Fact-checked by Grok 2 weeks ago

Haloperidol

Haloperidol is a first-generation medication that primarily exerts its therapeutic effects by antagonizing D2 receptors in the , achieving maximum efficacy at approximately 72% receptor occupancy. Developed as part of a research effort into potent analgesics at Janssen Pharmaceutica in , it was synthesized on February 11, 1958, by and rapidly progressed to clinical trials due to its unexpected neuroleptic properties observed in animal models, where it induced rather than analgesia. Marketed under the brand name Haldol® starting in October 1959, haloperidol quickly became a cornerstone of psychiatric treatment for its efficacy in managing acute and chronic psychotic symptoms, marking a pivotal advancement in 20th-century . Approved by the U.S. for , (to control motor and vocal tics), and severe behavioral disorders or hyperactivity in children when other interventions fail, haloperidol is available in oral, injectable, and long-acting depot formulations for flexible administration. It is also employed off-label for conditions such as agitation in , acute , , and intractable hiccups, reflecting its broad utility in both psychiatric and non-psychiatric settings. Therapeutic serum levels typically range from 1 to 10 ng/mL, with dosing initiated low and titrated based on response to minimize risks. While effective, haloperidol is associated with significant adverse effects, including such as , , , and , as well as , , and QT interval prolongation that can lead to cardiac arrhythmias. It is contraindicated in patients with or due to exacerbation of motor symptoms, and caution is advised in those with a history of seizures, QT prolongation, or concurrent use of other QT-prolonging drugs. Additionally, its use in elderly patients with dementia-related is not recommended owing to an increased risk of mortality, primarily from cardiovascular or infectious causes. Despite these limitations, haloperidol remains widely prescribed globally for its rapid onset and established role in and long-term management of .

Medical uses

Schizophrenia and other psychoses

Haloperidol, a first-generation , exerts its therapeutic effects in primarily through strong antagonism of D2 receptors in the , which reduces hyperactivity associated with positive symptoms such as hallucinations and delusions. This mechanism underlies its efficacy in alleviating core psychotic features, though it may have limited impact on negative symptoms like social withdrawal. Clinical trials since the 1960s have demonstrated haloperidol's superiority over in reducing psychotic symptoms and preventing relapse in . For instance, early double-blind studies showed significant improvements in symptom scores on scales like the Brief Psychiatric Rating Scale, with response rates exceeding 70% compared to 20-30% for . Typical oral dosing for ranges from 2 to 20 mg per day, titrated based on response and tolerability, often starting at 1-5 mg daily and divided into multiple administrations. Haloperidol is approved for maintenance therapy in to sustain remission and reduce hospitalization risk, as well as for managing acute in psychotic episodes, where intramuscular formulations provide rapid symptom control. In comparisons with atypical antipsychotics like , haloperidol shows similar overall efficacy for prevention but higher rates of in long-term studies (e.g., 60% vs. 34% at one year). Historically, haloperidol became a first-line agent in the 1960s for chronic management in institutional settings, revolutionizing care by enabling deinstitutionalization through effective symptom control. While effective, its use is tempered by risks of , as detailed elsewhere.

Tourette syndrome and agitation

Haloperidol was approved by the U.S. (FDA) for the treatment of tics in in 1969 for adults and in 1978 for children, marking it as one of the first medications indicated for this neurological disorder. The drug's efficacy in reducing motor and vocal tics stems from its potent D2 receptor blockade, which modulates the hyperactivity implicated in tic generation. Controlled clinical trials have demonstrated significant tic suppression with haloperidol, with one double-blind study of 57 patients showing a 70% reduction in tic severity achieved in 64% of participants treated with the drug compared to . Typical oral dosages for range from 0.5 to 5 mg per day, administered in divided doses, with adjustments based on response and tolerability; lower doses (e.g., 0.5-2 mg/day) are often sufficient for moderate symptoms, while higher doses up to 5 mg/day may be needed for severe cases. Haloperidol is generally reserved for patients with severe, refractory tics that do not respond to behavioral therapies such as or comprehensive behavioral intervention for tics. Long-term use requires careful monitoring for extrapyramidal side effects, including the risk of , which has been reported in patients on stable haloperidol doses. Haloperidol is also FDA-approved for the short-term of severe behavioral disorders in children aged to 12 years, including hyperactivity and combative, explosive, or aggressive behavior that does not respond to or other medications. Typical oral doses for these pediatric indications start at 0.5 to 1.5 mg/day (up to 0.1 mg/kg/day), titrated to 2 to 6 mg/day based on response, with careful monitoring for and other adverse effects. In managing acute , haloperidol is employed for severe behavioral disturbances in conditions such as or , offering rapid symptom control through its and properties. Intramuscular () of 2 to 5 provides quick onset for acute episodes, with repeat doses every 4 to 8 hours as needed until oral can be initiated. This approach is particularly useful in emergency psychiatric settings where immediate is required, though it is not FDA-approved specifically for -related due to increased mortality risks in elderly patients.

Delirium and other indications

Haloperidol is employed off-label for the management of agitated in (ICU) and settings, typically administered intravenously at low doses of 0.5 to 2 mg as needed for symptom control. In patients with advanced cancer, haloperidol at doses of 0.5 to 5 mg per day orally or subcutaneously has demonstrated efficacy in reducing symptoms, including , as measured by the Delirium Rating Scale, without causing excessive . A 2024 multicenter trial involving patients with advanced cancers found that scheduled intravenous haloperidol combined with every 4 hours significantly decreased agitated severity at 24 hours compared to alone or , with fewer rescue doses required and minimal additional . In , haloperidol serves as an anti-emetic for opioid-induced and , particularly in end-of-life scenarios where patients receive for pain control. A 2024 randomized prospective study in cancer patients showed that prophylactic oral haloperidol at 2.5 mg nightly effectively prevented (mean scores of 1-2 on a severity scale) with a response rate comparable to metoclopramide, though it was less effective against . It is also used off-label for at doses of 0.5-2 mg or PO. Recent reviews from 2024 affirm haloperidol's role in preventing and in advanced cancer, highlighting its utility as a second-line option when first-line agents like metoclopramide are contraindicated or ineffective. Haloperidol has off-label applications for intractable hiccups, typically at 1-3 mg/day orally, based on case reports and small studies showing rapid resolution. Emerging applications of haloperidol include through its modulation of sigma-1 receptors, which play a role in neuropathic and inflammatory pathways. A 2024 narrative review summarized preclinical and clinical evidence indicating that haloperidol, at doses of 1 to 5 mg intravenously or orally, reduces in conditions like (with VAS score improvements of up to 4.0 points) and migraines, while also decreasing requirements in palliative and settings. Additionally, low-dose haloperidol shows potential in preventing postoperative , with perioperative administration (e.g., 1 mg twice daily) reducing incidence and severity in elderly surgical patients according to a 2024 . In , haloperidol is used adjunctively for associated , with doses of 5 to 10 mg intravenously helping control alongside benzodiazepines, though evidence is primarily from observational data. Despite these uses, haloperidol is not recommended as a first-line agent for due to the scarcity of high-quality randomized controlled trials isolating its effects, with most studies featuring small samples and heterogeneous dosing that limit generalizability. In patients with hepatic impairment, haloperidol requires cautious dosing adjustments, as reduced liver and protein binding can elevate plasma levels, potentially increasing toxicity risks; lower initial doses and frequent monitoring of liver function are advised.

