it offers a reliable, albeit small, improvement in sleep latency in exchange for a superior safety profile compared to more potent agents.

4.2. Investigational and Off-Label Use: Delirium Prevention

Perhaps the most promising and clinically significant application for Ramelteon lies outside its approved indication, in the prevention of delirium. Delirium is a common and severe form of acute brain dysfunction in hospitalized patients, associated with increased morbidity, mortality, and healthcare costs, yet it has no FDA-approved pharmacological treatment. The pathophysiology of delirium is strongly linked to disruption of the sleep-wake cycle and circadian rhythms, making Ramelteon's mechanism of action an ideal therapeutic target.

A growing body of evidence from systematic reviews and meta-analyses supports this application. An early systematic review in 2014 found Ramelteon to be beneficial for preventing delirium in medically ill individuals.[1] More recently, a 2023 meta-analysis of eight randomized controlled trials (RCTs) involving 587 patients provided robust evidence. The analysis concluded that Ramelteon was associated with a 50% lower odds of delirium occurrence compared to placebo (Odds Ratio 0.50; 95% Confidence Interval [CI] 0.29-0.86).[16] A trial sequential analysis confirmed that this finding was statistically reliable and not a result of random error.[16] The prophylactic effect appears to be most pronounced in elderly patients (OR 0.28) and when administered over multiple days.[16]

However, evidence from studies in specific surgical populations is mixed. While some retrospective studies and RCTs have shown a benefit in preventing postoperative delirium after procedures like liver resection or thoracic surgery [32], other trials in patients undergoing general or cardiac surgery found no significant effect.[33] This suggests the benefit may be context-dependent. There is also emerging evidence that combination therapy with an orexin receptor antagonist, such as suvorexant, may be particularly effective in preventing delirium.[1] Several ongoing clinical trials, including NCT05069428, are further investigating Ramelteon's role in preventing and treating delirium in the intensive care unit (ICU) setting, aiming to solidify its place in this area of high unmet medical need.[35] The potential for Ramelteon to become a standard-of-care agent for delirium prophylaxis represents a significant evolution from its original, modestly effective role in primary insomnia.

4.3. Investigational and Off-Label Use: Circadian Rhythm and Psychiatric Disorders

4.3.1. Circadian Rhythm Sleep Disorders (CRSDs)

Given that Ramelteon's mechanism directly targets the circadian system, its use in treating CRSDs like Delayed Sleep Phase Syndrome (DSPS) is a logical extension. Melatonin agonists as a class are considered effective for regulating sleep-wake cycles in these disorders.[1] Indeed, Ramelteon was evaluated in a proof-of-concept clinical trial for its ability to advance the timing of sleep in individuals with DSPS (NCT00593736).[36] Despite this mechanistic rationale, Ramelteon is not approved for the treatment of any CRSD. Development for these indications, including jet lag disorder and shift work disorder, was officially discontinued.[1] This outcome may reflect a failure to meet primary endpoints in clinical trials or a strategic business decision to focus on the larger primary insomnia market.

4.3.2. Bipolar Disorder

The link between circadian rhythm disruption and bipolar disorder is well-established, leading to the hypothesis that stabilizing the sleep-wake cycle with an agent like Ramelteon could improve mood stability and prevent relapse.[37] Early research provided a promising signal. A small, 24-week, placebo-controlled trial (n=83) in patients with euthymic bipolar disorder and sleep disturbances (the Ram-TIME study, NCT00552760) found that adjunctive Ramelteon was associated with a significantly lower risk of relapse into a manic or depressive episode. Patients treated with Ramelteon were approximately half as likely to relapse as those receiving placebo (OR 0.48, p=0.024), and the median time to relapse was more than doubled (188 days vs. 84 days).[37]

This promising early finding, however, failed to be replicated in larger, more definitive studies. A subsequent large, multi-center Phase 3 trial (NCT01677182) evaluating a sublingual formulation of Ramelteon as an adjunctive maintenance therapy was terminated early for futility. A planned interim analysis revealed no significant difference between any dose of Ramelteon and placebo in preventing mood episode relapse.[39] Other related trials were also withdrawn or terminated for business reasons prior to enrollment.[41] This trajectory represents a common narrative in drug development, where a strong signal from a small, early-phase study does not translate into success in a larger, more rigorous Phase 3 setting. It underscores that while sleep and circadian disruption are core features of bipolar disorder, targeting this system with a single chronobiotic agent is likely insufficient to overcome the complex, multifactorial neurobiology of mood relapse.

