MOBILE VIEW  | 

CITALOPRAM AND RELATED AGENTS

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Citalopram hydrobromide is a phthalane derivative like other second generation antidepressant agents. However, it is currently the most highly selective and potent serotonin reuptake inhibitor. Citalopram enhances serotoninergic neurotransmission through selective and potent inhibition of neuronal serotonin reuptake. Anticholinergic effects may occur with this agent.
    B) Escitalopram is a selective serotonin reuptake inhibitor (SSRI) and is the S(+)-enantiomer of citalopram.

Specific Substances

    A) CITALOPRAM (synonym)
    1) LU-10-171
    2) Nitalapram hydrobromide
    3) 1-(3-dimethylaminopropyl)-1-(p-fluorophenyl)-5-phthalan carbonitrile
    4) CAS 59729-33-8 (citalopram)
    5) CAS 59729-32-7 (citalopram hydrobromide)
    6) Molecular Formula: C20-H21-F-N2-O, HBr
    ESCITALOPRAM (synonym)
    1) S-Citalopram (synonym)
    2) S(+)-Citalopram (synonym)
    3) LU-26-054
    4) Cipralex

Available Forms Sources

    A) USES
    1) Citalopram is used in the treatment of depression (Prod Info Celexa(R) oral tablets, 2014).
    2) Escitalopram is approved for the acute and maintenance treatment of depression in adults and pediatric patients aged 12 to 17 years. It is also approved to treat generalized anxiety disorder in adults (Prod Info Lexapro(R) oral tablets, oral solution, 2014).

Life Support

    A) This overview assumes that basic life support measures have been instituted.

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Selective serotonin reuptake inhibitors (SSRIs) used to treat major depressive disorder. Also used for treating obsessive-compulsive disorder, panic disorder, premenstrual dysphoric syndrome, anxiety disorder, and posttraumatic stress disorder.
    B) PHARMACOLOGY: Selectively inhibits the presynaptic reuptake of serotonin. Stimulation of postsynaptic 5-HT1 receptors results in the antidepressant and anxiolytic effects.
    C) TOXICOLOGY: Many of the toxic effects are mediated by stimulation of the 5-HT2 receptors causing excessive serotonin effect or serotonin syndrome.
    D) EPIDEMIOLOGY: Overdose is becoming increasingly frequent. Overdose of a single SSRI is usually well tolerated with mild to moderate severity. More severe toxicity (serotonin syndrome) may develop when another agent that increases CNS serotonin is ingested in addition to citalopram.
    E) WITH THERAPEUTIC USE
    1) ADVERSE EFFECTS: COMMON: Nausea, vomiting, dry mouth, somnolence, insomnia, sweating, tremor, diarrhea, dyspepsia, anxiety, decreased libido, asthenia, myalgia, rash, and weight gain.
    2) ADVERSE EFFECTS: SEVERE: Suicidality, worsening depression, serotonin syndrome, mania, seizure, elevated liver enzymes, hyponatremia, SIADH, priapism, anaphylactoid reaction, hypoglycemia, extrapyramidal reaction, tachycardia, abnormal platelet aggregation, and hepatitis. Abrupt cessation can result in withdrawal syndrome.
    3) Escitalopram is the S(+)-enantiomer of citalopram, and the adverse effects profile is similar to that of citalopram.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Agitation, confusion, tremor, nausea, vomiting, hyperreflexia, occasional clonus and myoclonus, hypoglycemia, hypertension, and bradycardia.
    2) SEVERE TOXICITY: Seizure, rigidity, hyperthermia, hypertension or hypotension, QRS and QTc interval prolongation, coma, and rarely, death. Serotonin syndrome may develop and is characterized by mental status changes (confusion, hypomania), agitation, myoclonus, hyperreflexia, diaphoresis, shivering, tremor, diarrhea, incoordination, and fever. Wide complex tachyarrhythmias, torsade de pointes, and cardiac arrest have all been observed after massive citalopram overdose, sometimes occurring quite late after the ingestion. Neuroleptic malignant syndrome has been reported following a citalopram overdose.
    0.2.20) REPRODUCTIVE
    A) Citalopram and escitalopram are classified as FDA pregnancy category C. Limited data on the use of citalopram during human pregnancy have not demonstrated apparent teratogenic risk; however, SSRI administration lasting more than 30 days during the second or third lunar month of pregnancy was associated with a significant 80% increased risk of clubfoot occurrence in infants. In addition, third trimester exposure has been associated with an increased risk for nonteratogenic effects (pulmonary hypertension of the newborn (PPHN) and findings consistent with serotonin syndrome) and neonatal intensive care. Animal studies demonstrated fetal abnormalities, reduced birth weights, death, and mild maternal toxicity with citalopram and escitalopram use during pregnancy; however, these occurred at doses considerably greater than the maximum recommended human dose. The manufacturer suggests considering a dose reduction of citalopram or escitalopram during the third trimester of pregnancy. Citalopram and escitalopram are excreted in human breast milk and long-term effects of exposure to SSRIs via breast milk on the cognitive development of the infant have not been determined. In fertility studies of citalopram use in male and female rats, reduced fertility and increased duration of pregnancy were observed at doses 5 and 8 times higher than the recommended human dose, respectively. An increased risk for social-behavioral abnormalities at 2 to 6 years of age was reported in children exposed to SSRIs or SNRIs in utero who developed neonatal abstinence syndrome (NAS) at birth.

