C20H21N
303-53-7
Muscle Spasms
Cyclobenzaprine is a centrally-acting skeletal muscle relaxant widely prescribed for the symptomatic relief of acute muscle spasms. First synthesized in 1961 and approved for human use in the United States in 1977, its clinical application has been focused on its role as an adjunct to rest and physical therapy for painful musculoskeletal conditions.[1] However, to fully comprehend its pharmacological profile, therapeutic limitations, and extensive safety considerations, one must look to its developmental origins.
The history of cyclobenzaprine is not that of a molecule designed from the outset as a muscle relaxant. Instead, it was initially investigated for use as an antidepressant, a path dictated by its striking structural similarity to the class of tricyclic antidepressants (TCAs).[1] It differs from amitriptyline, a cornerstone TCA, by only a single double bond within its tricyclic ring system.[1] While it ultimately failed to demonstrate sufficient efficacy for an antidepressant indication, this developmental history is not a mere historical footnote; it is the single most important factor in predicting and understanding its entire pharmacological identity. Its structural heritage as a TCA derivative foretells its mechanism of action, its broad spectrum of receptor interactions, its characteristic adverse effect profile, and its most serious contraindications. Consequently, a more accurate conceptualization of the drug is not as a simple muscle relaxant, but as a TCA-like agent with pronounced muscle relaxant properties. This nuanced understanding transforms the clinical approach to its use, shifting it from a straightforward symptomatic treatment to a complex pharmacological agent that demands careful patient selection, meticulous monitoring, and a thorough appreciation of its potential for serious drug interactions and adverse events.
Cyclobenzaprine is classified as a small molecule drug and is a tricyclic amine salt.[1] Chemically, it is a derivative of dibenzo[a,d]annulene.[2] The molecule is available and used pharmaceutically as its hydrochloride salt to enhance its physicochemical properties for oral administration.
The existence of two primary forms—the free base and the hydrochloride salt—is a critical distinction in the pharmaceutical sciences. The free base is described as being soluble in organic solvents like DMSO but not in water, a characteristic that would severely limit its absorption from the aqueous environment of the gastrointestinal tract and result in poor and erratic oral bioavailability.[6] The conversion to the hydrochloride salt is a deliberate and necessary step in the manufacturing process to overcome this limitation. The reaction of the parent amine with hydrochloric acid creates a salt that is freely soluble in water, ensuring adequate dissolution and subsequent absorption after oral administration.[3] This chemical modification is fundamental to the drug's clinical viability and highlights a core principle of medicinal chemistry: optimizing a molecule's physicochemical properties is as crucial as its pharmacological activity.
A comprehensive summary of its chemical identifiers and properties is provided in Table 1.
Table 1: Physicochemical and Identification Data for Cyclobenzaprine
Parameter | Value | Source(s) |
---|---|---|
DrugBank ID | DB00924 | 1 |
Type | Small Molecule | 1 |
CAS Number (Free Base) | 303-53-7 | 2 |
CAS Number (HCl Salt) | 6202-23-9 | 2 |
Molecular Formula (Free Base) | C20H21N | 1 |
Molecular Formula (HCl Salt) | C20H22ClN or C20H21N⋅HCl | 5 |
Average Weight (Free Base) | 275.39 g/mol | 1 |
Molecular Weight (HCl Salt) | 311.9 g/mol | 5 |
IUPAC Name | N,N-dimethyl-3-(2-tricyclo[9.4.0.03,8]pentadeca-1(15),3,5,7,9,11,13-heptaenylidene)propan-1-amine | 2 |
InChIKey | JURKNVYFZMSNLP-UHFFFAOYSA-N | 2 |
SMILES | CN(C)CCC=C1C2=CC=CC=C2C=CC3=CC=CC=C31 | 2 |