Table 3: Summary of Pivotal and Investigational Clinical Trials for Ramelteon

IndicationTrial Identifier / StudyPhasePopulationInterventionPrimary EndpointKey Result / OutcomeSource(s)
Chronic InsomniaZammit et al. (2007)3Adults with chronic insomniaRamelteon 8 mg vs. PlaceboChange in Latency to Persistent Sleep (LPS)Significant reduction in LPS at Week 1 (32.2 min vs 47.9 min for placebo; p<0.001), maintained for 5 weeks.9
Chronic InsomniaNCT004922324Adults with chronic insomnia on zolpidemRamelteon 8 mg vs. PlaceboFacilitation of zolpidem discontinuationInvestigated use of Ramelteon to aid in tapering off Z-drugs.42
Delirium PreventionHatta et al. (Meta-analysis, 2023)Meta-Analysis (8 RCTs)Hospitalized patients (n=587)Ramelteon vs. PlaceboIncidence of deliriumRamelteon associated with 50% lower odds of delirium (OR 0.50, 95% CI 0.29-0.86).16
Delirium PreventionNCT050694284ICU patientsRamelteon vs. PlaceboDelirium reduction (CAM-ICU-7 score)Ongoing trial to assess efficacy in prevention and treatment of ICU delirium.35
Bipolar Disorder (Maintenance)Norris et al. (Ram-TIME)4Euthymic bipolar I patients (n=83)Adjunctive Ramelteon 8 mg vs. PlaceboTime to mood relapseRamelteon reduced relapse risk by ~50% (OR 0.48, p=0.024) over 24 weeks.37
Bipolar Disorder (Maintenance)NCT016851513Bipolar I patientsRamelteon SL (0.1, 0.4, 0.8 mg) vs. PlaceboTime to mood relapseStudy terminated early for futility; no significant difference between Ramelteon and placebo.40
Delayed Sleep Phase SyndromeNCT005937362Adults with DSPSRamelteon vs. PlaceboAdvancement of sleep timingProof-of-concept study; development for this indication was later discontinued.36

Section 5: Safety, Tolerability, and Risk Management

The safety profile of Ramelteon is its most compelling clinical attribute and the primary feature that distinguishes it from all other classes of hypnotic medications. Its tolerability is excellent, and it is devoid of the risks of abuse and dependence that plague traditional sleep aids.

5.1. Adverse Reaction Profile

In clinical trials, Ramelteon was generally well-tolerated. The most common adverse reactions reported were mild and occurred at rates only slightly higher than placebo.[1] The adverse events occurring in at least 3% of patients and more frequently than with placebo were somnolence (3% for Ramelteon vs. 2% for placebo), dizziness (4% vs. 3%), fatigue (3% vs. 2%), and nausea (3% vs. 2%).[1] Overall, side effects led to discontinuation of the drug in 1% or fewer people, a rate comparable to placebo.[1]

A specific area of attention is Ramelteon's effect on endocrine function, a predictable consequence of its action on the melatonin system, which is linked to the hypothalamic-pituitary-gonadal axis. Clinical studies have found that Ramelteon can cause a slight increase in prolactin levels in women (+34%) and a modest decrease in free testosterone levels in men (ranging from 3% to 18% depending on age).[1] While often subclinical, these hormonal changes can manifest as symptoms such as decreased libido, amenorrhea, galactorrhea, or problems with fertility. The FDA label advises that assessment of prolactin and testosterone levels should be considered for patients who present with such unexplained symptoms.[43]

5.2. Warnings, Precautions, and Contraindications

Despite its favorable overall safety profile, the use of Ramelteon carries several important warnings and precautions, some of which are class-wide labels for all hypnotic drugs.

  • Severe Allergic Reactions: Rare but potentially life-threatening cases of angioedema (swelling of the tongue, glottis, or larynx) and anaphylaxis have been reported following the first or subsequent doses of Ramelteon. These reactions can cause airway obstruction and may be fatal. Any patient who develops angioedema after treatment should not be rechallenged with the drug.[13]
  • Abnormal Thinking and Behavioral Changes: In association with the use of hypnotics, a variety of cognitive and behavioral changes have been reported. For Ramelteon, these may include hallucinations, bizarre behavior, and agitation.[13] In patients with pre-existing depression, the use of hypnotics has been associated with a worsening of depression, including suicidal ideation and completed suicides.[1] Any new or worsening behavioral change requires immediate evaluation.
  • Complex Sleep Behaviors: A significant warning for the hypnotic class is the potential for complex sleep behaviors. These are activities performed while not fully awake, such as "sleep-driving," preparing and eating food, making phone calls, or having sex, often with amnesia for the event.[13] The risk for these behaviors is increased by the concomitant use of alcohol or other CNS depressants. Discontinuation of Ramelteon should be strongly considered for any patient who reports such an event. While this is a required class warning, the mechanistic basis for these behaviors is thought to be a state of incomplete arousal and dissociated consciousness characteristic of profound GABAergic CNS depression. Given that Ramelteon does not act on the GABA system and does not cause global CNS depression or significantly alter sleep architecture, it is mechanistically less likely to induce these parasomnias compared to agents like zolpidem.[24] The reports are likely very rare, and the warning may represent a broad regulatory requirement for the hypnotic class rather than a specific, high-frequency risk attributable to Ramelteon's mechanism.
  • Contraindications and Precautions: The most critical contraindication is the concomitant use of fluvoxamine, a strong CYP1A2 inhibitor, due to the risk of a massive increase in Ramelteon exposure.[13] A history of angioedema to Ramelteon is also an absolute contraindication.[13] Caution is advised in patients with moderate hepatic impairment, and the drug is not recommended for use in patients with severe hepatic impairment or severe obstructive sleep apnea.[28]