Laboratory Monitoring

    A) Monitor vital signs (including temperature) and mental status.
    B) Monitor serum electrolytes (including potassium, bicarbonate) following significant overdose.
    C) Obtain baseline ECG, continuous cardiac monitoring, and serial ECGs following a significant exposure.
    D) Monitor serial blood glucose, especially in patients with depressed mental status; severe hypoglycemia has been reported after citalopram overdose.
    E) Plasma concentration is not readily available and does not correlate well with therapeutic or adverse effects. It is not indicated for the acute management of overdose.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Most patients require only supportive care. Control agitation and confusion with either benzodiazepines or serotonin antagonist such as cyproheptadine or chlorpromazine. Hypertension and tachycardia are generally mild and well tolerated, and do not require specific treatment.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Early intubation, neuromuscular paralysis, ventilation assistance, and aggressive cooling should be performed if the patient presents with respiratory depression, severe muscle rigidity, and severe hyperthermia. Adequate circulatory support with IV fluids and vasopressors (if needed) should be assured if patient presents with circulatory collapse. Treat seizures with benzodiazepines; use barbiturates or propofol for recurrent seizures. Patients with wide-complex dysrhythmias should be treated with hypertonic sodium bicarbonate boluses. Although QTc interval prolongation is well described, torsade de pointes is rare. If torsade de pointes occurs, patients should be treated using standard interventions (magnesium sulfate 2 g IV, external or internal cardiac pacing).
    C) DECONTAMINATION
    1) PREHOSPITAL: Activated charcoal can be considered within the first hour after a large ingestion (greater than 600 mg citalopram), if the patient has an appropriate level of consciousness, has a patent airway, and is able to drink the charcoal.
    2) HOSPITAL: Administer activated charcoal if the patient presents early after large ingestion (greater than 600 mg citalopram), if the patient has an appropriate level of consciousness, patent airway and can drink the charcoal, or if the patient is intubated. Severe toxicity is rare; gastric lavage is rarely, if ever, warranted.
    D) AIRWAY MANAGEMENT
    1) Perform early with neuromuscular paralysis if the patient presents with respiratory or CNS depression, severe muscle rigidity, or severe hyperthermia.
    E) ANTIDOTE
    1) CYPROHEPTADINE: A serotonin antagonist with high affinity for the 5-HT2 receptors; effective for milder cases of serotonin syndrome. Dose: ADULT: 12 mg orally or nasogastric tube, followed by 4 to 8 mg every 4 to 6 hours. CHILD: 0.25 mg/kg/day orally or nasogastric tube, divided every 6 hours, maximum dose 12 mg/day.
    2) CHLORPROMAZINE: A phenothiazine antipsychotic with 5-HT2 antagonist activity; indicated in severe serotonin syndrome cases. Dose: 12.5 to 50 mg IV, followed by 25 to 50 mg every 6 hours. It is NOT generally recommended because it may cause severe hypotension.
    F) SEIZURES
    1) IV benzodiazepines, barbiturates, or propofol for recurrent seizures.
    G) SEROTONIN SYNDROME
