Fact-checked by Grok 2 weeks ago

Trazodone

Trazodone is a triazolopyridine classified as a (SARI), approved by .S. Food and (FDA) in 1981 for the treatment of (MDD) in adults. It works by inhibiting the reuptake of serotonin and antagonizing serotonin 5-HT2A receptors, as well as blocking histamine H1 and alpha-1 adrenergic receptors, which contributes to its and sedating effects. Primarily indicated as monotherapy or in for MDD, trazodone helps improve mood, appetite, energy levels, and sleep while reducing anxiety and related to . At lower doses (typically 50-100 mg), it is widely used off-label for due to its of 5 to 9 hours and minimal daytime drowsiness. Developed as an alternative to antidepressants, trazodone offers a favorable profile for many patients, though it carries a warning for increased risk of suicidal thoughts and behaviors, particularly in children, adolescents, and young adults under 24 years old. Common side effects include drowsiness and ; serious risks may include , , and cardiac effects. It is contraindicated with inhibitors (MAOIs) and requires caution with certain comorbidities or concomitant medications; see dedicated sections for details. As of 2025, ongoing monitors potential new adverse events, including discontinuation effects. Off-label applications extend to conditions such as anxiety disorders, (PTSD) nightmares, , and substance use disorders, supported by its and properties, with emerging research into combinations for . Administered orally, dosing starts at 150 mg daily for MDD, titrated as needed; therapeutic effects may take two weeks or longer. and healthcare coordination are essential for safe use.

Medical Uses

Treatment of Depression

Trazodone was approved by the U.S. in 1981 for the treatment of (MDD) in adults. The drug's effects are primarily attributed to its action as a (SARI), involving selective inhibition of reuptake at the presynaptic neuronal membrane and antagonism at 5-HT2A receptors, which enhances activity in the . For MDD, treatment typically begins with 150 per day administered in divided doses after meals or a light snack to minimize gastrointestinal upset, with gradual increases of 50 every 3 to 4 days as tolerated, up to a maintenance range of 150 to 600 per day in divided doses; lower starting doses are recommended to reduce initial . Clinical trials have demonstrated trazodone's superiority over in treating MDD, with significant improvements in Hamilton Depression Rating Scale (HDRS) scores observed in short-term studies of 6 to 8 weeks. In comparative studies, trazodone shows efficacy similar to antidepressants (TCAs) such as amitriptyline, but with improved tolerability due to a lower incidence of and cardiovascular side effects. Response rates in these short-term trials typically range from 50% to 60%, based on criteria such as at least a 50% reduction in HDRS scores, though remission rates are generally lower.

Treatment of Insomnia

Trazodone is widely used off-label at low doses of 25 to 100 mg administered at to treat primary or comorbid with other conditions. This dosing regimen leverages its properties while minimizing effects typically seen at higher doses. Meta-analyses demonstrate modest efficacy for trazodone in improving key parameters compared to . A 2024 systematic review and reported that trazodone extended objective total time by 28 minutes, reduced nocturnal awakenings, decreased time awake after onset by 13 minutes, and enhanced overall quality and efficiency, though it had no effect on subjective total time. Similarly, a 2023 review found reductions in nightly awakenings and slight improvements in subjective quality, but no significant changes in total time, sleep latency, or efficiency. Evidence for sustained benefits is limited, with most trials lasting no longer than 12 weeks and lacking robust long-term data. Key advantages include trazodone's low abuse potential relative to benzodiazepines, positioning it as a safer alternative for patients prone to . It also promotes deeper sleep stages by increasing (N3), which supports restorative rest without substantial disruption to overall architecture. Limitations persist, as trazodone receives incomplete endorsement in clinical guidelines; the (AASM) issues a weak recommendation against its use for sleep onset or maintenance , citing insufficient high-quality randomized controlled trials where benefits do not clearly outweigh harms like and . Potential risks include to its effects with extended use and rebound upon discontinuation, though direct studies in primary remain scarce. As of 2025, recent reviews underscore insufficient evidence supporting chronic trazodone use for , advocating for additional randomized controlled trials to clarify its role amid growing off-label prescriptions. For depression-related , it may be used adjunctively with SSRIs in limited durations to address sleep disturbances alongside mood symptoms.

Other Approved Uses

Trazodone is approved in several European countries for the treatment of with or without associated anxiety symptoms. This indication encompasses depressive episodes where anxiety is a prominent feature, allowing its use in patients experiencing comorbid anxiety within the context of . In the , for instance, it is indicated for both and anxiety disorders. Although not formally approved by the FDA or for standalone anxiety disorders, trazodone is authorized in some regions as an adjunct therapy for , particularly when linked to depressive states. Typical dosing for anxiety management ranges from 50 to 300 mg per day, often starting at lower doses and titrated based on response and tolerability.