Pregnancy and lactation

Haloperidol is assigned to FDA C, indicating that animal reproduction studies have shown adverse effects on the , such as increased resorption and pup mortality in at doses up to three times the maximum dose, with no teratogenic effects observed except for cleft in mice at higher doses, while adequate and well-controlled studies are lacking. Use during is recommended only if the potential benefit justifies the potential risk to the . Third-trimester may lead to neonatal , such as agitation, , , or , and/or withdrawal symptoms including , respiratory distress, and feeding difficulties, which can range from mild and self-limited to severe cases requiring prolonged hospitalization. Post-2000 registry and cohort data, including analyses from the Swedish Birth Register and prospective studies, indicate no significant increase in major congenital malformations with first-trimester exposure to haloperidol or other typical antipsychotics, though associations with shorter , lower , and have been reported. One population-based study found an elevated risk of among women using typical antipsychotics like haloperidol (adjusted 1.77, 95% 1.04-3.03), with 4.4% incidence compared to 1.7% in unexposed pregnancies. According to American College of Obstetricians and Gynecologists (ACOG) guidelines, haloperidol may be used during for treating psychotic disorders when benefits outweigh risks, with routine screening for recommended for those on antipsychotics; atypical antipsychotics are generally preferred over typical agents like haloperidol due to their profiles, though haloperidol maintains a relatively established reproductive safety record. Regarding lactation, haloperidol passes into at low levels, with maternal doses up to 10 mg daily yielding milk concentrations ranging from undetectable to 23.5 mcg/L and infant levels of 0.8–2.1 mcg/L, typically without adverse effects on the breastfed . Exposed infants have shown normal development in most cases, but rare instances of excessive , poor feeding, and motor slowing have occurred, particularly with concurrent use of other antipsychotics; monitoring for drowsiness and developmental milestones is advised.

Dosage and administration

Haloperidol is available in oral formulations, including tablets and concentrated solution, for the management of psychotic disorders. For adults with moderate psychotic symptoms, the initial oral dose is typically 0.5 to 2 mg administered two to three times daily, while for severe symptoms, it ranges from 3 to 5 mg two to three times daily. Doses are titrated based on clinical response to achieve the lowest effective level, with maintenance therapy often ranging from 1 to 15 mg per day in divided doses; higher daily totals up to 100 mg may be required in cases but are uncommon. The oral concentrate should be diluted in water, milk, or juice prior to to improve and ensure accurate dosing. Parenteral administration via intramuscular (IM) injection is indicated for acute agitation in psychotic patients, starting with 2 to 5 mg, which may be repeated every 60 minutes as needed, not exceeding 20 mg per day. Intravenous (IV) use, though not FDA-approved, is sometimes employed off-label for at infusion rates of 0.5 to 1 mg per hour, with careful for cardiac effects. For long-term maintenance, the decanoate depot formulation is administered IM monthly at 50 to 200 mg, calculated as 10 to 20 times the previous daily oral dose (initially), with lower multipliers (10 to 15 times) for maintenance; injections should be deep IM using a 21-gauge needle, limited to 3 mL per site. Dosage adjustments are essential for vulnerable populations to minimize risks. In elderly patients, initial oral doses should be reduced to 0.25 to 0.5 mg two to three times daily due to altered and increased sensitivity, with gradual . For hepatic , caution is advised as liver accounts for primary clearance; severe cases may require dose reductions of up to 50% due to elevated levels from decreased protein and . Renal generally does not necessitate adjustments, given minimal renal , though is recommended in advanced disease. Pediatric use is FDA-approved for oral formulations (ages 3-12 years) in and severe behavioral disorders but not established for safety and efficacy in injectable or decanoate formulations.

Safety profile

Contraindications

Haloperidol is contraindicated in patients with known to the drug or any of its components, as severe allergic reactions may occur. It is also absolutely contraindicated in individuals with severe (CNS) depression or comatose states from any cause, due to the risk of exacerbating respiratory and cardiovascular depression. Additionally, haloperidol should not be used in patients with or , as it can worsen and lead to severe motor deterioration in these populations. Relative contraindications include a history of (NMS), a potentially life-threatening condition characterized by hyperpyrexia, muscle rigidity, and autonomic instability; prior exposure increases recurrence risk, necessitating avoidance or extreme caution. Use is also relatively contraindicated in patients with prolonged (greater than 500 ms), as haloperidol can further extend QTc and precipitate or sudden cardiac death. Cardiac risks warrant careful consideration; haloperidol should be avoided in patients with uncorrected or hypomagnesemia, conditions that predispose to QT prolongation, and in those concurrently using other QT-prolonging drugs. A baseline electrocardiogram (ECG) is recommended prior to initiation, particularly in at-risk individuals, to assess and monitor for changes. The FDA has issued a warning for haloperidol due to increased mortality in elderly patients with dementia-related ; analyses of 17 -controlled trials showed a 1.6-1.7 times higher risk of death compared to (4.5% vs. 2.6% over 10 weeks), primarily from cardiovascular or infectious causes, and it is not approved for this indication.

Adverse effects

Haloperidol, a , is associated with a range of adverse effects, primarily due to its potent D2 receptor blockade. The most common are (EPS), which occur in up to 50% of patients treated with typical antipsychotics like haloperidol, particularly in the first few weeks of therapy. EPS include acute , characterized by involuntary muscle contractions such as or , affecting 10-30% of patients, especially young males; , an inner sense of restlessness, seen in approximately 20% of cases; and drug-induced , manifesting as bradykinesia, rigidity, and , which can occur in up to 50% over 6-12 weeks. These symptoms are dose-dependent and more frequent with high-potency agents like haloperidol compared to atypical antipsychotics. Management typically involves anticholinergic agents such as benztropine for acute relief, with dose reduction or switching medications for prevention. Tardive dyskinesia (TD), a potentially irreversible involving choreoathetoid movements, particularly of the face and , carries a cumulative risk of 5% per year with haloperidol use, with higher incidence in elderly women and long-term users. Monitoring with the Abnormal Involuntary Movement Scale (AIMS) is recommended every 3-6 months during prolonged therapy to detect early signs. Discontinuation or switching to atypical antipsychotics may halt progression, though established TD often persists. Metabolic effects are less pronounced than with antipsychotics but still notable; haloperidol is linked to modest , averaging 1-2 kg over 10 weeks, compared to 4 kg or more with agents like . Hyperprolactinemia occurs in up to 90% of patients, resulting from tuberoinfundibular blockade, and can lead to , menstrual irregularities, or ; levels should be monitored in symptomatic patients. Cardiac adverse effects include dose-dependent prolongation, typically up to 20 ms at therapeutic doses, though 2020s meta-analyses of over 12,000 patients indicate low risk of arrhythmias or in short-term use, with no significant increase compared to . Electrocardiogram monitoring is advised in patients with cardiac risk factors or concurrent QT-prolonging drugs. (NMS), a rare but life-threatening reaction, has an incidence of 0.01-0.02% with haloperidol, presenting with , severe muscle rigidity, autonomic instability, and altered consciousness. Immediate discontinuation, supportive care, and or may be required for management.

Drug interactions

Haloperidol undergoes metabolism primarily via enzymes, including and , making it susceptible to pharmacokinetic interactions with inhibitors of these pathways. Strong inhibitors, such as , elevate haloperidol plasma concentrations, which can heighten the risk of (EPS) and other adverse effects; dose reduction of haloperidol is recommended during co-administration. Pharmacodynamic interactions occur with other QT-prolonging agents, including selective serotonin reuptake inhibitors (SSRIs) like and antiarrhythmics such as or , resulting in additive prolongation of the and increased risk of . Such combinations should generally be avoided, especially in patients with a exceeding 500 ms, with electrocardiographic monitoring advised if unavoidable. Haloperidol potentiates the hypotensive effects of antihypertensives, particularly alpha-blockers, leading to enhanced risk of . It also antagonizes the therapeutic effects of agonists like levodopa by blocking , which may exacerbate parkinsonian symptoms and necessitate adjustments in antiparkinsonian therapy. Co-administration with or other (CNS) depressants, such as opioids or anesthetics, produces additive CNS depression, increasing , respiratory depression, and impairment of psychomotor skills. Patients should avoid and exercise caution with concomitant CNS depressants to minimize these risks.

