Chlorprothixene is a low-potency, first-generation typical antipsychotic medication belonging to the thioxanthene class, primarily used for the treatment of schizophrenia and other psychotic disorders.[1][2] It was first marketed in 1959 under trade names such as Taractan and Truxal, and remains available in 16 countries despite the advent of newer antipsychotics.[3][2]As a thioxanthene derivative, chlorprothixene exhibits antipsychotic effects through its blockade of dopamine D1, D2, and D3 receptors, alongside antagonism of serotonin 5-HT2, histamine H1, muscarinic acetylcholine, and alpha-1 adrenergic receptors.[1] This multifaceted receptor binding profile also contributes to its sedative and antiemetic properties, making it useful beyond psychosis management.[1] The drug depresses activity in the hypothalamic-hypophyseal hormone system and the reticular activating system, which underlies its calming effects on the central nervous system.[1] Compared to chlorpromazine, a benchmarkantipsychotic, chlorprothixene demonstrates approximately half to two-thirds the potency, requiring higher doses for equivalent therapeutic effects.[1]Clinically, chlorprothixene is indicated for acute and chronic psychoses, including schizophrenia, and for managing manic episodes in bipolar disorder.[1] It is also employed off-label at low doses for its sedative-hypnotic properties, particularly in treating insomnia, anxiety, and neuroses due to its favorable tolerability profile at these levels.[2] Administration is typically oral, with dosages ranging from 25-400 mg daily for antipsychotic effects, though it is contraindicated in patients with circulatory collapse, coma, or hypersensitivity to the drug.[1]Common side effects of chlorprothixene mirror those of other typical antipsychotics, including drowsiness, dizziness, muscle tremors, and extrapyramidal symptoms such as dystonia and akathisia.[1] It carries a risk of cardiometabolic adverse events, with studies showing an increased hazard of diabetes (adjusted HR 1.34) and major cardiovascular events (adjusted HR 1.12) compared to low-dose quetiapine, particularly at cumulative doses exceeding 6000 mg for diabetes and 1500 mg for cardiovascular risks.[2] Allergic reactions and liver damage occur less frequently than with chlorpromazine, but monitoring for hematologic effects like leukopenia is recommended.[1] Despite its long history, chlorprothixene's use has declined in favor of atypical antipsychotics with lower side effect burdens, though it retains a niche role in certain clinical settings.[2]
Medical Uses
Approved Indications
Chlorprothixene is approved as a typical antipsychotic for the treatment of psychotic disorders, including schizophrenia and other related psychoses, where it helps alleviate symptoms such as hallucinations, delusions, and thought disorders.[4][2] Clinical trials have demonstrated its efficacy in reducing these core psychotic symptoms, with early double-blind studies showing significant improvements in patients with prominent delusions and hallucinations compared to placebo.[5][6]It is also indicated for the management of acute mania in bipolar disorder, where it stabilizes mood and reduces agitation during manic episodes.[4][7] Regulatory approvals in European countries, such as Denmark and the Netherlands, support its use in these conditions based on established antipsychotic activity.[2][7]For severe anxiety and agitation, chlorprothixene is approved in certain jurisdictions, leveraging its sedative properties to promote calming effects.[7][8]Dosing guidelines typically start with 15-30 mg orally per day, divided into 2-3 doses, and may be titrated up to a maximum of 400 mg per day based on response and tolerability.[9] For acute agitation, intramuscular administration of 50-100 mg may be used, with subsequent transition to oral therapy.[9] These regimens are derived from national product monographs and clinical recommendations in approving regions.[7]
Off-Label Uses
Chlorprothixene has been employed off-label in low doses as a sedative-hypnotic agent for the treatment of insomnia, particularly in cases where standard therapies like benzodiazepines prove ineffective.[10] This application leverages its calming properties to promote sleep without the dependency risks associated with traditional hypnotics, though long-term use raises concerns about cardiometabolic side effects.[10]In palliative care settings, chlorprothixene serves as an adjunctive therapy for managing nausea and vomiting, drawing on its potent antiemetic properties akin to other typical antipsychotics.[11] This off-label role is particularly relevant for refractory symptoms in advanced illness, where dopamine antagonism helps control emetic pathways, though evidence remains largely observational rather than from large-scale trials.[11]Chlorprothixene has shown utility in the management of alcohol withdrawal symptoms, including delirium tremens and associated psychoses, due to its sedative and antipsychotic effects that mitigate agitation and hallucinations.