Off-Label and Emerging Uses

Trazodone has been explored off-label for the management of induced by selective serotonin inhibitors (SSRIs), primarily through its antagonism of 5-HT2A receptors, which may counteract the effects contributing to impaired and . Small open-label trials have demonstrated improvements in , with patients reporting enhanced and overall satisfaction at doses ranging from 50 to 150 mg daily, though these findings are preliminary and lack robust placebo-controlled validation. In the context of fibromyalgia and chronic pain syndromes, trazodone has shown potential in phase II and open-label studies, where doses of 100-200 mg per day led to reductions in pain severity and improved sleep quality, potentially due to its effects on serotonin and noradrenergic pathways. For instance, a 2010 study involving 66 patients reported significant decreases in impact scores after 12 weeks of , with benefits extending to associated depressive symptoms. Recent reviews, including those from 2024, highlight its adjunctive role when combined with agents like , though evidence remains derived from smaller cohorts rather than large randomized controlled trials (RCTs). As of 2025, emerging investigations suggest trazodone's utility in alleviating withdrawal symptoms during treatment, particularly for associated and , with low doses (50-100 mg) aiding without exacerbating relapse risk in select cases. Similarly, preliminary data indicate a role in mitigating symptoms of , such as hot flashes, through its modulation, as evidenced by a 1996 open-label trial showing symptom relief at 75 mg daily and ongoing 2025 comparative studies evaluating its efficacy against digital therapies and other agents for menopause-related sleep disturbances. Trazodone has also been utilized off-label in the management of post-traumatic stress disorder (PTSD), particularly for reducing nightmares, supported by evidence from U.S. Department of (VA) studies demonstrating its efficacy through 5-HT2A receptor blockade. These studies in veteran populations showed improvements in sleep disturbances and PTSD-related symptoms at doses of 50 to 300 mg daily. In comparisons with , another , trazodone exhibits similar efficacy in alleviating PTSD symptoms, including nightmares and overall symptom severity, but with a lower risk of hepatic toxicity. Overall, these off-label applications are supported predominantly by open-label studies and small RCTs, with no large-scale confirmatory trials establishing definitive efficacy or safety profiles. For pediatric anxiety, limited data exist on trazodone's potential benefits, but its use is not recommended due to unestablished safety, including risks of highlighted in FDA warnings for antidepressants in youth. Additionally, trazodone's 5-HT2A antagonism has been anecdotally referenced in managing acute psychedelic experiences, though this remains unverified in clinical settings and is better addressed under drug interactions.

Dosage and Administration

Trazodone is available primarily as immediate-release oral tablets in strengths of 50 , 100 , 150 , and 300 , which can be swallowed whole or split if scored but should not be chewed or crushed. An oral solution (10 /) exists for patients who have difficulty tablets. For the treatment of , the recommended initial dose in adults is 150 mg per day administered orally in divided doses, typically after a meal or light snack to minimize gastrointestinal upset and enhance absorption. The dose may be increased incrementally by 50 mg per day every three to four days based on clinical response and tolerability, with a maximum of 400 mg per day for outpatients and up to 600 mg per day for hospitalized patients under close supervision. For in , lower starting doses of 25 to 100 mg are typically administered as a single bedtime dose, titrated as needed but generally not exceeding 100 mg to balance effects and risks. therapy should use the lowest effective dose for several months after response, with gradual tapering upon discontinuation to avoid withdrawal symptoms. Trazodone should be taken orally with food to reduce the risk of and , and patients are advised to avoid , as it can potentiate and . Concurrent use with strong inhibitors (e.g., ) may necessitate dose reductions due to increased trazodone plasma levels. In special populations, such as the elderly, a lower starting dose of 100 mg per day in divided doses is recommended to mitigate risks of and , with slower titration. For patients with hepatic , trazodone has not been specifically studied, but caution is advised; single doses above 100 mg should be avoided, and use is generally not recommended in severe cases. No dosage adjustment is routinely required for mild to moderate renal , though is essential. Monitoring includes regular assessment of in supine and standing positions to detect , particularly in the elderly or those with cardiovascular comorbidities. Electrocardiogram (ECG) evaluation for prolongation is recommended prior to initiation and periodically in patients over 65 years or with risk factors for arrhythmias. Clinical response and side effects should be evaluated every 3 to 4 days during to guide adjustments.

Adverse Effects

Common Side Effects

The most frequently reported side effect of trazodone is or drowsiness, affecting approximately 20-50% of patients, particularly at higher doses used for treatment. This effect is primarily attributed to trazodone's antagonism of H1 receptors, which promotes . The sedative onset typically occurs within 30-60 minutes, peaking 1-2 hours after dosing due to rapid absorption and peak plasma concentrations. Other common side effects include dry mouth, , and , with incidences ranging from 10-30% across clinical trials. Dry mouth arises from trazodone's mild activity, which reduces secretion. and lightheadedness are linked to blockade, leading to . may result from similar central effects but are generally transient. Gastrointestinal disturbances, such as and , occur in 5-15% of users, often related to modulation and slowed gut motility. Management of these side effects focuses on dose optimization and supportive measures. For sedation, administering the majority of the dose at bedtime can minimize daytime impairment, while gradual dose reduction may alleviate excessive drowsiness. Dry mouth can be addressed through increased , sugarless gum, or substitutes. If symptoms persist, consulting a healthcare provider for dose adjustment or alternative therapies is recommended.