    1) IV benzodiazepines, cooling measures. Cyproheptadine may be considered (ADULT: 12 mg orally or nasogastric (NG) tube, followed by 4 to 8 mg orally or NG tube every 4 to 6 hours if symptoms persist, up to a maximum of 32 mg in 24 hours. CHILD: 0.25 mg/kg/day divided every 6 hours, maximum dose 12 mg/day). Severe cases have been managed with benzodiazepine sedation and neuromuscular paralysis with nondepolarizing agents.
    H) TORSADES DE POINTE
    1) Hemodynamically unstable patients require electrical cardioversion. Emergent treatment with magnesium or atrial overdrive pacing is indicated. Detect and correct underlying electrolyte abnormalities.
    I) ENHANCED ELIMINATION
    1) Hemodialysis and hemoperfusion are NOT of value due to the large volume of distribution.
    J) PATIENT DISPOSITION
    1) HOME CRITERIA: Children and adults with mild symptoms (eg, vomiting, mydriasis, diaphoresis, mild somnolence) following an inadvertent ingestion of up to 100 mg citalopram or 50 mg escitalopram can be managed at home with instructions to call the poison center if symptoms develop. For patients already on citalopram or escitalopram, those with inadvertent ingestions of up to 5 times their own single therapeutic dose can be observed at home with instructions to call the poison center back if symptoms develop.
    2) OBSERVATION CRITERIA: Patients with deliberate ingestions, those with more than mild symptoms, and ingestions of more than 100 mg citalopram or 50 mg escitalopram should be sent to a health care facility. Patients on chronic citalopram or escitalopram therapy should be sent to a healthcare facility if they ingest more than 5 time their own single therapeutic dose. Ingestion of more than 600 mg citalopram or more than 300 mg escitalopram requires cardiac monitoring for 8 hours (11 hours if the patient did not receive activated charcoal within 4 hours of ingestion). Ingestion of more than 1000 mg citalopram or more than 500 mg escitalopram requires cardiac monitoring for 13 hours. At the end of the observation period, if the QTc is less than 450 msec, the monitoring can be discontinued, and the patient may be discharged if asymptomatic. Patients with symptoms of toxicity or a QTc of greater than 450 msec at the end of the observation period should be admitted for continued cardiac monitoring.
    3) ADMISSION CRITERIA: Patients with mild to moderate toxicity after intentional overdose need to be monitored for prolonged QTc for at least 13 hours. Patients with severe toxicity need to be admitted to an intensive care unit.
    4) CONSULT CRITERIA: Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity or an unclear diagnosis.
    K) PITFALLS
    1) Failure to aggressively control agitation, muscle rigidity, and hyperthermia with adequate benzodiazepines, intubation with neuromuscular paralysis, and body cooling.
    L) PHARMACOKINETICS
    1) CITALOPRAM: Peak plasma concentrations within 2 to 4 hours. Volume of distribution of 15 L/kg (large), protein binding 50%, and elimination half-life approximately 33 hours. Extensively metabolized by the liver.
    2) ESCITALOPRAM: Peak plasma concentrations within 3 to 4 hours. Volume of distribution of 1330 L and elimination half-life is 27 to 32 hours. Extensively metabolized by the liver via CYP 450 enzymes.
    M) DIFFERENTIAL DIAGNOSIS
    1) Other SSRI poisoning, serotonin syndrome, neuroleptic malignant syndrome, CNS infection, hypoglycemia, cocaine or amphetamine poisoning.