5.3. Abuse Potential and Dependence: A Paradigm Shift in Hypnotic Safety

The most significant safety differentiator for Ramelteon is its complete lack of abuse potential and physical dependence.[1] This feature is a direct result of its selective, non-GABAergic mechanism of action and has been rigorously demonstrated in a comprehensive series of preclinical and clinical studies.

Human abuse potential studies were conducted in populations of recreational polydrug abusers, the gold standard for assessing a drug's rewarding effects. In a double-blind, crossover study comparing Ramelteon (at therapeutic and supratherapeutic doses up to 160 mg, or 20 times the recommended dose), the benzodiazepine triazolam (a positive control), and placebo, the results were definitive. Triazolam produced clear, dose-dependent effects on measures of "drug liking" and subjective feelings consistent with its abuse liability. In stark contrast, Ramelteon at all doses was indistinguishable from placebo on all measures related to abuse potential. In fact, 79% of these experienced drug users identified the highest dose of Ramelteon as a placebo.[6]

These findings are corroborated by preclinical data. In animal models, monkeys trained to self-administer benzodiazepines like midazolam or diazepam did not recognize or self-administer Ramelteon, indicating it does not share the same subjective rewarding effects.[45] Furthermore, in monkeys treated with Ramelteon for one year, abrupt discontinuation did not produce any of the 33 signs of physical withdrawal (e.g., tremors, teeth grinding) typically seen with benzodiazepine withdrawal.[45] Consistent with this, clinical trials in humans have shown no evidence of rebound insomnia or withdrawal symptoms upon cessation of Ramelteon, even after a full year of continuous use.[1]

As a direct result of this robust and consistent body of evidence, the U.S. Drug Enforcement Agency (DEA) has not classified Ramelteon as a controlled substance. This makes it unique among all major prescription hypnotic drugs and establishes it as the safest option from the perspective of abuse and dependence.[1]

Section 6: Clinically Significant Drug-Drug Interactions

The pharmacokinetic profile of Ramelteon, particularly its heavy reliance on CYP450 enzymes for its extensive first-pass metabolism, makes it highly susceptible to clinically significant drug-drug interactions. A thorough understanding and screening for these interactions is a critical component of its safe clinical use.

6.1. Interactions via CYP450 Inhibition

Inhibitors of the primary metabolizing enzymes for Ramelteon can dramatically increase its systemic exposure, potentially leading to exaggerated effects or toxicity.

  • Fluvoxamine (Strong CYP1A2 Inhibitor): This interaction is the most critical and severe. Fluvoxamine is a potent inhibitor of CYP1A2, the primary enzyme responsible for Ramelteon's clearance. Co-administration results in a massive increase in Ramelteon exposure, with the area under the curve (AUC) increasing by approximately 190-fold and the peak concentration (Cmax) increasing by approximately 70-fold.[13] This interaction effectively turns a standard therapeutic dose into a massive overdose. Consequently, the concomitant use of Ramelteon and fluvoxamine is strictly contraindicated.[11]
  • Fluconazole (Strong CYP2C9 Inhibitor): Fluconazole is a strong inhibitor of CYP2C9, an enzyme that contributes to the secondary metabolic pathway of Ramelteon. Co-administration increases the systemic exposure (AUC) of Ramelteon by approximately 150%.[11] Due to this substantial increase, the FDA label advises that Ramelteon be administered with caution when used concomitantly with fluconazole.[28]
  • Ketoconazole (Strong CYP3A4 Inhibitor): Ketoconazole is a potent inhibitor of CYP3A4, the tertiary metabolic pathway for Ramelteon. This interaction leads to an increase in Ramelteon AUC by approximately 84% and Cmax by 36%.[11] While less dramatic than the interactions with CYP1A2 or CYP2C9 inhibitors, this is still a clinically meaningful increase, and caution is advised when these drugs are used together.[11]
  • Other Interactions: Other medications that are less potent inhibitors of these pathways can also affect Ramelteon exposure. For example, co-administration with donepezil or doxepin has been shown to increase Ramelteon AUC by approximately 100% and 66%, respectively. Close monitoring of the patient is recommended when these drugs are used together.[13]