pKa | 8.47 at 25°C | 3 |
Melting Point (HCl Salt) | 215.0 to 219.0 °C | 5 |
Solubility (HCl Salt) | Freely soluble in water, alcohol, and methanol | |
Appearance | White to off-white crystalline powder | 6 |
ATC Code | M03BX08 | 3 |
FDA UNII | 69O5WQQ5TI | 2 |
Cyclobenzaprine is a centrally-acting skeletal muscle relaxant whose therapeutic effects are mediated primarily within the central nervous system (CNS).[4] Preclinical and clinical evidence indicates that it does not act directly on skeletal muscle tissue or at the neuromuscular junction.[4] Instead, its primary site of action is the brainstem, with potential overlapping activity at the spinal cord level contributing to its overall effect.[1]
The core mechanism of action is understood to be a reduction of tonic somatic motor activity. It influences both gamma (γ) and alpha (α) motor systems, which are responsible for regulating muscle tone and voluntary muscle contraction, respectively.[1] This effect is thought to result from the inhibition of descending motor pathways, such as the coeruleus-spinal and reticulospinal tracts, ultimately leading to depressed activity of spinal cord interneurons that drive muscle hyperactivity.[1] More recent research has elucidated a more specific molecular target, suggesting that antagonism of the 5-hydroxytryptamine receptor 2A (5-HT2A) plays a significant role.[1] By blocking these receptors, cyclobenzaprine inhibits descending serotonergic pathways that can facilitate muscle spasm, contributing substantially to its observed antispasmodic effects.[1]
It is crucial to differentiate cyclobenzaprine's role as an antispasmodic from that of an antispasticity agent. It is effective for relieving skeletal muscle spasms of local origin, such as those arising from acute musculoskeletal injury.[4] However, it has not been found to be effective in treating spasticity, a condition of velocity-dependent muscle hyperactivity that originates from upper motor neuron lesions associated with cerebral or spinal cord disease, or in children with cerebral palsy.[2]
The clinical effects of cyclobenzaprine, both therapeutic and adverse, are the product of its promiscuous binding to multiple receptor systems—a direct legacy of its TCA structure. While its antispasmodic action is linked to the brainstem and 5-HT2A antagonism, its broader pharmacological footprint includes potent activity at several other "off-target" receptors. It possesses strong central and peripheral anticholinergic (muscarinic receptor antagonist) properties, antagonizes alpha-1 adrenergic receptors, and functions as a general CNS depressant.[4] These additional actions are not ancillary; they are central to the drug's clinical profile and directly responsible for its high burden of side effects. The anticholinergic activity causes the very common side effects of dry mouth, blurred vision, urinary retention, and confusion.[4] Its general CNS depressant effects lead to the profound drowsiness, somnolence, and fatigue reported by a large percentage of users.[14] Finally, alpha-1 adrenergic antagonism can contribute to dizziness, orthostatic hypotension, and reflex tachycardia.[4] This multi-target profile creates a narrow therapeutic window, as evidenced by clinical data showing a Number Needed to Treat (NNT) of 3 for benefit, which is uncomfortably close to the Number Needed to Harm (NNH) of 4 for adverse effects.[16] This classic pharmacological trade-off dictates its clinical niche: short-term use in acute situations where the benefit of interrupting a pain-spasm cycle is deemed to outweigh the near certainty of dose-limiting adverse effects.
The pharmacokinetic profile of cyclobenzaprine is characterized by moderate bioavailability, extensive distribution, comprehensive hepatic metabolism, and a long elimination half-life that varies significantly across patient populations.