Range Of Toxicity

    A) Acute ingestion of up to 100 mg citalopram or 50 mg escitalopram is not expected to result in toxicity. Serotonin toxicity may develop at therapeutic doses, particularly if another medication that increases CNS serotonin is used concomitantly. Minor toxicity develops with overdose less than 600 mg citalopram. Ingestion of more than 600 mg citalopram or more than 300 mg escitalopram requires cardiac monitoring for 8 hours (11 hours if the patient did not receive activated charcoal within 4 hours of ingestion). Ingestion of more than 1000 mg citalopram or more than 500 mg escitalopram requires cardiac monitoring for 13 hours. At the end of the observation period, if the QTc is less than 450 msec, the monitoring can be discontinued, and the patient may be discharged if asymptomatic. Patients with symptoms of toxicity or a QTc of greater than 450 msec at the end of the observation period should be admitted for continued cardiac monitoring. Seizures have been reported after ingestions of more than 600 mg citalopram, and serious toxicity after ingestion of 800 mg or more. PEDIATRIC: QTc prolongation has been reported after ingestion of escitalopram 200 mg.
    B) THERAPEUTIC DOSE: ADULT: Citalopram: Initially, 20 mg/day orally; max dose 40 mg/day. Escitalopram: 10 to 20 mg/day orally; max dose 20 mg/day. PEDIATRIC: Safety and effectiveness in children have not been established.

Summary Of Exposure

    A) USES: Selective serotonin reuptake inhibitors (SSRIs) used to treat major depressive disorder. Also used for treating obsessive-compulsive disorder, panic disorder, premenstrual dysphoric syndrome, anxiety disorder, and posttraumatic stress disorder.
    B) PHARMACOLOGY: Selectively inhibits the presynaptic reuptake of serotonin. Stimulation of postsynaptic 5-HT1 receptors results in the antidepressant and anxiolytic effects.
    C) TOXICOLOGY: Many of the toxic effects are mediated by stimulation of the 5-HT2 receptors causing excessive serotonin effect or serotonin syndrome.
    D) EPIDEMIOLOGY: Overdose is becoming increasingly frequent. Overdose of a single SSRI is usually well tolerated with mild to moderate severity. More severe toxicity (serotonin syndrome) may develop when another agent that increases CNS serotonin is ingested in addition to citalopram.
    E) WITH THERAPEUTIC USE
    1) ADVERSE EFFECTS: COMMON: Nausea, vomiting, dry mouth, somnolence, insomnia, sweating, tremor, diarrhea, dyspepsia, anxiety, decreased libido, asthenia, myalgia, rash, and weight gain.
    2) ADVERSE EFFECTS: SEVERE: Suicidality, worsening depression, serotonin syndrome, mania, seizure, elevated liver enzymes, hyponatremia, SIADH, priapism, anaphylactoid reaction, hypoglycemia, extrapyramidal reaction, tachycardia, abnormal platelet aggregation, and hepatitis. Abrupt cessation can result in withdrawal syndrome.
    3) Escitalopram is the S(+)-enantiomer of citalopram, and the adverse effects profile is similar to that of citalopram.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Agitation, confusion, tremor, nausea, vomiting, hyperreflexia, occasional clonus and myoclonus, hypoglycemia, hypertension, and bradycardia.
    2) SEVERE TOXICITY: Seizure, rigidity, hyperthermia, hypertension or hypotension, QRS and QTc interval prolongation, coma, and rarely, death. Serotonin syndrome may develop and is characterized by mental status changes (confusion, hypomania), agitation, myoclonus, hyperreflexia, diaphoresis, shivering, tremor, diarrhea, incoordination, and fever. Wide complex tachyarrhythmias, torsade de pointes, and cardiac arrest have all been observed after massive citalopram overdose, sometimes occurring quite late after the ingestion. Neuroleptic malignant syndrome has been reported following a citalopram overdose.