6.2. Interactions via CYP450 Induction

Inducers of CYP450 enzymes can have the opposite effect, accelerating the metabolism of Ramelteon and reducing its systemic exposure, which can lead to a loss of therapeutic efficacy.

  • Rifampin (Strong CYP Inducer): Rifampin is a potent inducer of multiple CYP enzymes, including CYP1A2, CYP2C, and CYP3A4. Co-administration with Ramelteon leads to a profound decrease in the exposure of both Ramelteon and its active metabolite M-II by approximately 80%.[11] This substantial reduction is likely to render Ramelteon clinically ineffective. Therefore, concomitant use may result in a loss of efficacy and should be considered when evaluating treatment response.[11]

6.3. Other Interactions

  • Alcohol: As with all CNS-active medications, alcohol can have additive effects. The combination of alcohol and Ramelteon results in additive psychomotor impairment. Patients should be advised not to consume alcohol in combination with Ramelteon.[13]
  • Lack of Interaction: Importantly, Ramelteon has been shown not to have clinically meaningful pharmacokinetic interactions with a number of commonly used drugs, including the SSRI fluoxetine, the proton-pump inhibitor omeprazole, and the cardiovascular drugs digoxin and warfarin.[11] The lack of interaction with warfarin, a drug with a narrow therapeutic index metabolized by CYP2C9, is a notable safety advantage.

Table 4: Clinically Significant Drug-Drug Interactions with Ramelteon

Interacting Drug / ClassMechanism of InteractionPharmacokinetic ConsequenceClinical Management RecommendationSource(s)
FluvoxamineStrong CYP1A2 inhibitor↑ Ramelteon AUC ~190-fold, ↑ Cmax ~70-foldCONTRAINDICATED. Do not use together under any circumstances.13
RifampinStrong CYP enzyme inducer↓ Ramelteon and M-II AUC ~80%Avoid concomitant use. Efficacy of Ramelteon will be substantially reduced or eliminated.11
FluconazoleStrong CYP2C9 inhibitor↑ Ramelteon AUC ~150%Use with caution. Monitor for increased Ramelteon effects.11
KetoconazoleStrong CYP3A4 inhibitor↑ Ramelteon AUC ~84%, ↑ Cmax ~36%Use with caution. Monitor for increased Ramelteon effects.11
DonepezilCYP2D6 and CYP3A4 inhibitor↑ Ramelteon AUC ~100%, ↑ Cmax ~87%Use with caution. Patients should be closely monitored.13
DoxepinCYP2D6 inhibitor↑ Ramelteon AUC ~66%, ↑ Cmax ~69%Use with caution. Patients should be closely monitored.13
AlcoholPharmacodynamic interactionAdditive psychomotor impairmentAvoid concomitant use.13
Warfarin, Digoxin, Fluoxetine, MidazolamNo significant interactionNo clinically meaningful change in PKNo dose adjustment or special monitoring required.11

Section 7: Comparative Analysis and Place in Therapy

The clinical value of Ramelteon is best understood when it is positioned relative to other available hypnotic agents. Its introduction, along with that of the dual orexin receptor antagonists (DORAs), has fundamentally reshaped the therapeutic landscape of insomnia, creating a tiered, risk-stratified approach to treatment that did not exist when GABAergic agents were the only option. This modern paradigm allows for a more personalized selection of therapy based on a patient's specific clinical needs, comorbidities, and risk profile.

7.1. Ramelteon vs. Benzodiazepines (e.g., Temazepam) and Z-Drugs (e.g., Zolpidem)

Benzodiazepine receptor agonists (BZRAs), which include both the classic benzodiazepines and the nonbenzodiazepine "Z-drugs" (zolpidem, zaleplon, eszopiclone), have historically been the mainstay of insomnia treatment.

