C24H36O3
51022-71-0
Refractory Chemotherapy-Induced Nausea and Vomiting (CINV)
Nabilone is an orally active, synthetic cannabinoid and a structural analog of delta-9-tetrahydrocannabinol (Δ⁹-THC), the principal psychoactive constituent of cannabis. Identified by DrugBank ID DB00486 and CAS Number 51022-71-0, Nabilone functions as a partial agonist at both cannabinoid type 1 (CB1) and type 2 (CB2) receptors within the body's endocannabinoid system. Its therapeutic effects, particularly its antiemetic properties, are primarily mediated through its action on CB1 receptors in the central nervous system.
The United States Food and Drug Administration (FDA) has approved Nabilone for a narrow indication: the treatment of nausea and vomiting associated with cancer chemotherapy in patients who have failed to respond adequately to conventional antiemetic therapies. This positions Nabilone as a second-line or salvage agent in oncology support. Despite this limited approval, the drug is subject to extensive off-label use and investigation for a range of intractable conditions, including chronic non-cancer pain, fibromyalgia, spasticity-related pain in neurological disorders, and the suppression of nightmares associated with post-traumatic stress disorder (PTSD).
The clinical utility of Nabilone is fundamentally defined by a delicate balance between its potential therapeutic benefits and a prominent profile of adverse effects. Its safety and tolerability are dominated by dose-limiting central nervous system effects, including drowsiness, vertigo, euphoria, and potential psychiatric disturbances such as hallucinations and psychosis, which can persist for up to 72 hours after administration. Due to its psychoactive properties and potential for psychological dependence, Nabilone is classified as a Schedule II controlled substance in the United States and Canada, necessitating stringent prescribing and monitoring protocols.
Comparatively, Nabilone is more potent than dronabinol (synthetic THC) and possesses a distinct pharmacokinetic profile characterized by a longer duration of action. Its development history is unique, having been initially approved in 1985, withdrawn from the US market, and subsequently re-launched in 2006, reflecting a renewed interest in cannabinoid-based therapeutics. Current research continues to explore its potential in novel areas, such as managing agitation in Alzheimer's disease and aggression in developmental disabilities, underscoring its evolution from a niche antiemetic to a broad investigational tool for CNS disorders.
The precise identification of a pharmaceutical agent is foundational to its study and clinical use. Nabilone is a well-characterized small molecule with a comprehensive set of identifiers across various chemical, pharmacological, and regulatory databases.
Nabilone is known by several names and codes that facilitate its tracking in scientific literature and clinical practice. The primary brand name for Nabilone in the United States, Canada, the United Kingdom, and Mexico is Cesamet.[1] In Canada, multiple generic versions are also available, including ACT Nabilone, APO-Nabilone, pms-Nabilone, RAN-Nabilone, and TEVA-Nabilone.[3] The brand name Canemes is used in Austria and Germany.[4]
Historically and in research contexts, it has been referred to by synonyms such as Nabilon, Nabilona, Nabilonum, and the developmental codes Cpd 109514 and LY 109514.[5] The extensive catalog of identifiers from databases like ChEMBL (CHEMBL947), the Human Metabolome Database (HMDB0014629), and KEGG (D05099) reflects a long history of scientific investigation predating its modern clinical applications.[2]
Nabilone is a dibenzopyrane-derived synthetic cannabinoid.[6] As a raw material, it is described as a white to off-white polymorphic crystalline powder.[9] Its chemical structure is similar to that of Δ⁹-THC but is synthetically distinct.[2] The key physicochemical properties are summarized in Table 1. Its poor aqueous solubility (less than 0.5 mg/L across a pH range of 1.2 to 7.0) is a critical factor influencing its oral formulation and absorption characteristics.[10]
Table 1: Key Identifiers and Physicochemical Properties of Nabilone
Property | Value | Source(s) |
---|---|---|
DrugBank ID | DB00486 | 1 |
CAS Number | 51022-71-0 | 2 |
Chemical Formula | C24H36O3 | 1 |
Molecular Weight | 372.54 g/mol | 8 |
US Brand Name | Cesamet | 1 |
DEA Code Number | 7379 | 2 |
UNII | 2N4O9L084N | 2 |
Melting Point | 159.0 °C | 8 |
Boiling Point | 457.4 °C | 8 |
Solubility (Aqueous) | < 0.5 mg/L (pH 1.2-7.0) | 10 |
Storage | 2°C - 8°C | 8 |
Nabilone's therapeutic and adverse effects are a direct consequence of its interaction with the endogenous cannabinoid system. Its profile is characterized by high potency, complex central nervous system activity, and a pharmacokinetic profile that leads to a prolonged duration of action.