Absorption: Following oral administration, cyclobenzaprine has a moderate and variable bioavailability, estimated to be between 33% and 55%.[1] Peak plasma concentrations (
Cmax) ranging from 5 to 35 ng/mL are typically achieved approximately 4 hours (Tmax) after ingestion.[1] The presence of food has been shown to increase both the rate and extent (Area Under the Curve, AUC) of absorption, a factor that may be clinically relevant.[4]
Distribution: The drug is highly lipophilic, leading to extensive distribution into tissues. It is highly bound to plasma proteins, at approximately 93%.[1] It demonstrates a high affinity for human serum albumin and also binds to alpha-1 glycoprotein, an acute phase reactant protein that can be elevated during inflammatory conditions, potentially altering free drug concentrations.[1]
Metabolism: Cyclobenzaprine undergoes extensive metabolism in the liver through both oxidative and conjugative pathways, with very little of the parent drug being excreted unchanged.[1] The primary oxidative pathway is N-demethylation, which is catalyzed mainly by the cytochrome P450 enzymes CYP3A4 and CYP1A2, with a lesser contribution from CYP2D6.[1] The drug also undergoes N-glucuronidation, a conjugation reaction catalyzed by the enzymes UGT1A4 and UGT2B10.[1] Furthermore, cyclobenzaprine is subject to enterohepatic circulation, a process where the drug is excreted in the bile, reabsorbed in the intestine, and returned to the liver, which can contribute to its prolonged presence in the body.[1]
Excretion: Elimination occurs primarily via the kidneys, with metabolites (likely water-soluble glucuronide conjugates) being excreted in the urine. Only about 1% of the administered dose is recovered in the urine as the unchanged parent drug.[1] The effective elimination half-life (
t1/2) is notably long, averaging approximately 18 hours in healthy young adults.[1]
A critical aspect of cyclobenzaprine's clinical pharmacology is the significant disconnect between its long elimination half-life and its relatively short duration of clinical action, which is typically 4 to 6 hours.[1] This pharmacokinetic mismatch has profound implications for its use. The short duration of action necessitates multiple daily doses (e.g., every 8 hours) for the immediate-release formulation to maintain a therapeutic effect. However, the long half-life means that with each subsequent dose, the drug steadily accumulates in the body, reaching higher steady-state concentrations over several days. This accumulation risk is dramatically amplified in populations with reduced drug clearance. In elderly patients, the mean half-life extends to approximately 33 hours, and in patients with hepatic insufficiency, it can reach 46 hours.[1] In these individuals, standard dosing can lead to the rapid build-up of the drug to potentially toxic levels, markedly increasing the risk of CNS adverse effects such as confusion, excessive sedation, and falls. This pharmacokinetic behavior is the scientific foundation for several key clinical guidelines: it explains why therapy is strictly limited to 2-3 weeks (to prevent excessive accumulation), why geriatric and hepatically impaired patients require significant dose reductions and cautious titration, and why the extended-release formulation was developed to provide a smoother concentration profile for once-daily dosing, although this formulation is considered too risky for populations with impaired clearance.[1]
Table 2: Key Pharmacokinetic Parameters of Cyclobenzaprine
Parameter | Value (Healthy Young Adult) | Value (Special Populations) | Source(s) |
---|---|---|---|
Oral Bioavailability | 33% to 55% | Not specified, but exposure is increased in hepatic impairment and elderly. | 1 |
Tmax (Time to Peak) | ~4 hours | Not specified | 1 |
Plasma Protein Binding | ~93% | Not specified | 1 |
Primary Metabolic Enzymes | CYP3A4, CYP1A2, UGT1A4, UGT2B10 | N/A | 1 |
Elimination Half-life (t1/2) | ~18 hours | Elderly: ~33.4 hours; Hepatic Impairment: ~46.2 hours | 1 |
Plasma Clearance | ~0.7 L/min | Decreased in elderly and hepatic impairment. | 1 |
The approved indication for cyclobenzaprine is narrow and specific. It is indicated as a short-term (for periods of two to three weeks) adjunct to rest and physical therapy for the relief of muscle spasm associated with acute, painful musculoskeletal conditions.[1] It is one of the most well-studied drugs for this application.[6] Clinical evidence supports its effectiveness in improving the signs and symptoms of muscle spasm, including reducing local pain and tenderness and increasing range of motion, thereby helping to restore function.[4] Common conditions for which it is prescribed under this indication include acute low back pain, neck pain, strains, sprains, and other muscle injuries.[8]
Beyond its approved indication, cyclobenzaprine is frequently used off-label for a variety of conditions. The most prominent off-label application is in the management of fibromyalgia, a chronic condition characterized by widespread pain, fatigue, and sleep disturbances. In this context, it is used to reduce pain and improve sleep quality.[2] The interest in this indication led to the development of a novel sublingual formulation (Tonmya) intended for the chronic management of fibromyalgia; however, a Phase 3 clinical trial (NCT02829814) for this formulation was terminated.[6]
Other documented off-label and investigational uses include the treatment of Chronic Myofascial Pain, Temporomandibular Joint (TMJ) Disorder, sciatica, and certain types of headache, including migraine.[6] Clinical trial data from the Prospective Analgesic Compound Efficacy (PACE) Study (NCT02403687) show that cyclobenzaprine has been investigated as part of a multi-drug study for conditions including arthritis, tendonitis, and nerve root disorders, though its specific efficacy in these areas is not detailed.[24]
While cyclobenzaprine is established as an effective treatment for acute muscle spasm, a critical appraisal of the clinical evidence reveals a more nuanced picture of its overall benefit. Pooled analyses of clinical trials for low back pain have calculated a Number Needed to Treat (NNT) of 3, meaning that for every three patients treated with cyclobenzaprine instead of placebo, one additional patient will experience "global improvement" in symptoms by day 10.[16]
However, this seemingly favorable statistic is significantly tempered by two major caveats. First, the Number Needed to Harm (NNH) is 4, indicating that for every four patients treated, one will experience an adverse effect (typically drowsiness, dry mouth, dizziness, or nausea) that they would not have experienced on placebo.[16] The narrow margin between the NNT and NNH suggests a challenging risk-benefit ratio, where the likelihood of experiencing harm is nearly as high as the likelihood of experiencing benefit.