Discontinuation and withdrawal

Discontinuation of haloperidol requires careful to minimize the risk of and effects, particularly in patients with or other disorders. Abrupt cessation is associated with a significantly higher rate, with studies showing up to 77% of patients experiencing psychotic within one year compared to 31% when tapering occurs over more than 10 weeks. Gradual dose reduction is recommended to allow for adaptation of systems and reduce the likelihood of rebound . Tapering protocols typically involve reducing the dose by 10-25% of the current dose every 1-4 weeks, depending on the and duration of , with some approaches extending to monthly reductions over several months for long-term users. A hyperbolic tapering method, where reductions are proportional to the current dose (e.g., 25% every 3-6 months), has been proposed to more closely mimic the gradual decline in and minimize supersensitivity risks. For example, a on 4 mg daily might progress through steps like 3 mg, 2 mg, 1.5 mg, and lower, with intervals adjusted based on clinical response. Close monitoring for early signs of or symptom is essential during this process, with specialist oversight advised for complex cases. Withdrawal symptoms following haloperidol discontinuation can include relapse of , often within 1-2 weeks, as well as motor disturbances such as , , or , which may affect 31-50% of patients after abrupt stopping. Other manifestations encompass anxiety, , , sweating, and gastrointestinal upset, with motor symptoms potentially persisting for months. These effects arise from and unmasking of underlying conditions, though evidence quality remains low due to limited randomized trials. For patients on long-acting injectable haloperidol decanoate, discontinuation involves administering the final injection followed by a switch to oral tapering once plasma levels begin to decline, given the formulation's of approximately 3 weeks. Detectable levels may persist for up to 13 weeks post-last dose, necessitating extended monitoring for 3-6 months to detect delayed or . During this period, patients should be observed for , which can emerge or worsen upon depot clearance. Professional guidelines, such as those from psychiatric associations, emphasize slow tapering in stable patients to prevent adverse outcomes, with abrupt discontinuation contraindicated especially in those on high doses or with a history of relapse. For instance, reductions should only proceed after at least 12 months of stability in first-episode cases, with ongoing assessment of mental state and support systems. In all scenarios, multidisciplinary involvement ensures safe cessation tailored to individual risk factors.

Overdose

Signs and symptoms

Haloperidol overdose manifests as an exaggeration of its known pharmacological effects, primarily involving the , , and cardiovascular system. Symptoms typically onset rapidly, with extrapyramidal effects appearing within hours of ingestion. In mild cases, patients often present with drowsiness, sedation, and such as muscle rigidity, , and , accompanied by mild . These features reflect the drug's D2 receptor blockade and may overlap briefly with chronic adverse effects like observed during long-term use. Moderate overdose intensifies neurological involvement, leading to , , and electrocardiographic changes including prolongation. Seizures occur infrequently in these scenarios. Severe intoxication can progress to , profound respiratory depression, and life-threatening ventricular arrhythmias such as , with potentially culminating in . is dose-dependent and variable, but doses substantially exceeding therapeutic levels (e.g., beyond 100 mg daily) heighten the risk of fatal outcomes in adults. In pediatric patients, overdose carries a heightened risk of acute and due to greater sensitivity to extrapyramidal effects, with symptoms such as rigidity, , and altered consciousness potentially mimicking .

Management

Management of haloperidol overdose begins with immediate initial stabilization following the ABCs (airway, breathing, circulation). The airway should be secured with an , endotracheal , or tracheostomy if obstruction or is present, while supplemental oxygen is administered via nasal prongs, facemask, or for . Circulation is supported with intravenous fluids for , escalating to vasopressors such as norepinephrine if needed, while avoiding epinephrine due to potential exacerbation of arrhythmias. Gastrointestinal decontamination is indicated if ingestion occurred within 1 hour, with activated charcoal administration reducing drug absorption by approximately 50% in eligible cases. may be considered in early presentations, particularly for large ingestions, to remove unabsorbed drug. Symptom-specific interventions address acute manifestations. For extrapyramidal symptoms such as acute , benztropine 1-2 mg intravenously is administered promptly, followed by oral dosing if symptoms persist. Seizures are managed with benzodiazepines, such as or , titrated to effect. QRS complex widening on electrocardiogram, if present, is treated with to counteract blockade effects. Continuous cardiac monitoring is essential, with serial electrocardiograms to detect QTc prolongation exceeding 500 ms, a risk factor for . In cases of , 1-2 g intravenously over 15 minutes is given, with potential repetition as needed to stabilize rhythm. Supportive care includes admission to an for severe cases involving hemodynamic instability, arrhythmias, or altered mental status. is ineffective due to haloperidol's high of approximately 92%, limiting extracorporeal removal. Observation and monitoring continue until clinical stability, considering the drug's elimination of 14-26 hours as detailed in the pharmacokinetics section.

Prognosis

With prompt medical intervention, the from haloperidol overdose is low, typically less than 1%, as evidenced by rare fatalities among thousands of reported exposures. In contrast, rates are higher in the elderly or with , potentially reaching up to 5% or more if complicated by . Most cases resolve within 24-72 hours under supportive care, though symptoms such as extrapyramidal effects may persist for days to weeks in a subset of patients, necessitating prolonged therapy. Key risk factors for adverse outcomes include ingested doses exceeding 50 mg, intravenous , and comorbidities like preexisting cardiac disease; improves substantially with initiated within the first 2 hours of . Long-term sequelae are uncommon but can include rare instances of permanent or neurologic deficits if overdose leads to complications such as prolonged or ; psychiatric follow-up is advised upon discharge to address underlying issues.

Haloperidol primarily acts as a potent at D2 receptors, exhibiting high binding affinity with a Ki value of 1.2 , which underlies its therapeutic in treating psychotic disorders. By competitively blocking postsynaptic D2 receptors in the mesolimbic pathway, haloperidol reduces excessive transmission responsible for positive symptoms such as hallucinations and delusions. Optimal effects are achieved at approximately 60-80% D2 receptor occupancy in the brain. This mechanism also extends to modulation of activity, where D2 antagonism influences functional connectivity between and cortical regions, potentially contributing to improvements in cognitive aspects of . In addition to its strong D2 selectivity, haloperidol displays moderate antagonism at serotonin 5-HT2A receptors (Ki = 60 nM) and alpha-1 adrenergic receptors (Ki = 20 nM), while showing weaker binding to D1 receptors (Ki = 200 nM). The 5-HT2A blockade may enhance its profile by balancing serotonergic influences on release, though this effect is secondary to D2 antagonism. Alpha-1 adrenergic antagonism can lead to as an due to reduced vascular tone. These multi-receptor interactions distinguish haloperidol from more selective agents and contribute to its broad clinical utility, including properties via D2 receptor blockade in the of the . Adverse effects of haloperidol correlate with its D2 receptor antagonism in extrapyramidal pathways; blockade in the nigrostriatal tract disrupts dopaminergic inhibition of cholinergic interneurons, resulting in such as , , and . Furthermore, haloperidol functions as a high-affinity at sigma-1 receptors (Ki ≈ 2-6 nM), and this interaction has been investigated for potential applications, though its primary role remains in therapy.

Pharmacokinetics

Haloperidol exhibits variable absorption depending on the . results in good absorption from the , with a of 60-70% due to first-pass hepatic . Peak plasma concentrations are typically achieved within 2 to 6 hours after oral dosing. provides rapid within 10 to 30 minutes, with peak plasma levels reached in approximately 20 to 30 minutes. The long-acting intramuscular depot formulation, haloperidol decanoate, slowly releases the drug, achieving peak concentrations around 6 days post-injection and providing therapeutic levels over 3 to 4 weeks. The drug is widely distributed throughout the body, with a high of approximately 18 L/kg, reflecting its extensive penetration into tissues. Haloperidol is highly bound to proteins, at about 92%. Its lipophilic nature allows it to readily cross the blood-brain barrier, resulting in higher concentrations in brain tissue compared to , which underlies its effects. Haloperidol undergoes extensive hepatic metabolism, primarily via the enzymes and , as well as carbonyl reductase, leading to reduced metabolites. Key metabolites include reduced haloperidol and the active hydroxyhaloperidol, which contributes 20-30% of the overall pharmacologic activity. , mediated by UGT enzymes, accounts for 50-60% of the and is a major clearance pathway. Elimination of haloperidol occurs mainly through hepatic , with less than 1% excreted unchanged in the and the remainder as metabolites primarily via (about 40% of total radioactivity) and feces. The terminal elimination is 14 to 26 hours for oral and immediate-release intramuscular formulations, while the decanoate depot ester has a much longer of approximately 21 days due to gradual .