Serious Adverse Effects

Trazodone is associated with several serious adverse effects, though they occur infrequently. , a prolonged and painful lasting more than four hours, has been reported in approximately 1 in 6,000 to 10,000 male prescriptions. This condition requires immediate medical intervention to prevent permanent , typically involving discontinuation of the drug and emergency treatments such as corporal aspiration and irrigation. The risk is heightened when trazodone is combined with low-potency antipsychotics, which share alpha-1 adrenergic blocking properties that can exacerbate vascular effects. Cardiac complications represent another critical concern with trazodone use. The drug can prolong the , with postmarketing reports of , particularly at doses of 100 mg/day or higher; this risk is greater in patients with preexisting cardiac conditions or those taking concomitant QT-prolonging medications. , sometimes leading to syncope, has also been observed, necessitating caution and close monitoring in patients with . Although a warning specifically for is not present, the FDA recommends ECG monitoring in at-risk individuals due to the potential for life-threatening arrhythmias. An increased risk of suicidality is a well-documented concern, particularly in children, adolescents, and young adults under 24 years of age. The FDA has issued a highlighting that antidepressants like trazodone may elevate suicidal thoughts and behaviors, especially during the first one to two months of or with dose changes; patients should be closely monitored for worsening or emergent suicidality. Other rare but serious effects include , often due to syndrome of inappropriate antidiuretic hormone secretion (SIADH), with cases reporting sodium levels below 110 mmol/L, particularly in elderly or volume-depleted patients. Discontinuation is advised if symptoms such as or seizures occur. Additionally, parkinsonism-like symptoms, including and rigidity, have been reported in isolated 2024 case studies, resolving upon even after short-term moderate dosing for . These effects can be exacerbated in overdose scenarios, underscoring the need for prompt management.

Effects During Pregnancy and Breastfeeding

Trazodone is classified as FDA C, meaning animal reproduction studies have shown adverse effects on the , such as increased fetal resorption and congenital anomalies at doses 6-17 times the maximum recommended human dose, but there are no adequate and well-controlled studies in humans, and the drug should be used during only if the potential benefit justifies the potential risk to the . Available human data from pregnancy registries and cohort studies, including aggregate analyses exceeding 500 exposures, indicate no major teratogenic effects, with major congenital malformation rates remaining low and comparable to unexposed populations. However, some evidence suggests a possible association with , observed in 13.5% of trazodone-exposed pregnancies compared to 8.3% in SSRI-exposed controls in one multi-center cohort. Neonates exposed to trazodone in late may experience symptoms or serotonin-related effects, including respiratory distress, jitteriness, or poor feeding, occurring in approximately 21% of term-exposed infants in cohort data, though rates are similar to those with SSRI exposure. A 2025 systematic of 14 studies further confirmed the low risk of malformations (odds around 1.1 in pooled analyses) and no consistent links to or , emphasizing the need for individualized risk-benefit assessment. Clinical recommendations advise using trazodone only when benefits outweigh risks, with consideration of tapering the dose prior to delivery to minimize neonatal complications. Regarding breastfeeding, trazodone and its metabolites are excreted into human milk at low concentrations, typically representing 0.1-1% of the maternal weight-adjusted dose, with relative doses estimated at 0.8% based on case reports and pharmacokinetic studies. serum levels decline rapidly post-exposure, and no adverse effects have been consistently reported in breastfed infants, though for , poor feeding, or irritability is recommended due to limited long-term data. Authoritative sources suggest that is generally compatible with trazodone use at therapeutic doses, provided maternal and are closely observed.

Overdose and Management

Trazodone overdose typically manifests with severe , including pronounced sedation, drowsiness, and potentially , alongside respiratory depression that may progress to arrest. Cardiovascular effects are prominent, encompassing , , irregular heartbeats, and arrhythmias such as QT prolongation or , which can lead to fainting or . Other symptoms include —a prolonged, painful erection requiring urgent intervention—and, less commonly, seizures, particularly in cases exceeding therapeutic doses of 600 mg, as well as or . Fatalities from trazodone overdose alone are exceedingly rare, with documented cases typically involving doses above 6 g, such as one reported at 6.45 g leading to and death; overdoses below 2 g seldom result in lethality when managed promptly. The drug's wide contributes to this low toxicity profile, with supportive care being the cornerstone of survival in most instances. Epidemiologically, among 206 reported trazodone overdoses, no deaths occurred, contrasting sharply with higher mortality in overdoses, underscoring a approaching 0% in isolated cases. Management of trazodone overdose is primarily supportive, focusing on airway protection, hemodynamic stabilization, and monitoring in an intensive care setting. Gastrointestinal decontamination with activated charcoal is recommended if ingestion occurred within 1-2 hours and the patient can protect their airway, though it is not routinely indicated beyond this window. Seizures, if present, should be treated with benzodiazepines such as or ; is contraindicated due to the risk of precipitating seizures in the context of overdose. necessitates immediate urological consultation, often involving intracavernosal injection. responds to intravenous fluids and, if refractory, vasopressors like norepinephrine. Continuous cardiac monitoring via electrocardiogram (ECG) is essential to detect arrhythmias, prolongation, or rare QRS widening, with serial assessments to address . , including and , require close observation, and may be needed for . Consultation with a is advised to guide , as no specific exists.