Second, the primary outcome measure of "global improvement" is often nebulously defined in the underlying studies and, importantly, does not appear to integrate the negative impact of adverse effects into the overall assessment of benefit.[16] This raises a critical question: does a patient who has less pain but is functionally impaired by significant drowsiness and dizziness truly represent a case of "global improvement"? The therapeutic benefit appears to be most pronounced within the first four days of treatment, a period that coincides with the most acute sedative effects of the drug before tolerance develops.[16] This observation, coupled with findings from a high-quality randomized controlled trial suggesting minimal additional pain-relieving effect when cyclobenzaprine is added to naproxen for acute low back pain, leads to an alternative interpretation of its efficacy.[16] The primary clinical utility of cyclobenzaprine may not stem from a specific, potent muscle relaxant action, but rather from its powerful sedative properties, which enforce rest and help break the acute pain-spasm-pain cycle in the initial days of an injury. This reframes its role from a targeted therapy to a non-specific symptomatic treatment whose main mechanism of "relief" may be sedation. This has important implications for clinical practice, suggesting that for patients who cannot tolerate or afford the functional impairment of sedation (e.g., workers, drivers, caregivers), cyclobenzaprine may be an inappropriate choice, and alternative therapies should be prioritized.
Cyclobenzaprine is available for oral administration in two main formulations: immediate-release (IR) tablets and extended-release (ER) capsules.[19]
The recommended dosing for adults and adolescents aged 15 years and older is as follows:
Regardless of the formulation, the duration of therapy should be strictly limited. Cyclobenzaprine should only be used for short periods, up to a maximum of two to three weeks, as adequate evidence of effectiveness for more prolonged use is not available, and the underlying condition of acute muscle spasm is generally of short duration.[1]
The pharmacokinetic profile of cyclobenzaprine necessitates significant dosing adjustments and restrictions in certain high-risk populations. These guidelines are not arbitrary; they are a direct clinical application of the drug's known pharmacokinetic risks, primarily related to its extensive hepatic metabolism and long, variable half-life, which can lead to dangerous drug accumulation.
For optimal use, patients should be instructed on proper administration techniques.
The tolerability of cyclobenzaprine is significantly limited by its adverse effect profile, which is a direct reflection of its TCA-like pharmacology. The most frequently reported adverse reactions are related to its CNS depressant and anticholinergic properties.