  • Efficacy: In general, BZRAs are considered more potent and efficacious hypnotics than Ramelteon, particularly for sleep maintenance insomnia.[14] They produce more robust and consistent reductions in sleep latency and increases in total sleep time. However, some head-to-head evidence suggests a more comparable profile for sleep onset; one study found that Ramelteon 8 mg was as efficacious as zolpidem 10 mg in reducing subjective sleep latency over a 14-day period.[48]
  • Safety and Tolerability: This is where Ramelteon holds a decisive advantage. BZRAs act via global CNS depression and are associated with a significantly higher risk of next-day residual effects, including cognitive and motor impairment, dizziness, and an increased risk of falls, particularly in elderly populations.[15] A study directly comparing the effects of Ramelteon and zolpidem on middle-of-the-night balance and mobility in older adults found that zolpidem caused significant impairment, whereas Ramelteon was no different from placebo.[15]
  • Abuse and Dependence: This is the most critical differentiator. Benzodiazepines and Z-drugs are all classified as Schedule IV controlled substances by the DEA due to their well-documented potential for misuse, abuse, tolerance, and physical dependence, which can lead to significant withdrawal symptoms upon discontinuation.[14] Ramelteon, by virtue of its non-GABAergic mechanism, is devoid of these risks and is not a controlled substance.[3]

7.2. Ramelteon vs. Orexin Receptor Antagonists (DORAs; e.g., Suvorexant, Lemborexant)

DORAs represent another novel class of hypnotics that, like Ramelteon, do not target the GABA system. They work by blocking the wake-promoting signals of the orexin neuropeptide system.

  • Efficacy: The available evidence, including network meta-analyses, suggests that DORAs are more effective than Ramelteon for improving both sleep onset and sleep maintenance.[14] For example, one analysis ranked lemborexant as the most effective treatment for several objective sleep measures.[49]
  • Safety and Mechanism: Both classes are considered to have superior safety profiles compared to BZRAs.[14] However, a key distinction remains: DORAs are classified as Schedule IV controlled substances, indicating that they carry some risk of abuse or dependence, whereas Ramelteon does not.[46] Their mechanisms are also fundamentally different: Ramelteon actively promotes the physiological sleep signal via the melatonin system, while DORAs passively permit sleep by blocking a primary wakefulness signal.

7.3. Ramelteon vs. Eszopiclone

Eszopiclone is a Z-drug that is also approved for long-term use. Comparative data indicates that eszopiclone is more effective than Ramelteon, particularly for long-term treatment.[47] However, this greater efficacy comes at the cost of a less favorable side effect profile. Eszopiclone is associated with more adverse events, including a potential increased risk of infections, and patients are more likely to discontinue it due to side effects compared to Ramelteon.[47]

7.4. Place in Therapy

The evidence collectively positions Ramelteon within a specific therapeutic niche. The 2017 AASM clinical practice guidelines provide a "weak" recommendation for the use of Ramelteon for sleep-onset insomnia, acknowledging its favorable safety profile but small magnitude of benefit.[1] In contrast, some European guidelines do not recommend it at all due to concerns about its modest efficacy.[51]

Its primary and most appropriate place in therapy is as a first-line pharmacological agent for patients in whom safety is the paramount concern. This includes:

  1. Patients with a history of or active substance use disorder, for whom the non-controlled status and lack of abuse potential are critical advantages.
  2. Elderly patients, who are particularly vulnerable to the cognitive impairment, falls, and fractures associated with BZRAs.
  3. Patients with concerns about dependence or who require long-term treatment without the risk of developing tolerance or withdrawal.
  4. As a potential bridging therapy to facilitate the tapering and discontinuation of long-term BZRA use.[42]

Ramelteon's introduction created the "safety-first" tier in the modern, stratified treatment of insomnia. The clinical decision is no longer a simple choice between different BZRAs, but a nuanced assessment of the patient's risk profile. For a patient where safety is the priority, Ramelteon is an excellent choice. For a patient needing more potent effects on sleep maintenance without a history of substance abuse, a DORA may be considered. For acute, short-term insomnia requiring powerful sedation, a BZRA might still be used cautiously. Ramelteon's value, therefore, is not in being the most powerful hypnotic, but in being the anchor of the safest tier in this new therapeutic paradigm.

Table 5: Comparative Profile of Major Hypnotic Agent Classes

FeatureRamelteonZ-Drugs (e.g., Zolpidem)Benzodiazepines (e.g., Temazepam)DORAs (e.g., Suvorexant)
Mechanism of ActionSelective MT1/MT2 Receptor Agonist (Chronobiotic)GABA-A Receptor Positive Allosteric Modulator (Sedative)GABA-A Receptor Positive Allosteric Modulator (Sedative/Anxiolytic)Dual Orexin Receptor Antagonist (Wake-Blocking)
Primary EfficacySleep OnsetSleep Onset & MaintenanceSleep Onset & MaintenanceSleep Onset & Maintenance
Abuse/Dependence RiskNoneYesYesYes (Lower than BZRAs)
DEA ScheduleNot a Controlled SubstanceSchedule IVSchedule IVSchedule IV
Next-Day ImpairmentMinimal / NoneModerateModerate to HighLow to Moderate
Rebound InsomniaNoYesYesMinimal
Key Adverse EffectsSomnolence, dizziness, endocrine changes (prolactin, testosterone)Drowsiness, dizziness, amnesia, complex sleep behaviorsDrowsiness, cognitive impairment, ataxia, amnesiaSomnolence, headache, abnormal dreams, sleep paralysis
Source(s)1141414