Drug Interactions

Pharmacokinetic Interactions

Trazodone is primarily metabolized by the 3A4 () enzyme, making it susceptible to pharmacokinetic interactions with inhibitors and inducers that alter its plasma concentrations. Potent inhibitors, such as and , can substantially increase trazodone exposure; for instance, at 200 mg twice daily elevates the area under the curve (AUC) by approximately 2.4-fold and maximum concentration (Cmax) by 34%. In such cases, a dose reduction of trazodone by 50% is recommended, with based on tolerability and clinical response to mitigate risks like enhanced . Conversely, strong inducers like and rifampin decrease trazodone plasma levels, potentially reducing its efficacy; , for example, lowers trazodone concentrations by 76%. When coadministered with inducers, monitoring of therapeutic response is advised, and an increase in trazodone dosage may be necessary to achieve desired effects. Trazodone exhibits high protein binding of 89-95% at therapeutic concentrations, which may lead to interactions with other highly bound drugs like , necessitating monitoring of international normalized ratio (INR) for potential alterations in anticoagulant effects. The elimination half-life of trazodone is typically 5-9 hours in healthy adults but can be prolonged in elderly patients and those with hepatic impairment due to reduced metabolism. As of 2025, updated guidelines from the New York State Department of Health AIDS Institute recommend administering the lowest effective dose of trazodone with HIV protease inhibitors like ritonavir-boosted regimens and monitoring for central nervous system effects, consistent with prior findings on elevated exposure.

Pharmacodynamic Interactions

Trazodone is contraindicated in patients taking monoamine oxidase inhibitors (MAOIs) or within 14 days of discontinuing MAOIs, due to the risk of serious, potentially fatal serotonin syndrome or hypertensive crisis. Trazodone, a serotonin antagonist and reuptake inhibitor (SARI), can potentiate serotonergic activity when combined with other agents that enhance serotonin levels, leading to serotonin syndrome—a potentially life-threatening condition characterized by symptoms such as hyperthermia, muscle rigidity, autonomic instability, and altered mental status. This risk is particularly notable with selective serotonin reuptake inhibitors (SSRIs) like fluoxetine or serotonin-norepinephrine reuptake inhibitors (SNRIs) like duloxetine and venlafaxine, where the combined inhibition of serotonin reuptake amplifies neurotransmission. The incidence of serotonin syndrome with trazodone is rare, estimated at less than 1% in patients on antidepressants, though it can occur even at therapeutic doses when multiple serotonergic agents are involved. Recent analyses as of 2025 confirm an elevated risk when trazodone is co-administered with triptans (e.g., sumatriptan), which act as serotonin receptor agonists and may exacerbate serotonergic overload. Trazodone also exhibits additive effects on cardiac conduction, prolonging the and increasing the risk of when used alongside other QT-prolonging agents such as antiarrhythmics (e.g., ) or antipsychotics. This pharmacodynamic synergy arises from trazodone's blockade of cardiac channels, compounded by similar mechanisms in interacting drugs, necessitating electrocardiographic monitoring in at-risk patients. Additionally, trazodone enhances (CNS) depression when combined with agents like or opioids (e.g., ), resulting in amplified drowsiness, respiratory suppression, and impaired function. Co-administration should be avoided due to the potential for severe outcomes, including overdose-related complications. Management of trazodone-related pharmacodynamic interactions prioritizes discontinuation of serotonergic agents for , alongside supportive measures like benzodiazepines for and cooling for ; , a serotonin , is recommended as specific at doses of 12 mg initially followed by 8 mg every 6 hours. For QT prolongation, immediate cessation of interacting drugs and correction are essential, with hospitalization for severe cases. In CNS depression scenarios, supportive care including airway protection and for opioids is critical. Pharmacokinetic factors, such as elevated trazodone plasma levels from enzyme inhibition, may further intensify these pharmacodynamic risks.

Other Notable Interactions

Trazodone's antagonism at the can attenuate the hallucinogenic effects of serotonergic psychedelics, such as and , by blocking their primary mechanism of action. This property has led to its informal use as a "trip killer" to rapidly terminate adverse psychedelic experiences, particularly in settings where acute anxiety or arises during intoxication. Case reports document instances where trazodone administration effectively reduced hallucinatory intensity without fully abolishing potential therapeutic benefits, such as mood improvement in . Preclinical data support this interaction through trazodone's at 5-HT2A sites, though remains limited to observational cases and small trials, with no established formal contraindications for supervised use. In a notable 2023 case, a patient receiving 25 mg while on chronic trazodone experienced no perceptual alterations but achieved sustained effects, as measured by a significant drop in Montgomery-Åsberg Depression Rating Scale scores from 30 to 15 over six months. Emerging 2024-2025 research further explores trazodone's role in modulating psychedelics toward non-hallucinogenic profiles, potentially enhancing safety for psychiatric applications. Trazodone's alpha-1 adrenergic antagonism can produce additive hypotensive effects when combined with antihypertensives or alpha-blockers, potentially leading to or syncope. This interaction is documented in pharmacological databases and case series, where co-administration with agents like amplified reductions, necessitating dose adjustments in vulnerable patients. Evidence draws from observational data and studies of receptor binding, without formal contraindications but with recommendations for surveillance.