Nabilone exerts its pharmacological effects by acting on the endocannabinoid system (ECS), a crucial neuromodulatory system involved in regulating a wide array of physiological processes, including pain perception, memory, mood, appetite, and emesis.[2] The primary mechanism of action for Nabilone is its activity as a partial agonist at both cannabinoid type 1 (CB1) and type 2 (CB2) receptors, which are G protein-coupled receptors.[5]
Although structurally distinct from Δ⁹-THC, Nabilone mimics its pharmacological activity and is considered to be approximately twice as potent.[2]
In vitro studies have demonstrated its high binding affinity for both cannabinoid receptors, with dissociation constants (Ki) of 2.2 nM for CB1 and 1.8 nM for CB2.[8]
A critical nuance in Nabilone's pharmacodynamics is its classification as both highly potent and a "weak partial agonist".[2] This seemingly contradictory description highlights a key pharmacological principle. Its high potency means that it can elicit a response at very low concentrations, as reflected by its low
Ki values. However, as a partial agonist, it does not activate the receptor to its full capacity, unlike a full agonist. This may result in a "ceiling effect," where increasing the dose beyond a certain point does not produce a greater maximal response. This property could theoretically offer a safety advantage over full agonists by limiting maximal psychoactivity, but this is counterbalanced by its high potency, which means significant CNS effects occur even at therapeutic doses.
The complex effects of Nabilone on the CNS are dose-dependent and include the modulation of neurotransmitter systems, such as serotonin and dopamine, which contributes to its therapeutic actions and its side-effect profile.[9] These effects manifest as changes in mood (e.g., euphoria, depression, anxiety), decrements in cognitive performance and memory, and alterations in perception.[9]
The absorption, distribution, metabolism, and excretion (ADME) profile of Nabilone is crucial for understanding its clinical use, particularly its dosing schedule and the prolonged duration of its effects. The key pharmacokinetic parameters are summarized in Table 2.
This pronounced mismatch between the short half-life of the parent drug and the long half-life of its metabolites is of profound clinical importance. It explains why the central nervous system effects of Nabilone, both therapeutic and adverse, can persist for a variable and unpredictable period, often lasting 48 to 72 hours after the final dose.[11] This pharmacokinetic characteristic necessitates careful patient counseling regarding activities that require mental alertness and underscores the importance of supervision during initial therapy.
Table 2: Summary of Nabilone Pharmacokinetic Parameters
Parameter | Value | Clinical Implication | Source(s) |
---|---|---|---|
Bioavailability | Low (10-20%) | Significant first-pass metabolism limits systemic exposure from an oral dose. | 15 |
Time to Peak (Tmax) | ~2 hours | Onset of action is not immediate. | 5 |
Volume of Distribution (Vd) | ~12.5 L/kg | Extensive tissue distribution, including CNS penetration; accumulation in fat. | 5 |
Protein Binding | ~97% | High potential for displacement interactions with other protein-bound drugs. | 15 |
Metabolism | Hepatic (CYP2E1, 3A4, 2C8, 2C9) | Extensive metabolism; potential for drug-drug interactions. | 15 |
Elimination Route | Fecal (~60-67%), Renal (~22-24%) | Primarily eliminated via the biliary system. | 5 |
Half-life (Parent Drug) | ~2 hours | Parent drug is cleared from plasma relatively quickly. | 5 |
Half-life (Metabolites) | ~35 hours | Long-lasting metabolites are responsible for the prolonged clinical effects (48-72 hours). | 5 |
Nabilone's clinical profile is characterized by a single, narrowly defined approved indication, which stands in stark contrast to a broad and expanding landscape of off-label and investigational uses. This divergence reflects the drug's powerful, pleiotropic effects on the CNS, which clinicians have sought to harness for various treatment-refractory conditions.