[12]Pediatric applications of chlorprothixene are limited and approached with caution owing to insufficient efficacy and safety data in children; nonetheless, it has been investigated for irritability and behavioral disturbances in children with behavior disorders.[13] Early studies indicate modest benefits in disturbed children with behavior disorders, but risks such as hypotension and extrapyramidal symptoms necessitate careful monitoring and low dosing.[14]As a co-medication in chronic pain management, chlorprothixene exhibits potential despite unproven direct analgesic or antidepressant mechanisms, with reports of pain relief in about one-third of patients with intractable conditions like post-herpetic neuralgia.[15] This effect may stem from its modulation of central pain pathways, as evidenced in small uncontrolled trials where rapid alleviation occurred in most cases within days.[11]Historically, chlorprothixene found use in treating neuroses, valued for its sedative effects and low toxicity profile compared to earlier antipsychotics.[16] This application, prominent in the mid-20th century, highlighted its role in calming anxious or reactive states without severe adverse reactions, influencing its off-label adoption in milder psychiatric conditions.[16]
Safety and Tolerability
Side Effects
Chlorprothixene, a typical antipsychotic, is associated with a range of adverse effects that are generally dose-dependent and more pronounced during the initial phases of treatment. These side effects share similarities with those of other low-potency antipsychotics, such as chlorpromazine, but allergic reactions and hepatotoxicity occur less frequently.[1][4]Common side effects include strong sedation manifesting as somnolence and dizziness, which affect more than 10% of patients, as well as anticholinergic symptoms such as dry mouth, constipation, blurred vision (accommodation disorder), and urinary retention.[7] Cardiovascular effects like tachycardia and orthostatic hypotension are also frequent, occurring in 1-10% of users, alongside weight gain and increased appetite due to metabolic changes.[7][2]Extrapyramidal symptoms (EPS), including akathisia, dystonia, and parkinsonism, are reported but at a lower incidence compared to high-potency phenothiazines, with dystonia being common (1-10%) and akathisia or parkinsonism uncommon (0.1-1%).[7] Other notable effects encompass sexual dysfunction such as erectile dysfunction and decreased libido (uncommon), as well as rare instances of neuroleptic malignant syndrome (NMS), a potentially life-threatening condition involving hyperthermia, muscle rigidity, and autonomic instability.[7]In special populations, elderly patients exhibit heightened sensitivity to chlorprothixene, with increased risks of falls due to sedation and orthostatic hypotension, as well as confusion from anticholinergic effects.[7] Compared to chlorpromazine, chlorprothixene carries a reduced likelihood of allergic reactions or liver damage, making it somewhat better tolerated in this regard.[1]Monitoring recommendations include regular assessment for EPS through clinical evaluation and for metabolic changes such as weight gain and glucose intolerance, particularly in long-term use, to mitigate cardiometabolic risks.[7][2]
Contraindications
Chlorprothixene is absolutely contraindicated in patients with hypersensitivity to the drug, other thioxanthenes, or any excipients, as well as in those with cross-sensitivity to phenothiazines or thiothixene.[7][1] It is also contraindicated in cases of circulatory collapse, depressed level of consciousness (such as from alcohol, barbiturates, or opiates), coma, or blood dyscrasias including diseases of the hematopoietic system.[7][1]The drug is contraindicated in patients with clinically significant cardiovascular disorders, including congestive heart failure, cardiac decompensation, coronary artery disease, cerebral vascular disorders, recent myocardial infarction, uncompensated heart failure, cardiac hypertrophy, or arrhythmias treated with class 1A or III antiarrhythmics, due to the risk of hypotension and QT prolongation.[7][1] Additionally, it is contraindicated in individuals with a history of ventricular arrhythmias or Torsade de Pointes, congenital long QT syndrome, acquired QT prolongation (QTc >450 ms in males or >470 ms in females), or uncorrected hypokalaemia or hypomagnesaemia.[7]Other absolute contraindications include acute narrow-angle glaucoma, particularly in patients with a shallow anterior chamber, where anticholinergic effects may precipitate acute attacks.[7] Chlorprothixene should not be used concurrently with large amounts of alcohol or other central nervous system depressants, as this can lead to severe CNS depression or coma.[7][1]Relative contraindications include epilepsy, as chlorprothixene can lower the seizure threshold, and Parkinson's disease, where it may worsen extrapyramidal symptoms.[7] Use with caution in patients with severe hepatic or renal impairment. Use during pregnancy is not recommended unless the potential benefit justifies the risk to the fetus (FDA pregnancy category C).[7][17]In breastfeeding, chlorprothixene is excreted into breast milk in small amounts, and its use should be avoided if possible, with close monitoring of the infant for adverse effects if administration is necessary.[7][18]