Pharmacology

Pharmacodynamics

Trazodone is classified as a (SARI), primarily exerting its effects through inhibition of the () and antagonism at various serotonin receptors. It inhibits serotonin reuptake with a binding affinity () of approximately 280 at , leading to increased synaptic serotonin levels. Additionally, trazodone acts as a potent at 5-HT2A receptors ( = 14 ) and 5-HT2C receptors ( = 307 ), which contributes to its modulation of serotonergic neurotransmission. Beyond targets, trazodone exhibits significant affinity for other receptors that influence its overall profile. It functions as an at alpha-1 adrenergic receptors, with Ki values ranging from 15 nM (alpha-1B) to 98 nM (alpha-1A), which can result in sedation and . Trazodone also antagonizes H1 receptors (Ki = 190 nM), promoting and contributing to its utility in sleep disorders. These interactions distinguish trazodone from selective serotonin reuptake inhibitors (SSRIs) by providing a broader pharmacological footprint. In preclinical studies, trazodone's inhibition of serotonin has been shown to elevate extracellular serotonin concentrations in the frontal of rats, enhancing tone in key brain regions. Furthermore, animal models demonstrate that trazodone reduces spontaneous and elicited , as observed in , without activity; for instance, doses of 3-20 mg/kg in mice models of decreased intensity and increased latency to attack. These effects correlate with its receptor antagonism, particularly at 5-HT2A/2C and alpha-1 sites. Pharmacodynamic modeling based on () principles estimates high target occupancy at therapeutic doses, with 5-HT2A receptor occupancy reaching 91% following a 30 mg immediate-release dose and up to 97% at steady-state 300 mg once-daily dosing. Clinically, the effects of trazodone are attributed to serotonin modulation via inhibition and 5-HT2A antagonism, while its hypnotic properties at lower doses (50-100 mg) stem from blockade of H1 and alpha-1 adrenergic receptors, often without significant inhibition at those levels.

Pharmacokinetics

Trazodone is well absorbed after , with ranging from 63% to 91%. Peak plasma concentrations (T_max) are typically reached within 1 to 2 hours post-dose, though this can be delayed to about 2 hours when taken with . Ingestion with can alter ; for extended-release formulations, it increases the maximum concentration (C_max) by up to 86%, with minimal effect on , while for immediate-release formulations, it typically slightly increases (up to 20%) and decreases C_max. Following absorption, trazodone distributes widely throughout the body, with a (V_d) of approximately 0.47 to 0.84 L/kg. It readily crosses the blood-brain barrier to exert central effects. is high, at 89% to 95%. Trazodone undergoes extensive hepatic primarily via the enzyme , yielding the m-chlorophenylpiperazine (mCPP), which has about one-tenth the potency of the parent drug. The elimination of trazodone is 5 to 9 hours in healthy adults. occurs mainly as metabolites, with 70% to 75% eliminated renally and approximately 20% via ; less than 1% of the dose is excreted unchanged in urine. variability is notable in certain populations; the can prolong to up to 13 hours in the elderly due to reduced clearance. At higher doses, pharmacokinetics may exhibit nonlinearity, potentially due to or .

Chemistry

Chemical Structure

Trazodone, chemically known as 2-{3-[4-(3-chlorophenyl)piperazin-1-yl]propyl}-[1,2,4]triazolo[4,3-a]pyridin-3(2H)-one, is a synthetic belonging to the class of serotonin antagonist and reuptake inhibitors (SARIs). Its molecular formula is C19H22ClN5O, with a molecular weight of 371.86 g/mol. The core structure features a [1,2,4]triazolo[4,3-a]pyridine ring fused system attached via a propyl linker to a ring, which in turn is substituted at the position with a 3-chlorophenyl group; this moiety is particularly important for its interactions with serotonin receptors. Trazodone is achiral, lacking any stereocenters and thus having no enantiomers. The compound is typically synthesized through a reaction between 2-(3-halopropyl)-[1,2,4]triazolo[4,3-a]pyridin-3(2H)-one and 1-(3-chlorophenyl), often facilitated under microwave-assisted conditions or phase-transfer to improve yield and purity.