The sole indication for which Nabilone has received approval from the U.S. FDA and Health Canada is the treatment of nausea and vomiting associated with cancer chemotherapy in patients who have failed to respond adequately to conventional antiemetic treatments.[2] It is explicitly designated as a second-line or salvage therapy and is not intended for first-line or as-needed (PRN) use.[3]
This indication is a legacy of its initial development in an era before the advent of modern, highly effective antiemetics such as the serotonin 5-HT3 receptor antagonists (e.g., ondansetron) and neurokinin-1 (NK1) receptor antagonists.[20] Clinical trials from that period demonstrated that Nabilone could produce a significant reduction in the severity and duration of nausea and vomiting in approximately 50% to 70% of patients with severe symptoms refractory to then-conventional therapies like prochlorperazine.[20] In contemporary practice, its use is often governed by stringent criteria, with some guidelines and insurance policies requiring documented failure of a 5-HT3 antagonist and at least two other classes of antiemetics before Nabilone can be considered.[15]
The majority of current clinical interest in Nabilone lies outside its approved indication, focusing on its potential utility in neurology and psychiatry for conditions that often lack effective treatments. This has led to a pattern of pharmacological repositioning, where an older drug is explored for entirely new therapeutic purposes.
The off-label management of chronic non-cancer pain is one of the most common uses for Nabilone.[22] Conditions treated include neuropathic pain, fibromyalgia, and musculoskeletal pain.[1] However, the clinical evidence supporting this practice is mixed and often of low methodological quality, creating a significant gap between clinical practice and robust scientific validation.
Several retrospective chart reviews and small studies have suggested potential benefits. A review of 20 patients with chronic non-cancer pain treated with Nabilone for an average of 1.5 years found that 15 (75%) reported subjective overall improvement, with beneficial effects on sleep and nausea cited as primary reasons for continuing therapy.[26] A study in a pediatric chronic pain population found that 7 of 28 patients (25%) reported a slight improvement in pain symptoms.[25]
Conversely, systematic reviews and evidence-based guidelines have been more cautious. These analyses consistently highlight the mixed results and methodological limitations of the existing studies.[22] Consequently, recommendations from various organizations are conflicting, ranging from advising against the use of Nabilone for chronic pain to suggesting it may be considered as a third-line adjunctive therapy in select cases.[22] The uncertainty of the evidence suggests that the decision to use Nabilone for chronic pain is largely driven by clinical judgment and patient-reported outcomes in cases where conventional analgesics have failed.
A growing body of evidence suggests a promising role for Nabilone in managing treatment-resistant nightmares and insomnia associated with post-traumatic stress disorder (PTSD).[29] While the evidence is still emerging and largely derived from open-label trials, retrospective studies, and surveys, the findings have been notably consistent.
An early open-label chart review of 47 patients with PTSD found that 72% experienced either complete cessation or a significant reduction in nightmares when Nabilone was added to their existing treatment regimen.[31] More recently, an anonymous online survey of 60 active Canadian Armed Forces members with PTSD who were prescribed Nabilone for chronic nightmares found that 73% of those who continued the medication reported suppression of their nightmares.[30] Many respondents also reported ancillary benefits, such as improved sleep quality, fewer night sweats, and a reduction in daytime flashbacks.[34]
A key finding from these studies is that discontinuation of Nabilone often leads to a rapid recurrence of nightmares, typically within a week, suggesting that it provides a suppressive rather than a curative effect and may be required for long-term management.[29] Despite the promising results, the American Psychiatric Association has noted the methodological weaknesses of these studies and states that more compelling data is needed before Nabilone can be recommended as a standard treatment for PTSD.[31]
Nabilone has been investigated for its potential to alleviate spasticity and spasticity-related pain in neurological conditions such as multiple sclerosis (MS) and spinal cord injury (SCI).[22] A double-blind, placebo-controlled crossover trial in patients with upper motor neuron disease found that a low dose of Nabilone (1 mg daily) significantly reduced spasticity-related pain compared to placebo, although it did not significantly alter objective measures of spasticity itself.[35] A separate pilot study involving 11 patients with SCI found that Nabilone treatment led to a significant decrease in spasticity as measured by the Ashworth Scale.[36] However, broader evidence reviews have concluded that there is currently limited or insufficient evidence to support the use of cannabinoids, including Nabilone, as an effective treatment for spasticity in most neurological conditions.[37]
While Nabilone is sometimes used off-label as an appetite stimulant for patients with cachexia or wasting disease, its efficacy for this indication is not well established.[23] There is some confusion in the literature, with a few sources suggesting it is approved for anorexia and weight loss in patients with AIDS.[2] However, the more consistent consensus is that this indication is specific to dronabinol, and Nabilone is not formally approved for appetite stimulation.[38]
The continued exploration of Nabilone's therapeutic potential is evident in several ongoing or recent clinical trials targeting novel indications:
These trials signify a clear shift in the developmental trajectory of Nabilone, moving away from its historical role in oncology and toward its potential as a CNS-active agent for challenging neuropsychiatric and behavioral disorders.