Heent

    3.4.3) EYES
    A) WITH THERAPEUTIC USE
    1) In a meta-analysis of clinical studies, disturbances in accommodation were reported in approximately 9% of depressed patients treated with citalopram (n=746) (Milne & Goa, 1991).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) ELECTROCARDIOGRAM ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) Although ECG abnormalities appear to be less frequent with therapeutic citalopram use than with tricyclic antidepressant use, QRS widening, QTc prolongation, sinus tachycardia, wide complex tachycardia, and inferolateral repolarization abnormalities have been reported in overdose (Lung et al, 2013; Lung et al, 2012; Jimmink et al, 2008; Duncan et al, 2008; Kelly et al, 2004; Grundemar et al, 1997; Personne et al, 1997). Left bundle-branch block has also been reported (Snider , 2001).
    b) Widened QRS complexes have been observed at doses above 600 mg (15 to 30 times the usual therapeutic dose) (Lung et al, 2013; Grover & Caravati, 2001; Power, 1998; Grundemar et al, 1997).
    c) CASE SERIES
    1) In a retrospective review of reported citalopram overdoses in 26 patients, QTc interval prolongation occurred in 8 patients and bundle-branch block (BBB) occurred in 3 patients (2 patients with left BBB and 1 patient with right BBB). Atrioventricular block was reported in 1 patient and extrasystoles, in 2 patients. Among the 26 patients, the estimated amounts ingested ranged from 200 to 4960 mg (median 1190 mg) (Jimmink et al, 2008).
    2) In a series of 225 cases of overdose with newer antidepressants, patients suffering citalopram overdose (n=88) had significantly longer QT intervals on ECG than those taking venlafaxine, mirtazapine, or nefazodone (Kelly et al, 2004).
    3) In a review of 108 cases of citalopram overdose, approximately 25% of the patients had ECG changes such as nonspecific changes of the ST-T region and moderate widening of the ECG complexes. However, no clinically significant arrhythmias were reported (Personne et al, 1997).
    d) CASE REPORTS
    1) A 54-year-old woman with psychotic depression treated with citalopram 20 mg/day and zopiclone 7.5 mg/day developed delayed dysrhythmias after intentionally ingesting an unknown amount of citalopram with alcohol. She presented to the emergency department (time of ingestion unknown) with altered mental status, hypotension, and low O2 saturation. A positive bilateral Babinski (without sensory motor deficit) and jugular vein distension were noted upon examination. Laboratory analysis showed a blood ethanol level of 0.2 g/L, lactic acidosis, hypokalemia, metabolic acidosis, and a serum citalopram level of 5.88 mg/L (therapeutic range 0.01 to 0.2 mg/L). Urine screening was negative for opiates, cocaine, benzodiazepines, and tricyclic antidepressants. She was treated with 50 g activated charcoal and admitted to ICU. Upon admission to ICU, she seized and developed ventricular fibrillation (VF) which was treated successfully with midazolam and defibrillation. A prolonged QT interval of 0.44 seconds (QTc 0.62 seconds) with left bundle branch block was noted on ECG; however, cardiac enzymes were negative for myocardial infarction. Sustained ventricular tachycardia with VF episodes occurred 10 hours after ICU admission and continued for 48 hour, requiring repeated defibrillation. She recovered fully and was discharged 16 days after admission (Liotier & Coudore, 2011).
    2) A 31-year-old man developed sinus tachycardia (130 beats/min) with a widened QRS complex (124 msec) with a left bundle-branch block, and prolonged QTc (506 msec) after ingesting 400 mg citalopram. The QRS complex narrowed (96 msec) with resolution of the left bundle-branch block pattern, and the QTc interval shortened (495 msec) after administration of sodium bicarbonate (Engebretsen et al, 2003).