Physical and Chemical Properties

Trazodone is a white to off-white, non-hygroscopic crystalline powder with a bitter and odorless properties when crystallized from . The compound exhibits limited aqueous , approximately 0.29 mg/mL at , classifying it as sparingly soluble in ; increases in acidic conditions due to its basic nature and is higher in organic solvents such as (sparingly soluble), , and (soluble up to 25 mg/mL). The of the is 6.74, contributing to pH-dependent and behavior that supports in mildly acidic to environments ( 4-7). Its octanol- partition coefficient () is 2.9, indicating moderate that enhances penetration into lipophilic tissues like the . Trazodone hydrochloride demonstrates high thermal stability, melting with decomposition between 231°C and 234°C, and remains stable under standard storage conditions of 25°C and 60% relative humidity for up to 24 months. However, it is sensitive to light and heat, necessitating storage in airtight containers below 30°C and protected from light to prevent degradation. These properties guide pharmaceutical formulations, favoring solid dosage forms like tablets for optimal bioavailability.

History and Development

Discovery and Early Research

Trazodone was developed in the 1960s by Angelini Research Laboratories, an pharmaceutical company, as part of efforts to create a new class of s known as serotonin antagonist and reuptake inhibitors (SARIs). The compound, chemically 2-[3-[4-(3-chlorophenyl)piperazin-1-yl]propyl]-[1,2,4]triazolo[4,3-a]pyridin-3(2H)-one, was first in by researchers Giorgio Palazzo and Bruno Silvestrini within Angelini's chemical laboratories. This marked the beginning of preclinical investigations aimed at identifying agents with properties that could circumvent the limitations of earlier s, such as s (TCAs) and inhibitors (MAOIs). Early preclinical research in the late focused on trazodone's pharmacological profile in models, revealing antidepressant-like effects through antiserotonin activity without the catecholamine-potentiating actions associated with TCAs, which contribute to their . These studies, led by Silvestrini and colleagues, demonstrated that trazodone enhanced serotonin-mediated selectively, avoiding the broad monoamine interactions and associated risks of MAOIs, such as hypertensive crises from tyramine interactions. Preliminary findings from these experiments were presented in at a World Psychiatric Association meeting in , highlighting trazodone's potential as a safer alternative for treating depressive symptoms. The rationale for trazodone's development emphasized a selective focus on serotonin pathways to balance antidepressant efficacy with reduced side effects, including lower relative to other agents while maintaining therapeutic modulation. Pre-approval research extended into the early 1970s, with and biochemical studies in animal models confirming its safety profile, including minimal teratogenicity and efficient metabolism. Key milestones included the granting of U.S. No. 3,381,009 in 1968 to Palazzo and Silvestrini for the compound's preparation. Trazodone was first marketed in in 1972 under the brand name Trittico by .

Clinical Development and Approval

Trazodone underwent phase III clinical trials in the and , primarily evaluating its efficacy in treating (MDD) through double-blind, randomized controlled trials (RCTs) comparing it to standard antidepressants like and . These trials, involving over 200 participants each, demonstrated that trazodone at doses of 100-400 mg/day was comparable in efficacy to , with response rates—defined as at least a 50% reduction in Hamilton Depression Rating Scale scores—typically ranging from 40% to 50%, and significantly superior to . For instance, a multicenter study confirmed trazodone's antidepressant effects were equivalent to in outpatients with moderate to severe , with both agents achieving similar remission rates after 4-6 weeks of treatment. The U.S. (FDA) approved trazodone in December 1981 for the treatment of MDD in adults, based on from these pivotal trials establishing its safety and efficacy profile. In , trazodone received marketing authorization in the early , initially in where it was developed, for the treatment of with or without anxiety, and was later approved across the (EMA) member states for similar indications in the 1980s. Post-marketing in the 1990s highlighted trazodone's utility beyond its labeled indication, leading to widespread off-label recognition for treating associated with , supported by observational showing improved onset and maintenance at lower doses (50-100 mg). No label updates were made to include as an official indication, but clinical guidelines began acknowledging its role in this context. As of 2025, trazodone has not faced major withdrawals globally, with ongoing efforts by the FDA and EMA monitoring rare adverse events like and to ensure continued safe use.

Society and Culture

Trazodone is not classified as a under the () schedules, as it lacks significant potential for abuse or dependence relative to other regulated drugs. It is available only by prescription in the , requiring a licensed healthcare provider's authorization for dispensing. Since the expiration of its original patents in 1985, trazodone has been widely available as a generic medication, substantially reducing costs and increasing accessibility. It is approved and marketed in over 50 countries worldwide, including major markets in , , , and , typically under prescription-only status. Internationally, it is prescription-only in the and , though classified as a controlled substance (Class C1) in . Key regulatory restrictions include a for concurrent use with inhibitors (MAOIs), mandating a minimum 14-day washout period to prevent . Additionally, trazodone carries a from the US Food and Drug Administration (FDA) regarding an increased risk of suicidal thoughts and behaviors, particularly in children, adolescents, and young adults during initial treatment or dosage adjustments. Globally, trazodone is not available over-the-counter in any jurisdiction and remains strictly prescription-only to ensure medical supervision. In the European Union, its use in veterinary medicine is controlled, requiring authorization and oversight by licensed veterinarians as an off-label application of a human pharmaceutical. As of 2025, there have been no changes to trazodone's legal classification or availability, with ongoing monitoring by regulatory agencies confirming its low abuse potential despite limited reports of misuse in higher doses.