Therapeutic concentrations and monitoring

Therapeutic drug monitoring (TDM) for haloperidol involves measuring plasma concentrations to optimize efficacy in treating while minimizing risks such as (EPS). The established therapeutic range for haloperidol plasma levels is typically 1 to 10 ng/mL, with a minimum response around 2 ng/mL and optimal efficacy for control observed between 2 and 10 ng/mL. Levels exceeding 15 ng/mL are associated with increased risk of , including heightened EPS, without additional therapeutic benefit. Plasma concentrations of haloperidol exhibit significant interindividual variability, influenced primarily by genetic polymorphisms in the enzyme, which metabolizes approximately 60-70% of the drug. Poor metabolizers of , comprising about 7-10% of populations, achieve plasma levels approximately twice as high as normal metabolizers at equivalent doses, necessitating dose reductions to 60% of the standard amount to avoid supratherapeutic exposure. Drug interactions further contribute to variability; or inhibitors (e.g., or ) can elevate haloperidol levels by 50% or more, while inducers (e.g., ) may reduce them by 30-50%, altering and profiles. Clinical recommendations emphasize TDM in specific scenarios rather than routine use, due to its and limited . TDM is strongly advised for poor metabolizers, non-responders to standard dosing, or patients with suspected non-compliance, with trough levels measured 12-24 hours post-dose to guide adjustments and improve response rates. For long-term use, electrocardiogram (ECG) for prolongation is recommended, particularly weekly in patients with cardiac risk factors, as haloperidol can extend the QTc interval and precipitate at higher doses or in vulnerable individuals. Supporting evidence from 2010s studies, including the 2020 AGNP/ASCP consensus guidelines, demonstrates correlations between levels in the 2-10 ng/mL and clinical response in , with subtherapeutic levels (<2 ng/mL) linked to poor outcomes and supratherapeutic levels (>15 ng/mL) to diminished benefits and adverse effects. TDM is not routinely implemented in clinical practice owing to logistical challenges and expense but proves valuable in cases, where it can identify under- or overdosing responsible for up to 30% of non-responses.

Chemistry

Chemical structure and properties

Haloperidol is a member of the class of drugs, characterized by a central ring substituted at the 4-position with a hydroxy and a 4-chlorophenyl group, connected via a propyl chain to a 4-fluorobutyrophenone moiety. Its IUPAC name is 4-[4-(4-chlorophenyl)-4-hydroxypiperidin-1-yl]-1-(4-fluorophenyl)butan-1-one, with the molecular formula C21H23ClFNO2 and a molecular weight of 375.9 g/mol. Physically, haloperidol appears as a white to faintly yellowish, amorphous or microcrystalline powder that is odorless. It exhibits high , with a value of 4.3, and has a of 8.3 (or 8.65 in some measurements), indicating basic character due to the . Solubility is limited in (practically insoluble, approximately 14 mg/L at 25°C), but it is soluble in organic solvents such as (16.7 mg/mL at 25°C), , , acetone, and , as well as in dilute acids. Haloperidol is light-sensitive and requires protection from exposure to maintain , with storage recommended in airtight containers at . It demonstrates under neutral conditions and lacks readily hydrolyzable groups, but degrades under exposure to strong acids, bases, elevated temperatures, or . The compound is typically formulated as the free base or as salts such as the hydrochloride or lactate to enhance solubility for oral and injectable administration. Additionally, haloperidol decanoate is an ester prodrug used in long-acting injectable depots, providing sustained release upon hydrolysis in vivo.

Synthesis and formulation

Haloperidol's chemical synthesis was originally developed at Janssen Pharmaceutica and patented in processes filed starting in 1958, with the key U.S. patent (US3438991) describing the condensation of 4-(4-chlorophenyl)-4-hydroxypiperidine with γ-chloro-4'-fluorobutyrophenone in the presence of potassium iodide as a catalyst and sodium carbonate as a base, typically heated in an inert solvent such as toluene at 100–150°C for 24–72 hours to yield the product. The 4-(4-chlorophenyl)-4-hydroxypiperidine intermediate is prepared via a Grignard reaction, where 4-chlorophenylmagnesium bromide—formed from 1-bromo-4-chlorobenzene and magnesium in anhydrous ether—is added to N-protected piperidin-4-one (such as 1-benzylpiperidin-4-one), followed by acidic hydrolysis to form the tertiary alcohol and subsequent deprotection of the nitrogen (e.g., via hydrogenation for benzyl removal). The γ-chloro-4'-fluorobutyrophenone is synthesized through a Friedel-Crafts acylation of fluorobenzene with 4-chlorobutanoyl chloride in the presence of aluminum chloride, often in a solvent like carbon disulfide or dichloromethane at low temperature to control the reaction. This multi-step process, involving preparation of intermediates, coupling, and purification (typically via crystallization from ethanol or acetone), achieves overall yields of around 50–70% depending on scale and conditions, with early methods producing material of sufficient purity for pharmaceutical use after recrystallization. Modern industrial synthesis retains the core Grignard and steps but incorporates optimizations such as safer solvents (e.g., replacing with ) and alternative catalysts to minimize waste, including alternatives to aluminum chloride for the to reduce and environmental impact. The original , patented by Janssen, involved approximately five key stages from commercial starting materials like and 1-bromo-4-chlorobenzene, emphasizing stereochemical control at the tertiary alcohol center, which remains racemic in the final product. Pharmaceutical formulations of haloperidol are designed for both acute and maintenance therapy, with the base form used in oral tablets available in strengths of 0.5 mg, 1 mg, 2 mg, 5 mg, and 10 mg, often coated for ease of swallowing and stability. The salt is formulated as an for parenteral administration at 5 mg/mL, suitable for intramuscular or intravenous use in acute settings due to its and rapid onset. For long-acting therapy, the decanoate is prepared as an intramuscular oil-based in or medium-chain triglycerides at concentrations of 50 mg/mL and 100 mg/mL, providing depot release over 2–4 weeks after injection. An oral concentrate of 2 mg/mL is also available for flexible dosing in solution form, typically diluted in water or juice. Manufacturing of haloperidol originated exclusively with Janssen Pharmaceutica following the 1958 synthesis and subsequent patents, with production scaled up in for global distribution starting in the early 1960s. After patent expiry in the late 1980s (e.g., U.S. patent expiration around 1986), manufacturers worldwide adopted similar synthetic routes, leading to widespread availability through companies like and , with quality controlled under pharmacopeial standards such as or EP monographs ensuring purity above 98%.

History

Development and discovery

Haloperidol, the prototype of the butyrophenone class of antipsychotics, was synthesized on February 11, 1958, by chemist Bert Hermans under the supervision of Paul Adriaan Jan Janssen at Janssen Pharmaceutica in Beerse, Belgium. Initially codenamed R-1625, the compound emerged from a systematic structure-activity relationship study modifying pethidine analogs to develop potent analgesics with reduced narcotic effects, but it unexpectedly exhibited strong neuroleptic properties. This synthesis marked a pivotal advancement, as haloperidol became the first member of its chemical class to exhibit potent antipsychotic activity without the sedative or morphine-like side effects associated with earlier agents. Preclinical evaluation in animal models rapidly confirmed haloperidol's therapeutic potential. In studies conducted shortly after , the induced strong cataleptogenic effects in rats and dogs at low doses, a behavioral response linked to D2 receptor antagonism and predictive of efficacy. Compared to , the benchmark introduced in 1952, haloperidol demonstrated superior potency, requiring significantly lower doses to achieve equivalent cataleptogenic and anti-emetic responses while avoiding substantial . These findings, published in early pharmacological reports, underscored haloperidol's selectivity for neuroleptic action, distinguishing it from analgesics and setting the stage for its clinical exploration. The transition to human testing occurred swiftly, with the first clinical trials commencing in 1959 across . Cyrille Bloch in administered initial intravenous doses of 2 mg to patients experiencing , observing rapid calming effects without excessive sedation. Building on this, researchers at the expanded trials to individuals with and other psychotic disorders, where haloperidol effectively reduced and psychotic symptoms, leading to its quick embrace for managing acutely disturbed patients in psychiatric settings. A landmark symposium in September 1959 at Janssen's headquarters gathered European clinicians to present 17 studies affirming these outcomes, accelerating its adoption. Key milestones in haloperidol's early development included patent filings from 1959 to 1961, which protected Janssen Pharmaceutica's innovations in chemistry and propelled the compound toward commercialization. The name "haloperidol" was derived from the two atoms ( and ) incorporated into its structure. By October 1959, it was licensed and marketed in as Haldol, initiating widespread therapeutic use in .