The clinical use of Nabilone is intrinsically limited by its safety and tolerability profile. Its potent effects on the central nervous system are responsible for both its potential therapeutic actions and its frequent, often dose-limiting, adverse effects. A thorough understanding of this profile is essential for appropriate patient selection, counseling, and monitoring.
The adverse drug reaction (ADR) profile of Nabilone is dominated by its effects on the central nervous and psychiatric systems. The incidence of side effects is high, although they are typically mild to moderate in severity and may diminish with continued use as tolerance develops.[21] The most common and significant ADRs are detailed in Table 3.
Table 3: Comprehensive List of Adverse Drug Reactions by Frequency and System-Organ Class
System-Organ Class | Frequency | Adverse Reaction |
---|---|---|
Nervous System | Very Common (≥10%) | Drowsiness (up to 66%), Vertigo/Dizziness (up to 59%), Ataxia (up to 14%), Concentration difficulties (up to 12%) |
Common (1%-10%) | Headache, Sedation | |
Frequency not reported | Syncope, Tremor, Memory disturbance, Convulsions, Paresthesia, Coordination disturbance | |
Psychiatric | Very Common (≥10%) | Euphoria (up to 38%), Depression (up to 14%), Sleep disturbance (up to 11%) |
Common (1%-10%) | Dysphoria, Disorientation, Depersonalization syndrome | |
Frequency not reported | Hallucinations, Confusion, Anxiety, Paranoia, Psychosis, Panic disorder | |
Gastrointestinal | Common (1%-10%) | Dry mouth (up to 36%) |
Uncommon (0.1%-1%) | Nausea | |
Frequency not reported | Vomiting, Abdominal pain, Constipation | |
Ocular | Very Common (≥10%) | Visual disturbance (up to 13%) |
Frequency not reported | Eye dryness, Pupil dilation, Photophobia | |
Cardiovascular | Common (1%-10%) | Hypotension |
Frequency not reported | Orthostatic hypotension, Tachycardia, Palpitations, Hypertension | |
Metabolic | Common (1%-10%) | Anorexia, Increased appetite |
Other | Common (1%-10%) | Asthenia (weakness) |
Frequency not reported | Fatigue, Tinnitus, Flushing, Malaise, Irritability |
Source: Compiled from data in.[43]
Nabilone has the potential for numerous clinically significant drug interactions, primarily due to its CNS depressant effects and its metabolism by the cytochrome P450 system. The most critical interactions are summarized in Table 4.