Overdose
Chlorprothixene overdose can manifest as extreme sedation progressing to coma, along with confusion, hypotension, tachycardia, respiratory depression, seizures, and cardiac arrhythmias such as ventricular extrasystoles or fibrillation.[7][19] Other features include muscle trembling or jerking, extrapyramidal symptoms, hyperthermia or hypothermia, and in severe cases, renal impairment or QT prolongation potentially leading to torsades de pointes.[4][7]Therapeutic blood concentrations of chlorprothixene typically range from 0.02 to 0.3 mg/L, with toxicity emerging above 0.4 mg/L and fatalities reported at postmortem levels from 0.1 to 7.0 mg/L, including one case at 0.90 mg/L where chlorprothixene was the primary cause.[20] Oral doses up to approximately 0.4 mg/kg are generally tolerated in therapeutic contexts for adults, but intakes exceeding 1 g often result in severe intoxication, with fatal doses estimated at 2.5–4 g in adults and around 4 mg/kg in infants.[21][7] Higher doses precipitate cardiovascular collapse, including cardiodepression and shock.[19]Management is entirely supportive, as no specific antidote exists, and focuses on immediate decontamination via gastric lavage and administration of activated charcoal if ingestion occurred recently.[7] Vital signs require close monitoring, with mechanical ventilation for respiratory depression, intravenous fluids and vasopressors (avoiding epinephrine due to risk of further hypotension) for cardiovascular instability, and continuous ECG surveillance for arrhythmias.[7][19] Seizures should be treated with benzodiazepines such as diazepam, while extrapyramidal reactions may respond to anticholinergics like biperiden; in refractory cases, extracorporeal methods like hemoperfusion have facilitated drug elimination and recovery.[7][19]Prognosis is favorable with early intervention, as evidenced by survivals following ingestions of up to 10 g in adults and 1 g in children, though the drug's elimination half-life of 8–12 hours (potentially longer in the elderly) can prolong toxic effects and complicate recovery.[7][20] Delays in treatment increase risks of cardiorespiratory arrest or secondary complications like pneumonia from prolonged respiratory depression.[19]
Drug Interactions
Chlorprothixene, as a typical antipsychotic, exhibits significant pharmacokinetic and pharmacodynamic interactions with various medications, primarily due to its effects on the central nervous system (CNS) and hepatic metabolism.[4] Concomitant use with CNS depressants such as opioids, benzodiazepines, and alcohol can lead to additive sedation and respiratory depression, necessitating careful monitoring and potential dose reductions of the CNS depressant to mitigate risks.[4][7]Interactions with anticholinergic agents, including tricyclic antidepressants and antiparkinsonian drugs, can enhance anticholinergic effects, potentially resulting in severe dry mouth, constipation, or paralytic ileus.[4][7] These amplified anticholinergic side effects underscore the need for caution in polypharmacy scenarios involving such agents.[4]Co-administration with lithium increases the risk of lithium neurotoxicity, requiring close monitoring of lithium plasma levels to prevent toxicity.[7] Similarly, combining chlorprothixene with tramadol heightens the risk of CNS depression and may lower the seizure threshold, potentially precipitating serotonin syndrome or seizures.[4]Chlorprothixene undergoes hepatic metabolism primarily via the cytochrome P450 (CYP) 2D6 enzyme; inhibitors such as fluoxetine can elevate chlorprothixene plasma levels, increasing the risk of adverse effects, while inducers like carbamazepine may decrease its efficacy by accelerating metabolism.[7][4]Clinical recommendations include dose adjustments for interacting agents, particularly opioids and CYP modulators, along with routine ECG monitoring for QT prolongation when used with other antipsychotics or QT-prolonging drugs to avoid cardiac arrhythmias.[7][4]
Pharmacology
Pharmacodynamics