    3) An 82-year-old woman developed a generalized tonic-clonic seizure approximately 12 hours after ingesting citalopram 1.6 g in a suicide attempt. Although her cardiovascular and pulmonary examinations were unremarkable, an initial ECG showed sinus bradycardia with a ventricular rate of 58 beats/min, left axis deviation, and left bundle-branch block, which were consistent with her previous ECGs. In addition, the QRS duration, QT interval, and QTCs interval were 146 msec, 544 msec, and 534 msec, respectively. Three hours after she was transferred to telemetry service, marked bradycardia was observed. A 12-lead ECG showed a junctional escape rhythm with a ventricular rate of 40 beats/min, retrograde ventriculoatrial activation, and an underlying left bundle-branch block. The QRS duration, QT interval, and QTc interval were 152 msec, 646 msec, and 527 msec, respectively. The administration of sodium bicarbonate solution 50 mEq (50 mL of 8.4% solution) by direct intravenous injection on 2 different occasions caused a dramatic but temporary impact on the ECG findings. These effects quickly reverted to the previous junctional bradycardia. Approximately 3 hours after starting a continuous infusion of sodium bicarbonate 150 mEq in 1 liter of 5% dextrose in water (a rate of 75 mL/hour or 11.25 mEq/hour), normal sinus rhythm with a rate of 63 beats/min, a QT interval of 510 msec, and a QTc interval 504 msec were noted. After 36 hours, the infusion was stopped and no new ECG abnormalities were observed (Brucculeri et al, 2005).
    4) Persistent bidirectional wide-complex tachycardia, QRS prolongation, QTc widening, left bundle-branch block, and seizures were reported in a 22-year-old woman who ingested citalopram 1200 mg in a suicide attempt. She reverted to normal sinus rhythm within 5 hours of presentation (Sztajnkryeer et al, 2003).
    5) A 36-year-old woman developed palpitation, weakness, nausea, and numbness 36 hours after taking citalopram 1000 mg with alcohol. On day 3, her vital signs were blood pressure, 84/44 mmHg; temperature, 99.3 degrees F; pulse, 102 to 160 beats/min; and respiratory rate, 17 breaths/min. ECG revealed intermittent runs of wide-complex tachycardia with a QTc of 600 msec. Despite saline and magnesium sulfate infusions, she developed intermittent bigeminy, and despite IV lidocaine, intermittent torsade de pointes occurred. After receiving potassium chloride, transvenous pacemaker insertion, and isoproterenol infusion, she converted to normal sinus rhythm, and QTCs interval narrowed to 442 msec after 2 days. Her plasma citalopram level was 477 ng/mL (therapeutic range 40 to 110 ng/mL), with a corresponding desmethylcitalopram level of 123.2 ng/mL (therapeutic range 14 to 40 ng/mL) (Tarabar et al, 2003).
    6) A 21-year-old ingested 20 citalopram 20 mg tablets after drinking a "couple of beers" (blood alcohol 121 mg/dL) and taking one alprazolam 0.25 mg tablet. The patient received gastrointestinal decontamination. One hour after ingestion, the patient was in normal sinus rhythm with a QTc interval of 380 msec, and within 2 hours she had a QTc interval of 438 msec. At 7 hours the QTc was 450 msec, and at 13 hours it was 457 msec. Subsequent monitoring showed a QTc of 393 msec approximately 20 hours after ingestion. The delay in peaked effect was likely due to the cardiotoxicity of the metabolite DDCT. The authors noted that this overdose was relatively small (less than 7 times the recommended maximum daily dose of 60 mg), but QT prolongation still occurred (Catalano et al, 2001).
    7) Right bundle-branch block and QRS prolongation (136 msec) were reported in a 21-year-old man following an overdose of citalopram 1800 mg, fluoxetine 1000 mg, and acetaminophen 2500 mg. The patient's citalopram serum level was 840 ng/mL (ten times the reported therapeutic concentration of 80 ng/mL). ECG abnormalities resolved spontaneously (QRS 100 msec; normal axis) within 36 hours of ingestion (Grover & Caravati, 2001).