Clinical introduction and approvals

Haloperidol entered clinical use following promising early trials in during the late and early , where it demonstrated efficacy in managing psychotic symptoms through antagonism. Initial human studies began in in 1959, shortly after its synthesis in 1958, showing rapid effects in patients with and agitation. By 1960, it received national approvals in several European countries, including , for the treatment of , marking its introduction as a first-generation . In the United States, the (FDA) approved haloperidol on April 12, 1967, initially for and other psychotic disorders, based on double-blind, -controlled trials from the 1960s that reported response rates around 70% in reducing positive symptoms like hallucinations and delusions. These studies, involving hundreds of patients, highlighted its superiority over in acute management, with typical oral doses of 2-10 mg daily. The approval expanded in 1982 to include severe and vocal utterances in Tourette's syndrome, supported by controlled trials demonstrating tic reduction in pediatric and adult populations. The 1970s saw development of long-acting intramuscular depot formulations, with key trials evaluating haloperidol decanoate for maintenance therapy in chronic schizophrenia; these double-blind studies showed relapse prevention comparable to oral forms, leading to FDA approval of the decanoate injection in 1986. Globally, haloperidol's adoption accelerated after its inclusion on the World Health Organization's Model List of Essential Medicines in 1977, recognizing its role in treating schizophrenia and psychotic disorders in resource-limited settings; by the 1980s, it was available and routinely used in over 100 countries as a cornerstone antipsychotic. Its widespread use contributed to the deinstitutionalization of psychiatric patients, though early reports of tardive dyskinesia prompted refined guidelines by the 1970s. In the 2020s, clinical studies and guidelines have examined haloperidol's role in for off-label management of , , and in end-of-life settings, with mixed evidence supporting its utility in select cases, though no major new formal approvals have occurred; instead, emphasis has grown on its expanded off-label applications in intensive care and contexts to improve symptom control without introducing novel formulations.

Society and culture

Brand names and availability

Haloperidol is primarily marketed under the brand name Haldol by Janssen Pharmaceutica, which was the original developer and introduced the drug in 1959. Other notable brand names include Serenace (in various regions) and Haldol Decanoate for the long-acting intramuscular depot formulation. Haloperidol is widely available in generic forms, such as haloperidol tablets, oral solution, and injection for acute use. In the United States, haloperidol is available by prescription only and is not classified as a under the schedules, though it carries a warning against use in elderly patients with -related due to increased mortality risk. It is similarly prescription-only in the , where it is authorized through the for treating and other , with restrictions on use in elderly patients. In , haloperidol is broadly accessible via national regulatory approvals in countries like and , often as generics for psychiatric indications, but with similar warnings for vulnerable populations. The original for haloperidol expired in 1986, enabling the entry of multiple manufacturers worldwide and contributing to its low-cost availability in over 50 countries today.

Cost and access

Haloperidol, available as a medication, is relatively inexpensive in high-income countries. , a month's supply of oral haloperidol typically costs $10 to $50, depending on dosage and , with prices as low as $3.10 for a 30-day supply of 5 mg tablets using discount programs (as of November 2025). Depot injections, such as haloperidol decanoate, range from $20 to $60 per monthly dose (as of November 2025), reflecting production costs for long-acting formulations. In contrast, prices are significantly lower in countries like and , where oral haloperidol costs less than $5 per month; for example, a 5 mg tablet in is approximately ₹3.50 (about $0.04) (as of 2025). Insurance coverage enhances accessibility in many systems. In the , haloperidol is covered under plans, with patient copays generally ranging from $5 to $20 for a month's supply after meeting deductibles. In public health systems like the UK's (NHS), haloperidol is provided free of charge to eligible patients, including those exempt from prescription charges, which cover most treatments. Access challenges persist globally, particularly due to supply disruptions. Shortages of haloperidol, especially the decanoate injection, have occurred in the owing to manufacturing delays and discontinuations by some producers, affecting availability in the and other markets. In low-income countries, costs remain low due to widespread generic production, but barriers include limited healthcare and stigma around treatment. Economically, haloperidol remains cost-effective compared to atypical antipsychotics, often 20% to 50% cheaper—or more in some analyses—due to its status and lower acquisition costs, making it a preferred option in resource-limited settings. Its inclusion in WHO prequalification programs for supports procurement for global aid initiatives, improving equity in low- and middle-income countries; haloperidol is included on the World Health Organization's Model List of .

Research

Emerging human applications

Recent reviews have explored low-dose haloperidol (typically 0.5–2 mg) as an adjunct for managing chronic and neuropathic pain, particularly in palliative settings, where it may reduce opioid requirements through mechanisms such as sigma-1 receptor antagonism and NMDA channel blockade. A 2024 narrative review highlighted its efficacy in treating pain associated with gastroparesis, migraines, and facial neuralgia, with median daily doses around 1.5 mg showing promise for opioid-sparing effects in cancer patients, though evidence for broader neuropathic applications remains limited and calls for more prospective trials. In , haloperidol has gained expanded use for refractory and , as recommended in 2020s guidelines targeting stimulation from metabolic causes or medications. Doses of 0.5–1.5 mg orally or subcutaneously every 4–8 hours have demonstrated response rates up to 65% in advanced cancer patients, comparable to other antiemetics like metoclopramide, based on systematic reviews and ongoing comparative trials. For delirium management in intensive care units, including those with COVID-19 complications, haloperidol has shown potential to reduce delirium duration and rescue needs, with meta-analyses indicating a possible mortality (risk difference -0.03) in treated patients versus , though long-term outcomes require further confirmation. A 2025 Bayesian of ICU trials supported its use for acute delirium treatment, reporting a 68% probability of clinically important mortality reduction without increased serious adverse events. Despite these applications, challenges persist due to limited randomized controlled trials in non-psychotic populations, raising concerns over safety profiles including and prolongation at higher doses. Updated meta-analyses emphasize that while haloperidol appears safe short-term, its efficacy beyond in preventing or improving survival remains inconsistent, underscoring the need for larger RCTs to assess risks in diverse patient groups.

Veterinary uses

Haloperidol is utilized in as a neuroleptic agent to manage behavioral disorders and facilitate handling in various , particularly where sedation or antipsychotic effects are required. In and cats, it is commonly administered to calm aggressive or agitated behavior, with typical intramuscular doses ranging from 0.2 to 4 mg/kg every 6-8 hours as needed, though efficacy for long-term behavioral modification has been limited. In non-human primates, haloperidol serves as a model for studying and chronic exposure, often given orally at doses of 0.5-1.0 mg/kg daily to induce and maintain therapeutic effects in settings. Veterinary applications rely on human-approved formulations, including injectable solutions for intramuscular or intravenous use and oral concentrates or tablets adapted for animals, as no dedicated veterinary products like a specific "Haldol Vet" are widely available. Oral administration is particularly suited for zoo animals and primates, where it can be mixed with food such as fruit juice to premedicate for anesthesia or reduce stress during procedures, providing sedation lasting 2-4 hours. In the United States, haloperidol lacks specific FDA approval for veterinary use and is employed extra-label under the Animal Medicinal Drug Use Clarification Act (AMDUCA) of 1994, allowing veterinarians to prescribe it legally for off-label indications in non-food animals. In the European Union, its application in food-producing animals is restricted or prohibited due to the absence of established maximum residue limits (MRLs) for pharmacologically active substances, aiming to prevent potential residues in meat, milk, or eggs that could enter the human food chain. In dogs, the drug's indicate a prolonged elimination of approximately 20-30 hours, contributing to its sustained effects compared to shorter durations in some other species.