Section 8: Conclusion and Future Directions

8.1. Synthesis of Ramelteon's Clinical Profile

Ramelteon occupies a unique and important position in the pharmacotherapy of sleep disorders. Its clinical profile is defined by a central trade-off: it offers an unparalleled safety profile, rooted in its novel and highly selective chronobiotic mechanism, which is balanced by a clinical efficacy for its primary indication that is statistically significant but modest in magnitude. Its value as a therapeutic agent is defined as much by the significant risks it avoids—abuse, dependence, withdrawal, and major next-day impairment—as by the sleep-promoting benefits it provides. Unlike any other prescription hypnotic, its action is not one of forced sedation but of gentle facilitation of the body's natural sleep-wake cycle. This makes it a fundamentally different tool, best suited for clinical scenarios where safety and long-term tolerability are the highest priorities.

8.2. Recommendations for Clinical Practice

Based on the comprehensive evidence, the following recommendations are prudent for the clinical use of Ramelteon:

  1. Prioritize Patient Selection: Ramelteon is best reserved for patients with sleep-onset insomnia for whom safety is the primary concern. Ideal candidates include the elderly, patients with a history of substance use disorder, and those wishing to avoid the dependence potential of controlled substances.
  2. Manage Efficacy Expectations: Clinicians must educate patients that the effect of Ramelteon is subtle and not a powerful, immediate sedation. Its purpose is to help regulate the sleep cycle over time. This counseling is crucial to ensure patient adherence and prevent perceived treatment failure.
  3. Mandatory Drug Interaction Screening: Before prescribing, a thorough review of the patient's concomitant medications is essential. The contraindication with fluvoxamine is absolute. Caution and potential dose adjustment or heightened monitoring are required when co-administering with other potent inhibitors of CYP1A2, CYP2C9, or CYP3A4.
  4. Adhere to Administration Guidelines: Patients should be instructed to take the 8 mg dose within 30 minutes of bedtime and not with or immediately after a high-fat meal to ensure optimal absorption and onset of action.

8.3. Future Directions

The evolution of Ramelteon has revealed new avenues where its unique properties may be of even greater clinical value than in its primary indication. Future research should focus on several key areas:

  • Delirium Prevention: This is the most compelling future direction for Ramelteon. The strong positive signal from multiple meta-analyses warrants confirmation in large, prospective, well-designed RCTs. If these trials are successful, Ramelteon could become the first evidence-based, standard-of-care pharmacological agent for the prevention of delirium in high-risk hospitalized populations, addressing a major unmet medical need.
  • Benzodiazepine Receptor Agonist (BZRA) Discontinuation: While some studies have explored this, more formal research is needed to establish protocols for using Ramelteon as an adjunctive agent to facilitate tapering and discontinuation of long-term BZRA use. Its excellent safety profile makes it an ideal candidate to help patients transition off drugs with significant dependence liability.
  • Pediatric Use: Ramelteon has not been formally studied in children or adolescents.[26] Given the significant need for safe and effective sleep aids in certain pediatric populations (e.g., those with neurodevelopmental disorders), well-controlled clinical trials to establish its safety and efficacy in this age group would be a valuable contribution.