The primary concern is the additive or synergistic CNS depression when Nabilone is co-administered with other substances that slow brain activity. This includes alcohol, opioids, benzodiazepines, barbiturates, sedating antihistamines, and tricyclic antidepressants.[3] Such combinations can lead to profound sedation, respiratory depression, and impaired psychomotor function, and should generally be avoided.[11]
Table 4: Clinically Significant Drug Interactions with Nabilone
Interacting Drug/Class | Potential Effect | Clinical Management Recommendation | Source(s) |
---|---|---|---|
CNS Depressants (e.g., Alcohol, Opioids, Benzodiazepines, Barbiturates, Sedatives) | Additive or synergistic CNS depression, profound sedation, respiratory depression, psychomotor impairment. | Avoid combination. If co-administration is necessary, limit dosages and durations and monitor closely for sedation and respiratory depression. | 3 |
Sympathomimetic Agents (e.g., Amphetamines, Cocaine) | Additive hypertension and tachycardia; potential increase in cardiotoxic risk. | Use with caution and monitor cardiovascular parameters. | 15 |
Anticholinergic Agents (e.g., Atropine, Scopolamine, some Antihistamines) | Enhanced tachycardia and CNS effects, including sedation and confusion. | Use with caution and monitor for increased anticholinergic and CNS side effects. | 15 |
Highly Protein-Bound Drugs (e.g., Warfarin, Phenytoin) | Nabilone may displace other drugs from plasma proteins, increasing their free concentration and potential toxicity. | Monitor for altered pharmacological effects or toxicity of the co-administered drug. | 15 |
CYP450 Inducers/Inhibitors | Co-administration may alter the metabolism of Nabilone, affecting its plasma concentrations and clinical effects. | Use with caution and monitor for changes in efficacy or toxicity. | 11 |
The safety profile of Nabilone dictates a specific set of contraindications and warnings to mitigate the risk of serious adverse events.
As a synthetic cannabinoid with psychoactive effects similar to cannabis, Nabilone has a recognized potential for abuse and the production of psychological dependence.[2] This potential is the primary reason for its classification as a Schedule II controlled substance in the United States.[2] However, several factors may mitigate its real-world abuse liability. Research and expert opinion suggest that its abuse potential is lower than that of smoked cannabis due to its slow oral onset of action, longer time to peak effect, and a side-effect profile where unpleasant effects like drowsiness and vertigo are more prominent than the desired euphoria.[20] Population-level data and reviews of law enforcement and medical databases have found extremely rare instances of Nabilone abuse or diversion.[24] Nonetheless, prescribers must monitor patients for signs of misuse, abuse, or dependence.[11]
The prescribing and dispensing of Nabilone are strictly regulated due to its pharmacological properties and abuse potential. Its regulatory status varies internationally, reflecting different national approaches to cannabinoid-based medicines.
The legal classification of Nabilone is a critical factor governing its availability and use.
This regulatory heterogeneity highlights the complex and evolving global perspective on cannabinoid therapeutics, with the U.S. and Canada applying a high level of control based on abuse potential, while European nations have adopted a more varied, country-by-country approach.
Proper dosing and comprehensive patient education are paramount to maximizing the efficacy and minimizing the risks of Nabilone therapy.
Nabilone is marketed globally under several brand names. The originator brand is Cesamet.[1] The original developer of the compound was Eli Lilly and Company.[20] The rights were later acquired by Valeant Pharmaceuticals International, which has since been renamed Bausch Health Companies.[20] In Europe, manufacturers include AOP Orphan Pharmaceuticals GmbH (for Canemes) and Brown & Burk UK Ltd (for generic Nabilone).[4] Numerous other companies are involved in the manufacturing of the active pharmaceutical ingredient (API).[61]
To fully appreciate Nabilone's position in modern medicine, it is essential to compare it with its closest therapeutic relative, dronabinol, and to understand its unique developmental history.
Nabilone and dronabinol are the two most commonly prescribed synthetic cannabinoids in North America. While they share a similar mechanism of action and therapeutic class, they possess key differences in chemistry, pharmacology, regulation, and clinical use that are critical for informed prescribing. A comparative summary is provided in Table 5.