References

  1. [1]
    Haloperidol - StatPearls - NCBI Bookshelf
    Haloperidol, a first-generation typical antipsychotic, is commonly used worldwide to block dopamine D2 receptors in the brain and exert its antipsychotic action ...
  2. [2]
    Janssen, the discovery of haloperidol and its introduction ... - PubMed
    Apr 29, 2009 · The discovery of haloperidol at the end of the 1950s constitutes one of the greatest advances of 20th century psychiatry.
  3. [3]
  4. [4]
    Haloperidol: Uses, Interactions, Mechanism of Action - DrugBank
    Haloperidol is indicated for the treatment of the manifestations of several psychotic disorders including schizophrenia, acute psychosis, Tourette syndrome, and ...Identification · Pharmacology · Categories · References
  5. [5]
    Haloperidol bound D2 dopamine receptor structure inspired ... - NIH
    Feb 26, 2020 · and O4RE6 and the individual enantiomers were docked in order to understand their mechanism of action. ... schizophrenia. Nat. Rev. Drug Discov.
  6. [6]
    Haloperidol versus placebo for schizophrenia - PMC - PubMed Central
    Haloperidol was first developed in the late 1950s. Research subsequently showed its therapeutic effects on the symptoms of schizophrenia, such as hearing voices ...
  7. [7]
    Optimal haloperidol dosage in first-episode psychosis - PubMed
    Results: Optimal doses for the 36 subjects were 2 mg daily for 15 subjects, 5 mg daily for 11, 10 mg daily for 7, and 20 mg daily for 3.<|separator|>
  8. [8]
    A Comparison of Risperidone and Haloperidol for the Prevention of ...
    The Kaplan-Meier estimate of the risk of relapse at the end of the study was 34 percent for the risperidone group and 60 percent for the haloperidol group (P< ...
  9. [9]
    Classics in Chemical Neuroscience: Haloperidol - ACS Publications
    The discovery of haloperidol catalyzed a breakthrough in our understanding of the biochemical basis of schizophrenia, improved the treatment of psychosis, ...
  10. [10]
    Current Approaches and New Developments in the ...
    Jan 15, 2018 · Haloperidol was the first medication approved by the FDA for the treatment of adult patients with TS in 1969 and for children in 1978 [30].
  11. [11]
    A Review of the Current Treatment of Tourette Syndrome - PMC
    Haloperidol and Pimozide.​​ Both haloperidol and pimozide are FDA approved to decrease Tourette syndrome–associated tic severity. Studies have attributed the tic ...Missing: 1980s | Show results with:1980s
  12. [12]
    Pimozide cf. Haloperidol in Tourette Syndrome
    A 70% tic reduction was obtained in 64% of patients during either drug ... haloperidol treatment, a threefold higher frequency than with pimozide. [1].
  13. [13]
    Haloperidol Dosage Guide + Max Dose, Adjustments - Drugs.com
    Aug 6, 2025 · Usual Adult Dose for Psychosis: Oral Haloperidol Formulations: Moderate symptomology: 0.5 to 2 mg orally 2 to 3 times a day. Severe symptomology: 3 to 5 mg ...
  14. [14]
    Practice guideline recommendations summary: Treatment of tics in ...
    Forty-six recommendations were made regarding the assessment and management of tics in individuals with Tourette syndrome and chronic tic disorders.
  15. [15]
    Tardive dyskinesia in Tourette syndrome - Pediatric Neurology
    Three patients are reported who developed oral-buccal-lingual movements typical of tardive dyskinesia while being treated with stable doses of haloperidol ...
  16. [16]
    [PDF] haldol - accessdata.fda.gov
    HALDOL Injection is not approved for the treatment of patients with dementia-related psychosis (see BOXED WARNING). Cardiovascular Effects. Cases of sudden ...
  17. [17]
    Pharmacologic Management of End-of-Life Delirium - MDPI
    May 28, 2024 · The authors concluded that both haloperidol and olanzapine were efficacious in reducing delirium symptoms in patients with advanced cancer.
  18. [18]
    ESMO: Combination therapy reduced agitated delirium in patients ...
    Sep 13, 2024 · Patients that received the combination of haloperidol and lorazepam experienced a significant decrease in agitated delirium when measured at 24 ...
  19. [19]
    Comparison of Efficacy and Safety of Prophylactic Use of ... - NIH
    Oct 1, 2024 · While we used haloperidol to prevent morphine-induced nausea and vomiting only, we got a higher response rate by haloperidol. Gralla et al. ...Missing: end- | Show results with:end-
  20. [20]
    Haloperidol for Pain Management: A Narrative Review - MDPI
    Aug 21, 2024 · Haloperidol has good efficacy in the treatment of pain related to gastroparesis and migraines and has shown promise for opioid use reduction in patients with ...
  21. [21]
    The effect of haloperidol's perioperative application on postoperative ...
    Feb 3, 2024 · The meta-analysis revealed perioperative application of haloperidol could decrease the occurrence of postoperative delirium without obvious side effects in ...<|separator|>
  22. [22]
    Delirium Tremens (DTs) Treatment & Management
    Aug 26, 2024 · Benzodiazepines are the mainstay in treatment for alcohol withdrawal, but several adjuncts can be considered.
  23. [23]
    [PDF] haldol - accessdata.fda.gov
    Usage in Pregnancy. Pregnancy Category C. Rodents given up to 3 times the usual maximum human dose of haloperidol decanoate showed an increase in incidence ...
  24. [24]
    Antipsychotic drug labels updated on use during pregnancy - FDA
    Aug 4, 2017 · The new drug labels now contain more and consistent information about the potential risk for abnormal muscle movements (extrapyramidal signs or EPS) and ...
  25. [25]
    Use of Antipsychotic Drugs During Pregnancy - PMC - NIH
    Jan 30, 2019 · Two studies of infants of mothers who took typical antipsychotics noted decreased gestational age and lower birth weights, but were confounded ...
  26. [26]
    Antipsychotics During Pregnancy: Relation to Fetal and Maternal ...
    Conclusions Women who used antipsychotics during pregnancy had increased risks of gestational diabetes. The increased risks of giving birth to an SGA infant ...
  27. [27]
    [PDF] Treatment and Management of Mental Health Conditions During ...
    ACOG recommends against discontinuing mood stabi- lizers, except for valproate, during pregnancy due to the risk of recurrence or exacerbation of mood symptoms.
  28. [28]
    ACOG Guidelines on Psychiatric Medication Use During Pregnancy ...
    Sep 15, 2008 · Therefore, the routine use of these drugs during pregnancy and lactation is not recommended. Typical antipsychotics have a larger reproductive ...
  29. [29]
    Haloperidol - Drugs and Lactation Database (LactMed®) - NCBI - NIH
    Mar 15, 2025 · Limited information indicates that maternal doses of haloperidol up to 10 mg daily produce low levels in milk and usually do not affect the breastfed infant.
  30. [30]
    [PDF] HALDOL brand of haloperidol injection (For Immediate Release ...
    Haloperidol concentrations may increase in hepatically impaired patients, because it is primarily metabolized by the liver and protein binding may decrease.
  31. [31]
    Haloperidol: Package Insert / Prescribing Information - Drugs.com
    Jan 21, 2024 · The injection provides 5 mg haloperidol (as the lactate) with 1.8 mg methylparaben and 0.2 mg propylparaben per mL, and lactic acid for pH ...
  32. [32]
    [PDF] HALDOL® Decanoate 50 (haloperidol) - accessdata.fda.gov
    Also, the pharmacokinetics of haloperidol in geriatric patients generally warrants the use of lower doses (see DOSAGE AND ADMINISTRATION). Use in Hepatic ...
  33. [33]
    Haloperidol (oral route) - Side effects & dosage - Mayo Clinic
    Older adults—At first, 0.5 to 2 mg 2 or 3 times a day. Your doctor may increase your dose if needed. However, the dose is usually not more than 100 mg per day.
  34. [34]
    [PDF] haldol - accessdata.fda.gov
    HALDOL Injection is not approved for the treatment of patients with dementia-related psychosis (see WARNINGS). DESCRIPTION. Haloperidol is the first of the ...
  35. [35]
    Haldol: Package Insert / Prescribing Information - Drugs.com
    Jul 30, 2025 · Elderly patients with dementia-related psychosis treated with antipsychotic drugs are at an increased risk of death. HALDOL Injection is not ...