Works cited

  1. Ramelteon - Wikipedia, accessed August 7, 2025, https://en.wikipedia.org/wiki/Ramelteon
  2. Ramelteon | 196597-26-9 - ChemicalBook, accessed August 7, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB0496858.htm
  3. Ramelteon: A Novel Hypnotic Indicated for the Treatment of Insomnia - PMC, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2880941/
  4. What is the mechanism of Ramelteon? - Patsnap Synapse, accessed August 7, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-ramelteon
  5. Ramelteon: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed August 7, 2025, https://go.drugbank.com/drugs/DB00980
  6. Ramelteon demonstrates no abuse potential according to study results - EurekAlert!, accessed August 7, 2025, https://www.eurekalert.org/news-releases/593674
  7. Ramelteon: a novel hypnotic lacking abuse liability and sedative adverse effects - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/17015817/
  8. Rozerem (ramelteon) FDA Approval History - Drugs.com, accessed August 7, 2025, https://www.drugs.com/history/rozerem.html
  9. Evaluation of the Efficacy and Safety of Ramelteon in Subjects with Chronic Insomnia, accessed August 7, 2025, https://jcsm.aasm.org/doi/abs/10.5664/jcsm.26914
  10. Evaluation of the Efficacy and Safety of Ramelteon in Subjects with Chronic Insomnia, accessed August 7, 2025, https://jcsm.aasm.org/doi/10.5664/jcsm.26914
  11. Ramelteon Monograph for Professionals - Drugs.com, accessed August 7, 2025, https://www.drugs.com/monograph/ramelteon.html
  12. NDA 21-782 Page 4 ROZEREM™ (ramelteon) tablets DESCRIPTION ROZEREM™ (ramelteon) is an orally active hypnotic chemically desi - accessdata.fda.gov, accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2005/021782lbl.pdf
  13. This label may not be the latest approved by FDA. For current ..., accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/021782s021lbl.pdf
  14. A retrospective clinical practice study comparing the usefulness of dual-orexin receptor antagonists and a melatonin receptor agonist in patients switching from long-term benzodiazepine receptor agonists - PMC, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10985311/
  15. Comparative Study of Efficacy and Safety of Ramelteon vs Zolpidem in Patients of Insomnia in Tertiary Care Centre of East India - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/369137677_Comparative_Study_of_Efficacy_and_Safety_of_Ramelteon_vs_Zolpidem_in_Patients_of_Insomnia_in_Tertiary_Care_Centre_of_East_India
  16. Ramelteon for delirium prevention in hospitalized patients: An ..., accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/36726202/
  17. Ramelteon | CAS 196597-26-9 | SCBT - Santa Cruz Biotechnology, accessed August 7, 2025, https://www.scbt.com/p/ramelteon-196597-26-9
  18. Ramelteon | TAK-375 | CAS#196597-26-9 | melatonin receptor agonist | MedKoo, accessed August 7, 2025, https://www.medkoo.com/products/7753
  19. 196597-26-9・Ramelteon・184-03371・180-03373[Detail Information] | [Life Science] - FUJIFILM Wako Chemicals, accessed August 7, 2025, https://labchem-wako.fujifilm.com/us/product/detail/W01W0118-0337.html
  20. Ramelteon – A Melatonin Receptor Agonist in the Treatment of Insomnia - touchNEUROLOGY, accessed August 7, 2025, https://www.touchneurology.com/wp-content/uploads/sites/3/2015/06/private_articles_8222_pdf_levien.pdf
  21. Ramelteon 196597-26-9 | Tokyo Chemical Industry Co., Ltd.(APAC), accessed August 7, 2025, https://www.tcichemicals.com/OP/en/p/R0216
  22. Ramelteon (TAK-375, CAS Number: 196597-26-9) | Cayman Chemical, accessed August 7, 2025, https://www.caymanchem.com/product/20389/ramelteon
  23. Clinical Pharmacology Biopharmaceutics Review(s) - accessdata.fda.gov, accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2005/021782s000_Rozerem_biopharmr.pdf
  24. How does Ramelteon (rozerem) work and why is it not habit-forming? - Dr.Oracle, accessed August 7, 2025, https://www.droracle.ai/articles/124011/how-does-ramelteon-works-why-its-not-habit-forming
  25. center for drug evaluation and - accessdata.fda.gov, accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2005/021782s000_Rozerem_prntlbl.pdf
  26. Ramelteon (Rozerem) for Insomnia - AAFP, accessed August 7, 2025, https://www.aafp.org/pubs/afp/issues/2006/0415/p1437.html
  27. Metabolism of ramelteon in human liver microsomes and correlation with the effect of fluvoxamine on ramelteon pharmacokinetics - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/20478852/
  28. NDA 21-782/S-008, S-009, S-010 Page 5 NDA 21-782 ROZEREM (ramelteon) Takeda Global Research and Development Center, Inc. One T - accessdata.fda.gov, accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021782s008s009s010lbl.pdf
  29. FDA Final Labeling 21782/Supp 011 11.9.10, accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021782s011lbl.pdf