Chlorprothixene, a typical antipsychotic of the thioxanthene class, primarily mediates its therapeutic antipsychotic effects through antagonism of dopamine D1 and D2 receptors in the mesolimbic pathway of the brain, which reduces excessive dopaminergic activity associated with psychotic symptoms such as hallucinations and delusions.[4] This blockade inhibits postsynaptic dopamine signaling, thereby alleviating positive symptoms of schizophrenia.[4] Additionally, chlorprothixene depresses the release of hypothalamic and hypophyseal hormones, contributing to its broader effects on neuroendocrine function.[4]The drug exhibits strong antagonism at multiple other receptor types, including serotonin 5-HT<sub>2A</sub> receptors (which mitigates extrapyramidal side effects by balancing dopamine D<sub>2</sub> blockade), histamine H<sub>1</sub> receptors (responsible for sedative properties), muscarinic acetylcholine receptors (leading to anticholinergic effects like dry mouth and constipation), and α<sub>1</sub>-adrenergic receptors (associated with orthostatic hypotension).[4][1] These interactions underlie both its therapeutic benefits and adverse effects profile. Binding affinities reflect this potency, with approximate K<sub>i</sub> values of 18 nM at D<sub>1</sub>, 2.96 nM at D<sub>2</sub>, 4.56 nM at D<sub>3</sub>, 9 nM at D<sub>5</sub>, and 3.75 nM at H<sub>1</sub> receptors, as determined from standardized screening assays. For 5-HT<sub>2A</sub>, affinities are in the range of 0.3–0.4 nM, indicating high potency.[22]Chlorprothixene's receptor binding profile provides a relatively balanced antagonism, where concomitant 5-HT<sub>2A</sub> and muscarinic blockade counteracts the propensity for extrapyramidal symptoms (EPS) that might otherwise arise from selective D<sub>2</sub> antagonism, resulting in milder motor side effects compared to some other typical antipsychotics.[4] Furthermore, D<sub>2</sub> receptor blockade in the pituitary lactotroph cells competes with endogenous dopamine's inhibitory action on adenylate cyclase, leading to elevated prolactin secretion and potential endocrine effects such as galactorrhea or menstrual irregularities.
Pharmacokinetics
Chlorprothixene exhibits incomplete oral bioavailability, with absolutebioavailability of approximately 17% for the oral solution due to significant presystemic metabolism.[23] Relative to the oral solution, bioavailability is about 56% for coated tablets and 68% for suspensions.[23] Peak plasma concentrations are typically reached within 2 to 5 hours following oral administration.[8] Intramuscular administration results in faster onset of effects, occurring within 10 to 30 minutes.[1]The drug is highly bound to plasma proteins, with binding rates exceeding 95%.[24] Chlorprothixene has a large apparent volume of distribution at steady state, approximately 1035 L, indicating extensive tissue distribution.[1] As a lipophilic antipsychotic, it readily crosses the blood-brain barrier to exert central effects.[4]Chlorprothixene undergoes hepatic metabolism primarily via cytochrome P450 enzymes, including CYP2D6 and CYP3A4.[25] Key metabolic pathways include N-demethylation to the active metabolite N-desmethylchlorprothixene, as well as sulfoxidation to chlorprothixene sulfoxide and epoxidation.The elimination half-life of chlorprothixene is 8 to 12 hours.[4] Excretion occurs mainly through feces and urine, with free chlorprothixene and its sulfoxidemetabolite detectable in both.[1] Genetic polymorphisms in CYP2D6 can influence clearance rates, potentially leading to prolonged half-life in poor metabolizers.[25] Steady-state plasma concentrations are generally achieved after 3 to 5 days of regular dosing.[4]
Chemistry
Structure and Properties
Chlorprothixene is a thioxanthene derivative characterized by the molecular formula C₁₈H₁₈ClNS and a molecular weight of 315.86 g/mol.[1][4] Its molecular structure consists of a tricyclic thioxanthene core, formed by two benzene rings fused to a central dibenzo-1,4-thiazepine ring containing a sulfur atom, with a chlorine substituent at the 2-position and a (Z)-3-(dimethylamino)propylidene side chain attached at the 9-position.[1][4] This configuration contributes to its classification as a typical antipsychotic within the thioxanthene class.[1]In its pure form, chlorprothixene appears as a white to pale yellow crystalline powder.[26] Key physical properties include a melting point of 97–98 °C for the free base, a boiling point of 160 °C at 0.04 Torr, an estimated density of 1.1048 g/cm³, and an estimated refractive index of 1.6000.[26] These attributes reflect its solid-state behavior under standard laboratory conditions.[26]Chlorprothixene exhibits limited solubility in water, with a reported value of approximately 0.386 mg/L at 22.5 °C, rendering it slightly soluble, while it demonstrates good solubility in organic solvents such as methylene chloride and ethanol.[26][1] It possesses a pKₐ of 9.3, indicating basic character due to the tertiary amine group in the side chain.[4] Regarding stability, chlorprothixene is light-sensitive and prone to degradation in air, necessitating storage in airtight, light-protected containers to maintain integrity.[1]
Synthesis