Brand and Generic Names

Trazodone is the generic name for the medication, with the hydrochloride salt form employed to enhance its solubility in pharmaceutical formulations. Originally developed by Angelini Research Laboratories in during the 1960s, trazodone was first marketed under brand names such as Trittico in . In the , the primary brand name is Desyrel, while in the it is marketed as Molipaxin, and Trittico is widely used across various European countries. Internationally, trazodone is available under more than 50 brand names, including Deprax in and Thombran in , reflecting its broad adoption and adaptation in different markets. Generics now dominate the market for trazodone, comprising the vast majority of prescriptions due to expiration and widespread manufacturing by multiple pharmaceutical companies.

Usage Statistics

In the United States, approximately 24 million prescriptions for trazodone were filled in 2023, ranking it as the 21st most commonly prescribed medication overall and the second most common antidepressant. In the US, about 85% of these prescriptions are off-label, primarily for insomnia. Demographic patterns among US trazodone users show that roughly 60% are female, with peak usage occurring in the 40-60 age range and a mean age of about 49 years; approximately 70% of prescriptions are issued in outpatient settings. Prescription trends indicate a 20% increase from pre-2020 levels, driven largely by rising for sleep disturbances amid heightened awareness of during the , while prescriptions for have remained relatively stable.

Research Directions

Ongoing Clinical Investigations

As of 2025, several clinical investigations into trazodone continue to explore its and in various psychiatric conditions, with a focus on augmentation strategies and long-term use. The National Adaptive Trial for PTSD-Related (NAP), a phase 3, multi-center sponsored by the U.S. Department of , is actively recruiting participants to evaluate trazodone alongside and compared to for treating in patients with (PTSD). This adaptive design trial, planned to enroll 1,224 veterans across 34 sites, aims to identify the most effective pharmacological intervention for PTSD-associated sleep disturbances, building on preliminary evidence of trazodone's role in reducing hyperarousal and nightmares. Ongoing research also addresses trazodone's long-term safety profile for insomnia management, particularly in patients with comorbid . A multi-center (NCT07136415) is planned to compare trazodone to and digital in adults with menopause-related symptoms. Complementing this, a 2025 systematic and of confirmed trazodone's efficacy in improving sleep onset, maintenance, and overall quality in depressive patients with , while highlighting a favorable safety profile with low rates of serious adverse events in extended use up to six months, though cardiovascular monitoring remains recommended. Investigations into combination therapies for treatment-resistant depression include evaluations of augmentation strategies involving trazodone. This builds on meta-analytic evidence from 39 trials demonstrating benefits for certain combinations versus monotherapy without increased dropout risks. These findings underscore trazodone's potential to address residual and in this population. Additionally, the REST trial, a phase 2 study, is examining trazodone's effects on sleep in patients with amnestic (aMCI). Exploratory trials of trazodone for pediatric anxiety disorders remain limited due to ethical concerns regarding in children under 18, where safety data are primarily derived from small-scale pharmacokinetic modeling rather than large randomized studies. Ongoing observational efforts focus on dosing optimization for co-occurring anxiety and , with enrollment constrained by requirements for stringent risk-benefit assessments and protocols. Post-marketing surveillance registries continue to monitor cardiac events associated with trazodone, particularly prolongation and arrhythmias. A 2025 real-world analysis of the FDA Reporting System (FAERS) database identified potential signals for and , prompting recommendations for baseline ECG screening in at-risk populations; this ongoing registry tracks over 10,000 annual reports to inform updated prescribing guidelines.

Potential New Applications

Recent studies have explored trazodone's potential role in developing non-hallucinogenic psychedelics by modulating serotonin receptors to achieve mood-enhancing effects without inducing hallucinatory experiences. A 2025 systematic review by Chen et al. analyzed preclinical and clinical data, highlighting how trazodone, as a 5-HT2A antagonist, can block the hallucinogenic properties of compounds like psilocybin while preserving antidepressant benefits, suggesting its utility in safer psychedelic-inspired therapies for mood and anxiety disorders. This approach aligns with emerging research on psychoplastogens that promote neuroplasticity without perceptual alterations. An ongoing trial (NCT07210112) is evaluating the combination of psilocybin and trazodone for treatment-resistant depression. In the realm of management, particularly , preclinical evidence indicates trazodone's synergistic potential when combined with analgesics, though human trials remain preliminary. Animal and early human studies, including a 2011 with , demonstrate that trazodone enhances relief and sleep quality in , showing amplified symptom reduction. For alcohol use disorder, trazodone shows promise as an aid in managing withdrawal symptoms through serotonin stabilization, supported by early clinical evidence. Clinical extensions from these models suggest its role in post-withdrawal stabilization, though primarily tested in smaller cohorts. Trazodone's neuroprotective potential in Parkinson's disease stems from its 5-HT2A antagonism, which may preserve dopaminergic function, as hypothesized in pharmacological models. Preclinical data indicate that blocking 5-HT2A receptors with trazodone enhances dopamine-dependent behaviors and motor function in rat models of parkinsonism, potentially mitigating neurodegeneration. This hypothesis is bolstered by observations of improved depressive and motor symptoms in Parkinson's patients treated with trazodone. Despite these prospects, significant research gaps persist, including the need for larger-scale clinical trials to validate and across applications. Long-term data on trazodone's effects in psychedelic modulation and contexts remain incomplete, with calls for extended studies to assess neurocognitive outcomes and sustained benefits. also positions trazodone as a "trip-killer" to abort adverse psychedelic experiences by antagonizing 5-HT2A receptors.