  30. Ramelteon Completed Phase 3 Trials for Disorders of Initiating and Maintaining Sleep Treatment - DrugBank, accessed August 7, 2025, https://go.drugbank.com/drugs/DB00980/clinical_trials?conditions=DBCOND0079328&phase=3&purpose=treatment&status=completed
  31. Evaluation of the Efficacy and Safety of Ramelteon in Subjects with Chronic Insomnia - PMC, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC1978328/
  32. Preventative effects of ramelteon against postoperative delirium after elective liver resection, accessed August 7, 2025, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0241673
  33. The Efficacy of Ramelteon to Prevent Postoperative Delirium After General Anesthesia in the Elderly: A Double-Blind, Randomized, Placebo-Controlled Trial | Request PDF - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/372614388_The_Efficacy_of_Ramelteon_to_Prevent_Postoperative_Delirium_After_General_Anesthesia_in_the_Elderly_A_Double-Blind_Randomized_Placebo-Controlled_Trial
  34. Suvorexant with or without ramelteon to prevent delirium: a systematic review and meta‐analysis - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/356899038_Suvorexant_with_or_without_ramelteon_to_prevent_delirium_a_systematic_review_and_meta-analysis
  35. Study Details | Delirium Reduction With Ramelteon - ClinicalTrials.gov, accessed August 7, 2025, https://www.clinicaltrials.gov/study/NCT05069428
  36. Study Details | Efficacy of Ramelteon on Speeding Up Sleep in Subjects With Delayed Sleep Phase Syndrome | ClinicalTrials.gov, accessed August 7, 2025, https://www.clinicaltrials.gov/study/NCT00593736?intr=ROZEREM&aggFilters=studyType:int&viewType=Table&rank=8
  37. A double-blind, randomized, placebo-controlled trial of adjunctive ..., accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/22963894/
  38. Study Details | Ramelteon for the Treatment of Insomnia and Mood Stability in Patients With Euthymic Bipolar Disorder | ClinicalTrials.gov, accessed August 7, 2025, https://clinicaltrials.gov/study/NCT00552760
  39. Safety and Efficacy Study of Ramelteon (TAK-375) Tablets for Sublingual Administration (SL) in Adults With Bipolar 1 Disorder | ClinicalTrials.gov, accessed August 7, 2025, https://clinicaltrials.gov/study/NCT01677182?term=NCT01467713%20NCT01677182%20NCT01685151&rank=2
  40. Efficacy and safety of sublingual ramelteon as an adjunctive therapy in the maintenance treatment of bipolar I disorder in adults: A phase 3, randomized controlled trial - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/28662460/
  41. A Safety and Efficacy Study of Ramelteon Tablets for Sublingual Administration (TAK-375SL) in the Maintenance Treatment of Bipolar 1 Disorder | ClinicalTrials.gov, accessed August 7, 2025, https://clinicaltrials.gov/study/NCT01685151
  42. Ramelteon Completed Phase 4 Trials for Insomnia Chronic Treatment | DrugBank Online, accessed August 7, 2025, https://go.drugbank.com/drugs/DB00980/clinical_trials?conditions=DBCOND0080650&phase=4&purpose=treatment&status=completed
  43. Medication Guide - accessdata.fda.gov, accessed August 7, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021782s011Medguide.pdf
  44. The mechanism of zolpidem and ramelteon to treat insomnia - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/389413988_The_mechanism_of_zolpidem_and_ramelteon_to_treat_insomnia
  45. New sleep medication shows less potential to foster abuse and dependence, accessed August 7, 2025, https://www.apa.org/news/press/releases/2006/06/sleep-medication
  46. What are the strongest sleeping pills? - Drugs.com, accessed August 7, 2025, https://www.drugs.com/medical-answers/what-strongest-sleeping-pills-3578510/
  47. Comparative effects of pharmacological interventions for the acute and long-term management of insomnia disorder in adults: a systematic review and network meta-analysis - PubMed, accessed August 7, 2025, https://pubmed.ncbi.nlm.nih.gov/35843245/
  48. Comparative Study of Efficacy and Safety of Ramelteon vs Zolpidem ..., accessed August 7, 2025, https://globalresearchonline.net/ijpsrr/v78-2/23.pdf
  49. Comparative efficacy of lemborexant and other insomnia treatments: a network meta-analysis - PMC, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10394202/
  50. Meta-Analyses of Hypnotics and Infections: Eszopiclone, Ramelteon, Zaleplon, and Zolpidem - PMC, accessed August 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2725260/
  51. Safety of dual orexin receptor antagonists: a real-world pharmacovigilance study | Request PDF - ResearchGate, accessed August 7, 2025, https://www.researchgate.net/publication/389255975_Safety_of_dual_orexin_receptor_antagonists_a_real-world_pharmacovigilance_study
  52. The efficacy of add-on ramelteon and subsequent dose reduction in benzodiazepine derivatives/Z-drugs for the treatment of sleep-related eating disorder and night eating syndrome: a retrospective analysis of consecutive patients, accessed August 7, 2025, https://jcsm.aasm.org/doi/10.5664/jcsm.9236

Published at: August 7, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.