Table 5: Comparative Profile of Nabilone and Dronabinol
Feature | Nabilone (Cesamet®) | Dronabinol (Marinol®) | Source(s) |
---|---|---|---|
Chemical Identity | Synthetic THC analog | Synthetic THC | 38 |
US DEA Schedule | Schedule II | Schedule III | 38 |
FDA-Approved Indications | Refractory CINV | Refractory CINV; Anorexia in AIDS | 15 |
Potency (vs. THC) | ~2x more potent | 1x (chemically identical) | 2 |
Onset of Action | Slower | Faster (~30 mins) | 38 |
Duration of Action | Longer (8-12+ hours) | Shorter | 38 |
Key PK Difference | Fewer metabolites | More metabolites | 14 |
Allergen Risk | None specified | Contains sesame oil | 38 |
Cost | Typically less expensive | Typically more expensive | 38 |
The history of Nabilone is that of a drug developed for one purpose, overshadowed by newer innovations, and later revived for a new era of medicine. Its journey began in the early 1970s at Eli Lilly and Company, spurred by anecdotal reports from young cancer patients that smoking cannabis alleviated the severe nausea and vomiting caused by chemotherapy.[20]
Nabilone received its first FDA approval in 1985 for refractory CINV.[2] However, its market presence was short-lived. This period coincided with the development and launch of the first 5-HT3 receptor antagonists, such as ondansetron, which offered superior antiemetic efficacy with a far more favorable side-effect profile. Faced with this new class of drugs, older antiemetics with significant CNS side effects, including Nabilone, became less commercially viable. Consequently, Eli Lilly withdrew Nabilone from the U.S. market in 1989, citing "commercial reasons".[20]
Despite its withdrawal from the U.S., Nabilone remained approved and in use in other countries, notably Canada, where it was first approved in 1981.[24] A renewed interest in the therapeutic potential of cannabinoids in the early 2000s set the stage for its return. In 2004, Valeant Pharmaceuticals International (now Bausch Health Companies) acquired the rights to Cesamet from Eli Lilly.[59] After securing a new marketing approval from the FDA, Valeant re-launched Nabilone in the United States in 2006.[2] This re-launch was not aimed at competing with first-line antiemetics but rather at filling the niche for refractory CINV and capitalizing on the growing exploration of cannabinoids for a wide range of off-label indications.
Nabilone occupies a unique and complex position in contemporary pharmacotherapy. Its profile is defined by a central therapeutic dilemma: the need to balance its potential efficacy in difficult-to-treat conditions against a substantial and predictable burden of central nervous system adverse effects. Originally developed as an antiemetic, it has been largely superseded in this role by safer, more effective agents. Yet, it has found a second life as an important tool for clinicians and researchers exploring the therapeutic potential of the endocannabinoid system.
The established efficacy of Nabilone is confined to its narrow, FDA-approved indication for refractory CINV, a clinical niche that has shrunk with the advancement of modern antiemetic regimens. The true drivers of its current use are the numerous off-label applications in neurology and psychiatry, particularly for chronic neuropathic pain, fibromyalgia, and PTSD-associated nightmares. In these areas, where conventional therapies often fail, Nabilone offers a novel mechanism of action. However, the evidence supporting these uses remains largely preliminary, derived from small, often uncontrolled studies. This has created a significant evidence-practice gap, where clinical use has outpaced rigorous scientific validation.
The primary barrier to broader application and higher-dose therapy is Nabilone's safety profile. The high incidence of dose-limiting CNS effects—such as somnolence, dizziness, cognitive impairment, and potential psychiatric disturbances—necessitates careful patient selection, intensive counseling, and stringent monitoring. Its classification as a Schedule II controlled substance further underscores the regulatory concerns regarding its abuse potential, even though real-world abuse appears to be rare. The drug's pharmacokinetic properties, especially the long half-life of its active metabolites, result in a prolonged duration of action that can be both a therapeutic advantage and a safety concern, as adverse effects can persist for days after discontinuation.
Looking forward, the future of Nabilone lies in clarifying its role in these off-label and investigational domains. There is an urgent need for high-quality, long-term, randomized controlled trials to definitively establish its efficacy and safety in chronic pain, PTSD, and spasticity. The ongoing trials in Alzheimer's-related agitation and aggression in developmental disabilities represent promising steps in this direction. Further research into the pharmacology of its long-lasting metabolites could also provide a clearer understanding of its extended duration of effect and inform the development of future cannabinoid-based therapeutics with improved safety profiles.
In conclusion, Nabilone is a historically significant cannabinoid therapeutic that serves as a valuable, albeit challenging, option for a small subset of patients with refractory CINV. Its more important contemporary role may be as a pharmacological probe, providing critical insights into the function of the endocannabinoid system and paving the way for the next generation of medicines for some of the most intractable disorders of the central nervous system.
Published at: September 2, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.