Table 3: Common and Serious Adverse Reactions to Cyclobenzaprine
System Organ Class | Adverse Reaction | Incidence/Frequency | Source(s) |
---|---|---|---|
Nervous System | Drowsiness/Somnolence | Very Common (up to 39%) | 14 |
Dizziness | Common (up to 11%) | 14 | |
Headache | Common (5%) | 15 | |
Decreased Mental Acuity | Common (1-3%) | 14 | |
Seizures, Ataxia, Tremors | Rare/Postmarketing | 14 | |
Psychiatric | Fatigue | Common (6%) | 14 |
Nervousness, Irritability | Common (1-3%) | 14 | |
Confusion, Hallucinations, Agitation, Anxiety, Abnormal Dreams | Rare/Postmarketing | 1 | |
Gastrointestinal | Dry Mouth | Very Common (up to 32%) | 14 |
Nausea | Common (1-3%) | 14 | |
Constipation, Dyspepsia | Common (1-3%) | 14 | |
Vomiting, Gastritis, Diarrhea | Rare/Postmarketing | 14 | |
Ocular | Blurred Vision | Common (1-3%) | 14 |
Cardiovascular | Tachycardia, Arrhythmia, Palpitation, Hypotension, Syncope | Rare/Postmarketing | 14 |
Hepatic | Abnormal Liver Function, Hepatitis, Jaundice, Cholestasis | Rare/Postmarketing | 14 |
Hypersensitivity | Rash, Urticaria, Pruritus, Angioedema, Anaphylaxis | Rare/Postmarketing | 14 |
The use of cyclobenzaprine is absolutely contraindicated in several clinical situations where the risk of serious harm is unacceptably high. These contraindications are directly linked to its TCA-like pharmacology.
Beyond its absolute contraindications, the use of cyclobenzaprine carries several significant warnings and requires numerous precautions to ensure patient safety.
Serotonin Syndrome: A major, potentially life-threatening risk associated with cyclobenzaprine is the development of serotonin syndrome. This risk is a direct consequence of its inherent serotonergic activity, a property inherited from its TCA origins. The danger is particularly acute when cyclobenzaprine is co-administered with other serotonergic drugs, such as selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), other TCAs, triptans, tramadol, bupropion, and MAOIs.[1] The common classification of cyclobenzaprine as merely a "muscle relaxant" dangerously masks this risk, creating a potential for prescribers to inadvertently trigger this serious adverse event through common drug combinations. Case reports have documented severe serotonin syndrome in patients who received cyclobenzaprine while on stable doses of other serotonergic agents like phenelzine (an MAOI) and duloxetine (an SNRI).[31] Clinicians must maintain a high index of suspicion and monitor for symptoms, which include mental status changes (e.g., confusion, agitation, hallucinations), autonomic instability (e.g., tachycardia, labile blood pressure, sweating, hyperthermia), and neuromuscular abnormalities (e.g., tremor, hyperreflexia, myoclonus, rigidity).[1] If serotonin syndrome is suspected, cyclobenzaprine and all other serotonergic agents must be discontinued immediately.[17]
Cardiac Effects: The cardiac risks of cyclobenzaprine are a ghost of its TCA past. These are not general precautions but specific, mechanistically-driven risks related to the blockade of fast-acting sodium channels in the cardiac conduction system, an action characteristic of TCAs.[4] This can slow cardiac depolarization, prolong the conduction time, and lead to arrhythmias, sinus tachycardia, and, in severe cases, myocardial infarction and stroke.[4] While some evidence suggests cyclobenzaprine may be less cardiotoxic in overdose (at doses less than 1000 mg) than older TCAs in otherwise healthy individuals, the existence of absolute contraindications for patients with
any pre-existing cardiac conduction disease implies that even standard therapeutic doses can be sufficient to destabilize a compromised cardiovascular system.[4] This risk profile effectively disqualifies a large and highly relevant patient population—older adults with musculoskeletal pain who often have comorbid cardiovascular disease—from using the drug safely.