  36. [36]
    Second‐Generation Antipsychotics and Extrapyramidal Adverse ...
    Jun 3, 2014 · It is estimated that approximately 50% of patients treated with high-potency FGAs (such as haloperidol) develop acute EPS within the first ...Abstract · Background · Extrapyramidal Symptoms · Conclusion
  37. [37]
    Antipsychotic drugs and extrapyramidal side effects in first episode ...
    Nov 5, 2011 · This systematic review aimed to determine whether the risk of extrapyramidal side effects (EPS) differed between antipsychotic drugs used in first episode ...Included Studies And... · Antipsychotics And Dose · Eps Rating Scale Outcomes
  38. [38]
    A Meta-Analysis of the Risk of Acute Extrapyramidal Symptoms with ...
    The results presented here confirm that the currently available intramuscular SGAs have a significantly lower risk of acute EPS compared to haloperidol alone.
  39. [39]
    Lower Risk for Tardive Dyskinesia Associated With Second ...
    By contrast, the weighted mean annual tardive dyskinesia risk for haloperidol in the three randomized, double-blind studies involving adults (N=408) was 5.4% ( ...
  40. [40]
    Almost All Antipsychotics Result in Weight Gain: A Meta-Analysis - NIH
    A refined statistical method (network analysis) allowed for mutual comparison between AP and shows that haloperidol has the least impact weight gain.
  41. [41]
    Hyperprolactinemia with Antipsychotic Drugs in Children and ...
    These drugs have prominent side effects of interest to pediatric endocrinologists, including weight gain and associated metabolic risk factors and ...
  42. [42]
    Effects of the classical antipsychotic haloperidol and atypical ...
    Antipsychotic drug-induced side effects of weight gain and sexual dysfunction have clinical significance adversely affecting both compliance and morbidity.
  43. [43]
    Major adverse cardiac events with haloperidol: A meta-analysis
    Jun 25, 2025 · We did not find that haloperidol was arrhythmogenic or increased mortality in these largely short-duration trials.
  44. [44]
    Neuroleptic Malignant Syndrome - StatPearls - NCBI Bookshelf - NIH
    Incidence rates range from 0.01% to 3.2% of patients taking neuroleptic medications. The incidence is decreasing due to newer agents, which are less likely to ...
  45. [45]
    [PDF] HALDOL brand of haloperidol injection - accessdata.fda.gov
    Usage in Pregnancy. Pregnancy Category C. Rodents given up to 3 times the usual maximum human dose of haloperidol decanoate showed an increase in incidence ...
  46. [46]
    Drug Information Group | University of Illinois Chicago
    If the baseline QT interval is ≥500 ms, IV haloperidol should be avoided. References. Micromedex Solutions [database online]. Greenwood Village, CO: Truven ...
  47. [47]
    Haloperidol and levodopa Interactions - Drugs.com
    Haloperidol may reduce the effectiveness of levodopa. In addition, using these medications together may increase the risk and/or severity of side effects.
  48. [48]
    A Method for Tapering Antipsychotic Treatment That May Minimize ...
    Abruptly stopping antipsychotic medication is the method most likely to induce relapse and withdrawal symptoms. These concepts are germane to the broader ...
  49. [49]
    Stopping and switching antipsychotic drugs - Australian Prescriber
    Oct 1, 2019 · For patients with serious psychiatric illness, stopping or switching antipsychotics requires referral to a specialist if possible. However, for ...<|control11|><|separator|>
  50. [50]
  51. [51]
    Haloperidol Withdrawal Symptoms - Consensus
    These studies suggest that haloperidol withdrawal symptoms can include anxiety, dyskinesia, behavioral dopamine supersensitivity, biochemical alterations, ...
  52. [52]
  53. [53]
    [PDF] Long-acting injectable antipsychotics: What to do about missed doses
    One study found that haloperi- dol plasma concentrations were detectable 13 weeks after the discontinuation of halo- peridol decanoate.Missing: protocol | Show results with:protocol
  54. [54]
    Guidelines for Discontinuation of Antipsychotics in Patients ... - NIH
    To prepare for discontinuation, doses should be reduced to levels lower than those mentioned above. In all studies, doses were tapered over 1 to 6 months.
  55. [55]
    Stopping and switching antipsychotic drugs - PMC - NIH
    Oct 1, 2019 · In general, specialist advice should be sought when stopping or switching antipsychotics While antipsychotics are often needed long term, ...
  56. [56]
    Haloperidol Overdose - News-Medical
    Studies suggest that for haloperidol, the dose required for acute poisoning is relatively high compared to therapeutic doses. Overdose does not occur with ...
  57. [57]
    [PDF] HALDOL® - Medsafe
    Adjust the haloperidol decanoate dose by up to 25 mg every 4 weeks (based on individual patient response) until an optimal therapeutic effect is obtained. •.
  58. [58]
    Accidental haloperidol poisoning in children - PMC - PubMed Central
    Though his vitals were stable, he had anticholinergic symptoms such as diaphoresis, hyperthermia and urinary retention. On detailed CNS exam, generalized ...
  59. [59]
    First-generation (typical) antipsychotic medication poisoning
    Jun 4, 2024 · Outline · SUMMARY AND RECOMMENDATIONS · INTRODUCTION · EPIDEMIOLOGY · PHARMACOLOGY AND CELLULAR TOXICOLOGY · KINETICS · CLINICAL FEATURES OF OVERDOSE.
  60. [60]
    Activated charcoal for acute overdose: a reappraisal - PMC - NIH
    Among studies in which the dose of AC was at least 50 g, the average reduction in systemic drug absorption was 47.3% at 30 min, 40.1% at 60 min and 16.5% at 120 ...Missing: haloperidol | Show results with:haloperidol
  61. [61]
    Cogentin (benztropine) dosing, indications, interactions, adverse ...
    Drug-Induced Extrapyramidal Disorders. 1-2 mg IV/IM/PO q8-12hr; titrate gradually at 0.5 mg increments at 5-6 day intervals based on response and tolerability; ...
  62. [62]
    Torsade de Pointes - Medscape Reference
    May 6, 2024 · Magnesium can be given at 1-2 g IV initially in 30-60 seconds, which then can be repeated in 5-15 minutes. Alternatively, a continuous infusion ...
  63. [63]
    Haloperidol in palliative care: Indications and risks - ScienceDirect
    It is widely used in acute and chronic disorders such as intractable hiccups, hallucinations, restlessness and agitation, psychosis [[4], [5], [6], [7]], and ...
  64. [64]
    Neuroleptic Agent Toxicity: Background, Pathophysiology, Etiology
    Jul 24, 2025 · Mortality is relatively rare with overdose of antipsychotic medication. However, if NMS occurs, the mortality rate can reach up to 10-12%. Risk ...Missing: onset | Show results with:onset
  65. [65]
    HALOPERIDOL (UK PID) - Inchem.org
    Doses between 10 mg and 60 mg taken by a number of young people resulted in a variety of extrapyramidal symptoms, cold sweats, and insomnia. The extrapyramidal ...
  66. [66]
  67. [67]
    Pharmacokinetics of haloperidol: an update - PubMed
    In vivo pharmacogenetic studies have indicated that the metabolism and disposition of haloperidol may be regulated by genetically determined polymorphic CYP2D6 ...
  68. [68]
    Haloperidol | C21H23ClFNO2 | CID 3559 - PubChem - NIH
    Contraindications: Severe dystonic reactions have followed the use of haloperidol, particularly in children and adolescents. It should therefore be used ...
  69. [69]
    Haloperidol and Haloperidol Decanoate (Chapter 7) - The Clinical ...
    Oct 19, 2021 · The suggested therapeutic response threshold is 2 ng/ml, and the point of futility is 18 ng/ml. The 2020 AGNP/ASCP consensus paper strongly ...
  70. [70]
    Annotation of DPWG Guideline for haloperidol and CYP2D6 - ClinPGx
    The recommendation for CYP2D6 poor metabolizers is to use 60% of the normal dose of haloperidol and for CYP2D6 ultrarapid metabolizers to use 1.5 times the ...
  71. [71]
    Plasma concentrations of haloperidol are related to CYP2D6 ... - NIH
    In conclusion, we found that dose-corrected plasma concentrations of haloperidol were related to CYP2D6 genotype at haloperidol doses lower than 20 mg daily, ...