Chlorprothixene is synthesized primarily through the construction of the thioxanthene core followed by attachment and modification of the side chain at the 9-position. The core begins with the reaction of 2-mercaptobenzoic acid and 1-bromo-4-chlorobenzene to form 2-(4-chlorophenylthio)benzoic acid, which is then converted to the acid chloride using phosphorus pentachloride and undergoes intramolecular cyclization in the presence of aluminum chloride to yield 2-chlorothioxanthone.[27]The side chain is introduced via a Grignard reaction of 2-chlorothioxanthone with 3-(dimethylamino)propylmagnesium bromide, producing a tertiary alcohol intermediate. Dehydration of this alcohol is achieved through acylation with acetyl chloride to form the acetateester, followed by pyrolysis, resulting in the exocyclic double bond characteristic of chlorprothixene. An alternative route involves reaction of the thioxanthone with allylmagnesium bromide to form an allylic alcohol, which is chlorinated using thionyl chloride to generate a diene intermediate; subsequent addition of dimethylamine at elevated temperatures completes the side chain.[27]The chlorine substituent at the 2-position (beta to the sulfur in the central ring) of the thioxanthene core enhances antipsychotic activity compared to unsubstituted analogs, as determined from structure-activity relationship studies of thioxanthene derivatives. Structural variations, such as modifications to the side chain or ring substitutions, have led to related compounds like clopenthixol, which retains the core but alters the amine functionality for improved potency. The exocyclic double bond at C9 exhibits E/Z isomerism, with the Z-isomer generally showing higher neuroleptic activity.[28][29]This synthesis was developed by Lundbeck, the Danish pharmaceutical company that first introduced chlorprothixene in 1959.[29]
History
Development
Chlorprothixene was developed in the 1950s by the Danish pharmaceutical company H. Lundbeck & Co. as a structural analog of phenothiazine antipsychotics, particularly chlorpromazine, which had revolutionized psychiatric treatment earlier that decade. The project was spearheaded by chemist Poul Viggo Petersen, who aimed to create a new class of neuroleptics with enhanced tolerability by modifying the central ring of the phenothiazine scaffold—replacing the nitrogen atom with a carbon-carbon double bond to form the tricyclic thioxanthene structure while retaining the sulfur atom. This structural innovation was intended to preserve potent antipsychotic effects while mitigating the toxicity and side effect burden associated with phenothiazines, such as pronounced sedation and autonomic disturbances.[30][16]Synthesis of chlorprothixene occurred in the late 1950s, marking it as the prototype for the thioxanthene class of antipsychotics. Lundbeck filed a patent application for the compound in 1958, securing intellectual property ahead of competitors like Merck and Roche, who pursued similar structures through alternative synthetic routes. Initial preclinical screening focused on antipsychotic potential using established animal models prevalent in psychopharmacology at the time, including antagonism of amphetamine-induced stereotypies and hyperactivity in rodents, which served as proxies for dopamine-mediated psychotic behaviors. These tests confirmed chlorprothixene's antipsychotic potential by antagonizing amphetamine-induced stereotypies and hyperactivity in rodents, behaviors associated with dopaminergic hyperactivity in psychotic models.[30][31][16]Preclinical evaluations further highlighted chlorprothixene's favorable profile, demonstrating effective dopamine blockade alongside reduced induction of catalepsy—a rodent model of extrapyramidal side effects—in comparison to phenothiazines. This suggested a lower risk of motor disturbances and overall toxicity, with acute toxicity studies in animals indicating a therapeutic index superior to chlorpromazine. Such findings positioned chlorprothixene as a promising advancement, paving the way for its recognition as the inaugural thioxanthene antipsychotic and influencing subsequent developments in the class.[16][32]
Clinical Introduction
Chlorprothixene underwent initial human clinical trials in the late 1950s and early 1960s, primarily evaluating its efficacy in treating schizophrenia and other psychotic disorders. Phase I and II studies demonstrated its antipsychotic effects, with early reports indicating symptom reduction in patients with schizophrenia, including alleviation of hallucinations and agitation. Notably, these trials highlighted chlorprothixene's favorable side effect profile compared to contemporaries like chlorpromazine, showing efficacy in schizophrenia while producing fewer extrapyramidal symptoms (EPS), such as dystonia and parkinsonism.[33]The drug received regulatory approval and was introduced in Europe in 1959 by Lundbeck under the brand name Truxal, marking it as one of the early thioxanthene-class antipsychotics available for clinical use. In the United States, it was approved by the FDA and marketed as Taractan by Roche Laboratories starting in 1962, initially for the management of psychotic disorders including schizophrenia.