CNS Depression: Cyclobenzaprine is a potent CNS depressant. It can significantly impair mental and physical abilities required for performing hazardous tasks. Patients must be unequivocally warned against driving a car or operating heavy machinery until they are reasonably certain that the medication does not adversely affect their alertness or coordination.[18] The sedating effects are additive with those of other CNS depressants, including alcohol, barbiturates, benzodiazepines, and opioids. Co-administration can lead to profound sedation, respiratory depression, and an increased risk of overdose.[5]
Anticholinergic (Atropine-like) Action: Due to its potent anticholinergic effects, cyclobenzaprine should be used with caution in patients with a history of urinary retention, angle-closure glaucoma, or increased intraocular pressure, as it can exacerbate these conditions.[4]
Overdose and Toxicology: Overdose with cyclobenzaprine can be serious. The most common manifestations are significant drowsiness and tachycardia. Less common but more severe signs include tremor, agitation, confusion, hallucinations, ataxia, seizures, severe hypotension, cardiac dysrhythmias, and, rarely, cardiac arrest and neuroleptic malignant syndrome.[1] Management is complex and primarily supportive, often involving gastrointestinal decontamination and close cardiac monitoring. Consultation with a poison control center is strongly recommended. Due to extensive tissue distribution and high protein binding, dialysis is of no value in enhancing elimination.[1]
The potential for clinically significant drug-drug interactions with cyclobenzaprine is high, stemming from its multiple mechanisms of action and metabolic pathways. A thorough medication reconciliation is essential before initiating therapy. The most critical interactions are summarized in Table 4.
Table 4: Clinically Significant Drug Interactions with Cyclobenzaprine
Interacting Agent/Class | Potential Outcome | Clinical Management/Recommendation | Source(s) |
---|---|---|---|
Monoamine Oxidase Inhibitors (MAOIs) (e.g., phenelzine, tranylcypromine, selegiline) | Life-threatening Serotonin Syndrome, hyperpyretic crisis, severe seizures, death. | ABSOLUTELY CONTRAINDICATED. Do not use cyclobenzaprine within 14 days of MAOI administration. | 5 |
Other Serotonergic Agents (e.g., SSRIs, SNRIs, TCAs, triptans, tramadol, bupropion) | Increased risk of Serotonin Syndrome (agitation, hyperthermia, hyperreflexia, autonomic instability). | Avoid combination if possible. If concomitant use is clinically necessary, monitor patient closely for signs of serotonin toxicity, especially during treatment initiation or dose increases. | 17 |
CNS Depressants (e.g., Alcohol, Benzodiazepines, Opioids, Barbiturates, Sedating Antihistamines) | Additive CNS depression leading to excessive sedation, psychomotor impairment, respiratory depression, and increased risk of overdose. | Avoid combination, especially with alcohol. Counsel patients on risks and warn against driving or operating machinery. Consider dose reduction of one or both agents. | 5 |
Anticholinergic Agents (e.g., benztropine, oxybutynin, diphenhydramine, scopolamine) | Additive anticholinergic effects, resulting in severe dry mouth, blurred vision, urinary retention, constipation, and confusion (delirium), particularly in the elderly. | Use with caution. Monitor closely for anticholinergic toxicity. May require dose reduction or discontinuation of one agent. | 5 |
Tramadol | Increased risk of Serotonin Syndrome and increased risk of seizures, as both agents can lower the seizure threshold. | Use with extreme caution. Monitor for both serotonin syndrome and seizure activity. | 17 |
Verapamil | May interact to increase the risk of serotonin syndrome. | Monitor for signs and symptoms of serotonin syndrome. | 17 |
Cyclobenzaprine is a pharmacologically complex agent whose identity is more accurately defined by its tricyclic antidepressant heritage than by its simple classification as a skeletal muscle relaxant. This structural relationship dictates its entire clinical profile, from its central mechanism of action in the brainstem to its extensive adverse effect burden and serious safety considerations.
The evidence supports its efficacy as a short-term treatment for acute musculoskeletal spasms, where it can reduce pain and improve mobility. However, this modest benefit is achieved at the cost of a high incidence of adverse effects, primarily sedation and anticholinergic symptoms, resulting in a narrow therapeutic window. Critical analysis suggests that its primary therapeutic contribution may be its potent sedative effect, which enforces rest and helps interrupt the acute pain-spasm cycle, rather than a highly specific muscle relaxant action. Its long and variable elimination half-life creates a significant risk of drug accumulation, particularly in the elderly and those with hepatic impairment, making it a high-risk medication in these vulnerable populations. Furthermore, its inherent serotonergic and cardiotoxic properties, also inherited from its TCA structure, lead to absolute contraindications and the potential for life-threatening drug interactions.
To maximize benefit and minimize harm, the use of cyclobenzaprine should be guided by a clear understanding of its pharmacological profile and inherent risks. The following recommendations are provided for clinicians:
Published at: August 23, 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.