  72. [72]
    Blood Levels to Optimize Antipsychotic Treatment in Clinical Practice
    May 19, 2020 · The quantification of antipsychotic levels in blood, or therapeutic drug monitoring (TDM), could aid clinicians in numerous challenging ...
  73. [73]
    A review of a recently published guidelines' “strong recommendation ...
    Jul 1, 2019 · In a CYP2D6 polymorphism study,23 it was found that patients with a greater number of functional CYP2D6 alleles had lower trough HAL plasma ...
  74. [74]
    Haloperidol (PIM 253) - INCHEM
    ... and Health (Publication No. DHHS (NIOSH)) 2:1352. Scialli JUK & Thornton WE (1978) Toxic reactions from a haloperidol overdose in two children. Thermal and ...
  75. [75]
    Advancing thermal stability analysis of haloperidol
    Nov 5, 2024 · (1983) demonstrated haloperidol's instability under conditions involving elevated temperature, light exposure, and varying pH levels [12].
  76. [76]
    [PDF] Decanoate 50 (haloperidol) HALDOL - accessdata.fda.gov
    Usage in Pregnancy. Pregnancy Category C. Rodents given up to 3 times the usual maximum human dose of haloperidol decanoate showed an increase in incidence ...
  77. [77]
  78. [78]
    4-(4-chlorophenyl)piperidin-4-ol | 39512-49-7 | Benchchem
    $$50 deliveryMethodological Answer : The compound is typically synthesized via Grignard reactions, where 4-chlorophenylmagnesium bromide reacts with N-protected piperidinone ...
  79. [79]
    Haloperidol - an overview | ScienceDirect Topics
    Haloperidol, 4-[4-(p-chlorophenyl)-4-hydroxypiperidino]-4′-fluorobutyrophenone (6.3.8), is synthesized by the alkylation of 4-(4-chlorophenyl)-4- ...
  80. [80]
    [PDF] Haldol - accessdata.fda.gov
    HALDOL (haloperidol) is indicated for use in the treatment of schizophrenia. HALDOL is indicated for the control of tics and vocal utterances of Tourette's ...<|separator|>
  81. [81]
    [PDF] PRODUCT MONOGRAPH Pr Haloperidol Decanoate Injection ...
    Keep in carton until discarded. Haloperidol Decanoate Injection (haloperidol decanoate injection), 50 mg and 100 mg haloperidol/mL, is available in 1 mL single ...
  82. [82]
  83. [83]
    [The discovery of haloperidol] - PubMed
    Haloperidol was synthesized on the 11th of February 1958 by Bert Hermans at the Janssen Laboratories, Beerse, Belgium. Simple but ingenuous methods of ...Missing: development | Show results with:development
  84. [84]
    The consolidation of neuroleptic therapy: Janssen, the discovery of ...
    This substance was synthesized on the 11th February 1958 and received the generic name of haloperidol because of the two halogenated substitutes incorporated ...
  85. [85]
    The haloperidol story - PubMed
    Haloperidol was synthesized on the 11th of February 1958 at the Janssen Laboratories, in Belgium. Soon after its synthesis and animal studies, ...
  86. [86]
    Paul Janssen: From Haloperidol to Risperidone. David Healy's ...
    Clozapine was synthesized for the first time by a chemist working for the Swiss company Wander. Stille did the pharmacology and he realised that it was a weak ...Missing: patent | Show results with:patent
  87. [87]
    Haloperidol - American Chemical Society
    Apr 29, 2019 · Haloperidol is a venerable antipsychotic drug that is used to treat schizophrenia, Tourette syndrome, bipolar disorder, and other conditions.
  88. [88]
    Placebo Response in Antipsychotic Clinical Trials: A Meta-analysis
    Patients with schizophrenia have baseline cognitive difficulties that may be exacerbated during periods of acute psychosis, which may impair their capacity to ...
  89. [89]
    European clinical guidelines for Tourette syndrome and other tic ...
    Nov 10, 2021 · ... Drug Administration (FDA), e.g. haloperidol (in 1969) and pimozide (in 1984), whereas aripiprazole was approved in 2014. In European ...<|separator|>
  90. [90]
    Haloperidol Update: 1975 - Frank J Ayd, 1976
    First published January 1976. Haloperidol Update: 1975. Frank J Ayd, JrView all authors and affiliations. Volume 69, Issue 1_suppl.
  91. [91]
    Drug Approval Package: Haloperidol NDA #75-176
    Nov 20, 2001 · Haloperidol Decanoate Injection. Company: King Pharmaceuticals, Inc. Application No.: 75-176. Approval Date: 2/9/2000. Date created: November 20, 2001.
  92. [92]
    Haloperidol - eEML - Electronic Essential Medicines List
    First added in 1977 (TRS 615) for Schizophrenia or other primary psychotic disorders. Added in 1991 (TRS 825) for Palliative care. Wikipedia. Haloperidol.
  93. [93]
    Updates in palliative care – recent advancements in the ...
    The most frequently recommended antipsychotic in palliative care is haloperidol (a first-generation butyrophenone antipsychotic drug), followed by the newer ...
  94. [94]
    The application of Haloperidol in end-of-life care - PDC Rx
    Oct 25, 2023 · Control of Agitation and Aggression: Haloperidol can be used in emergency situations to manage severe agitation or aggression. Nausea and ...
  95. [95]
    Haldol and associated names - referral - European Medicines Agency
    Feb 24, 2017 · On 23 February 2017, the European Medicines Agency completed a review of Haldol. The Agency's Committee for Medicinal Products for Human Use ...<|separator|>
  96. [96]
    Label: HALOPERIDOL solution - DailyMed
    Packager: PAI Holdings, LLC dba PAI Pharma. Category: HUMAN PRESCRIPTION DRUG LABEL; DEA Schedule: None; Marketing Status: Abbreviated New Drug Application ...
  97. [97]
    [PDF] Dynamics of Demand for Prescription Drugs after Patent Expiration
    Feb 19, 2008 · ... patents expired during December 1984 - June 1985; (ii) late markets (doxepin, maprotiline and haloperidol) refer to the drugs that have ...
  98. [98]
  99. [99]
    Haloperidol Prices, Coupons, Copay Cards & Patient Assistance
    Oral Tablet ; 0.5 mg · $19.51 · 100, $0.20, $19.51 ; 1 mg · $19.25 · 100, $0.19 – $0.23, $19.25 – $23.47 ; 2 mg · $22.06 · 100, $0.22 – $0.45, $22.06 – $44.91 ; 5 mg ...
  100. [100]
  101. [101]
  102. [102]
    About haloperidol - NHS
    feelings of agitation or anxiety · schizophrenia, where you may see, hear or feel things that are not there, believe things that are not true, feel unusually ...Missing: maintenance acute
  103. [103]
    Drug Shortage Detail: Haloperidol Decanoate Injection - ASHP
    Fresenius Kabi had haloperidol decanoate on shortage due to short-term manufacturing delays. Mylan discontinued haloperidol decanoate 5 mL multi-dose vials.Missing: 2020s | Show results with:2020s
  104. [104]
    Table 1, Cost Comparison Table of Antipsychotic Drugs for ... - NCBI
    Drug/Comparator, Strength, Dosage Form, Price ($), Recommended Dose, Average Daily Drug Cost ($), Average Annual Drug Cost ($) ...
  105. [105]
    [PDF] WHO-PQ RECOMMENDED SUMMARY OF PRODUCT ...
    Nov 4, 2024 · This summary of product characteristics focuses on uses of the medicine covered by WHO's Prequalification ... haloperidol, risperidone and ...
  106. [106]
    Haloperidol for Pain Management: A Narrative Review - PMC
    Aug 21, 2024 · The sigma-1 receptor is a unique ligand-operated chaperone present in crucial areas for pain control in both the peripheral and central nervous ...
  107. [107]
    [PDF] Nausea and vomiting
    Oct 21, 2024 · Haloperidol for the treatment of nausea and vomiting in palliative care patients. 2015 Available from: http://onlinelibrary.wiley.com/doi ...<|separator|>
  108. [108]
    Haloperidol for the treatment of nausea and vomiting in palliative ...
    To evaluate the efficacy and adverse events associated with the use of haloperidol for the treatment of nausea and vomiting in palliative care patients.
  109. [109]
    Haloperidol in treating delirium, reducing mortality, and preventing ...
    Mar 20, 2025 · This study aimed to comprehensively evaluate the efficacy and safety of haloperidol for delirium treatment and prevention in ICU patients.
  110. [110]
    The efficacy and safety of haloperidol for the treatment of delirium in ...
    The use of haloperidol as compared to placebo has no significant effect on delirium-free days, overall mortality, length of intensive care unit and/or hospital ...