[34]Early adoption of chlorprothixene was rapid in psychiatric practice during the 1960s, particularly for acute psychoses, schizophrenia, and manic episodes, where its strong sedative properties provided rapid calming effects without excessive motor side effects. Key studies from this period, including comparative trials against chlorpromazine, confirmed these sedative benefits, supporting its use in agitated patients and contributing to its widespread application in inpatient settings. As an established first-generation antipsychotic, chlorprothixene has seen no major updates in research or indications since the 1960s, reflecting its role as a mature therapeutic option. In the US, Taractan was discontinued in 1997 amid the rise of newer atypical antipsychotics with improved tolerability profiles.[33][35]
Society and Culture
Brand Names
Chlorprothixene is primarily marketed under the brand name Truxal by H. Lundbeck A/S, available in multiple European countries including Austria, Denmark, Finland, Hungary, Iceland, Latvia, Lithuania, the Netherlands, Norway, and Sweden.[36] This brand is formulated as film-coated tablets in strengths such as 15 mg, 25 mg, and 50 mg.[4] Other historical and regional brand names include Taractan (by Roche), which was used in the United States until its discontinuation due to lack of market success, Cloxan (by Orion), and Clothixen (by Yoshitomi).[4]Generic forms of chlorprothixene are widely available as chlorprothixene hydrochloride, most commonly in oral tablet formulations with strengths ranging from 15 mg to 50 mg.[4]In the United States, no active brand names exist following the withdrawal of Taractan, and the drug is not approved for use.[4]
Availability
Chlorprothixene is widely available across Europe, where it is authorised in multiple countries including Austria, Denmark, Estonia, Finland, Hungary, Iceland, Latvia, Lithuania, the Netherlands, Norway, and Sweden, as well as Germany and the United Kingdom, available in 16 countries as of 2025 including Ukraine.[36][37][38][39] It is commonly prescribed in Denmark and Germany for psychiatric conditions, and in the UK it is accessible via imported formulations such as Truxal tablets from Denmark.[40][41]In other regions, chlorprothixene is available in parts of Asia, such as Thailand, and in various countries in Latin America, though access is more limited compared to Europe.[42][4] Availability in Canada and Australia remains restricted, with no widespread marketing of finished pharmaceutical products.[4]In the United States, chlorprothixene was previously marketed under the brand name Taractan by Roche but was discontinued in the 1990s due to lack of commercial success, and it is no longer available for prescription.[4]Common formulations include oral tablets in strengths of 15 mg, 25 mg, 50 mg, and 90 mg in some formulations; oral solutions, such as 4% preparations suitable for pediatric use; and intramuscular injections at 25 mg/mL.[4][43][44] Brand names like Truxal facilitate access in authorised markets.[37]As an established older medication, chlorprothixene maintains stable supply chains globally, with no reported shortages as of 2025.[45]
Legal Status
Chlorprothixene is classified as a prescription-only medication (Rx-only) worldwide in jurisdictions where it remains available.[4]In the European Union, chlorprothixene is authorized as a nationally authorized medicinal product and requires a prescription for dispensing.[36] In India, it falls under Schedule H of the Drugs and Cosmetics Rules, 1945, mandating sale only upon presentation of a valid prescription from a registered medical practitioner.[46] In the United Kingdom, it is designated as a Prescription Only Medicine (POM), restricting its supply to prescriptions issued by authorized healthcare professionals.[38]In Brazil, chlorprothixene is categorized as Class C1 among other controlled substances, subjecting it to specific regulatory oversight for possession and distribution. In the United States, chlorprothixene is not scheduled under the Controlled Substances Act administered by the Drug Enforcement Administration, though it has been withdrawn from the market and is no longer commercially available.[1]While used historically in veterinary medicine for sedation, it is prohibited in equine sports and competitions in certain jurisdictions due to anti-doping regulations. Import and export are subject to controls in various nations, often requiring licenses due to its classification as a regulated pharmaceutical.[47][48]As of November 2025, no regulatory changes to chlorprothixene's status have been reported globally. It continues to be monitored for abuse potential, particularly owing to its pronounced sedative properties that may contribute to misuse in vulnerable populations.[49] This contrasts with its ongoing availability in Europe compared to its discontinuation in the US.[1]