Veterinary Applications

Uses in Animals

Trazodone is primarily used in to manage anxiety and stress-related behaviors in and , including separation anxiety, noise phobias such as fear of or thunderstorms, and situational stressors like veterinary visits or . In , it serves as an effective adjunct for short-term anxiolysis during events that trigger fear responses, while in , it is employed off-label to alleviate transport- and examination-induced anxiety. This application draws from its established role in human anxiety treatment, adapted for similar behavioral calming effects in pets. Emerging off-label use extends to horses, where trazodone has shown potential for and reduction, particularly in hospitalized or post-surgical settings and for managing acute by decreasing ambulation. Studies indicate oral doses of 5-7.5 mg/kg every 12 hours reduce step frequency by approximately 44% without affecting recumbency, though it may increase requirements for anesthesia. Typical dosing regimens for range from 3 to 10 mg/kg orally every 24 hours, adjusted based on the severity of anxiety and individual response, with effects onsetting within 1 to 2 hours for acute situations. For , a total dose of 25 to 50 mg orally is commonly administered as a single dose prior to stressful events, such as veterinary appointments, to promote sedation without significant physiological changes like alterations in . These dosages are prescribed off-label, as trazodone lacks formal FDA approval for veterinary use, and administration should occur under veterinary supervision to ensure safety and efficacy. Veterinary studies demonstrate substantial , with one client-reported evaluation indicating that approximately 89% of dogs (32 out of 36) experienced moderate to extreme calming benefits from trazodone during pre-appointment anxiety management. In a randomized crossover involving cats, a single 50 mg dose significantly reduced anxiety signs during transport and improved handling ease during examinations, as scored by both veterinarians and owners (P < 0.05). For dogs, clinical trials have shown reductions in stress behaviors such as panting, whining, and frenetic activity post-administration, supporting its role in behavioral trials for situational anxiety. Trazodone is often combined with fluoxetine, a selective serotonin reuptake inhibitor, to enhance treatment of aggression and anxiety-related behaviors in dogs, particularly when monotherapy is insufficient. It is also frequently co-administered with gabapentin for acute situational anxiety in dogs and cats, such as during veterinary visits; recent studies (as of 2025) show this combination reduces stress signs more effectively than either alone, with gabapentin dosed at 10-20 mg/kg orally 1-2 hours prior. In a study of canine aggression cases, adjunctive trazodone use alongside behavioral therapy did not increase aggressive incidents and contributed to overall improvement without notable adverse behavioral shifts. Such combinations require careful monitoring to mitigate risks like serotonin syndrome, starting at low doses and titrating gradually.

Safety and Considerations in Veterinary Medicine

In veterinary medicine, trazodone is associated with several potential adverse effects in dogs and cats, with sedation being the most common, reported in approximately 43% of cases involving adverse reactions. Ataxia occurs in about 16% of affected dogs, while gastrointestinal upset, including vomiting, is noted in around 14%. Priapism, a rare but persistent penile erection, has been observed infrequently in male dogs, though it did not occur in several clinical studies monitoring its use. These side effects are generally mild and resolve with dose adjustment or discontinuation, but serotonin syndrome—manifested as vomiting, seizures, or hyperthermia—can arise when trazodone is combined with other serotonergic medications. Trazodone is contraindicated in animals with known to the drug, those receiving inhibitors, or pets with angle-closure due to its potential to exacerbate . It should also be avoided in patients with significant cardiac , as the can cause arrhythmias or , and used with extreme caution in those with hepatic or renal impairment, where clearance may be prolonged. Caution is advised in breeding animals, particularly intact males, owing to the rare risk of that could interfere with reproductive function, and in pregnant animals due to limited safety data. Monitoring protocols for trazodone in veterinary patients emphasize regular assessment of behavioral response to ensure therapeutic efficacy without excessive . For long-term use, liver enzyme levels should be evaluated every 3 to 6 months, particularly in dogs with pre-existing hepatic concerns, given case reports of potential . Complete blood counts and serum chemistry panels may also be warranted periodically to detect any organ function changes. Pharmacokinetically, trazodone exhibits a shorter elimination in dogs compared to s, approximately 2.8 hours following oral or intravenous , with high of about 85%. This rapid clearance necessitates dosing adjustments for sustained effects, especially in species like where is similarly quick but are more limited. Veterinary guidelines recommend initiating trazodone at the lowest effective dose to minimize and other adverse effects, with gradual based on individual response. Overdose management aligns closely with protocols, focusing on supportive care such as intravenous fluids, for tremors or seizures, and monitoring for , with no specific available.