    8) Five cases of QTc prolongation and sinus tachycardia have been reported following severe nonfatal citalopram overdoses (range 400 to 5200 mg) (Grundemar et al, 1997).
    9) Wide-complex right bundle-branch block with multiple episodes of monomorphic ventricular tachycardia, and a widened QRS were reported in a 19-year-old man who intentionally ingested an unknown amount of citalopram. The serum citalopram concentration, obtained at time of presentation, was 1500 ng/mL (therapeutic, 9 to 200 ng/mL). The patient's ECG abnormalities resolved following supportive therapy (Maddry et al, 2013).
    10) ESCITALOPRAM: A 32-year-old woman developed nausea, vertigo, confusion, palpitation, drowsiness, slight leukocytosis, mild fever, tachycardia (pulse 120 beats/min), and mild hypotension (blood pressure 100/70 mmHg) after ingesting escitalopram 190 mg. ECG revealed an increased QT interval and sinus tachycardia. Following supportive care, she recovered and was discharged 24 hours after hospitalization (Yuksel et al, 2005). Another patient, a 14-year-old girl, experienced a prolonged QTc interval after ingesting escitalopram 200 mg. She presented to the emergency department approximately 24 hours postingestion, and an ECG showed normal sinus rhythm with a QTc interval of 450 msec. The following day (2 days postingestion), a repeat ECG showed a QTc interval of 469 msec. That same day, her QTc interval normalized to 420 msec spontaneously, and she was transferred to a psychiatric facility (Scharko & Schumacher, 2008).
    11) Approximately 16 hours after a 46-year-old woman intentionally ingested 1400 mg of citalopram, an ECG was performed revealing a prolonged QTc interval of 541.6 ms. At approximately 29 hours post-ingestion, the initial citalopram serum concentration was 1231 ng/mL. Over the course of several days, the patient's serum citalopram concentration and QTc interval were determined within 24 hours of each other in 6 different instances, and determined on the same date in 5 cases, showing a similar rate of decline, indicating a direct correlation between the serum concentration of citalopram and QTc interval prolongation that was statistically significant (r=0.943; p<0.005) (Unterecker et al, 2012).
    12) ESCITALOPRAM: A 16-year-old girl, with a history of depression, presented to the emergency department with vomiting and slight lethargy approximately 2.5 hours after ingesting 500 mg of escitalopram, 25 tablets of tramadol/acetaminophen, and an unknown amount of hydrocodone/acetaminophen. An ECG revealed an incomplete right bundle branch block with a widened QRS complex (110 ms) and a QTc interval of 482 ms. Ten minutes following administration of an IV bolus of sodium bicarbonate (100 mEq), a repeat ECG revealed narrowing of the QRS complex (102 ms) and QTc interval prolongation (509 ms). A sodium bicarbonate infusion and IV magnesium sulfate were subsequently initiated, resulting in normalization of her QRS complex and QTc interval. The patient's serum escitalopram concentration, obtained approximately 8 hours post-ingestion, was 45 mcg/L (Schreffler et al, 2013).
    B) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Hypotension has been reported following nonfatal citalopram overdose (range 400 to 5200 mg) in several patients (Kelly et al, 2003; Rothenhausler et al, 2000; Grundemar et al, 1997) and in a fatal overdose with a potential maximum citalopram ingestion of 1.8 g, ingested in combination with other agents, including bupropion, clonazepam, and trazodone (Lung et al, 2012).
    b) CASE REPORT: A 54-year-old woman with psychotic depression treated with citalopram 20 mg/day and zopiclone 7.5 mg/day developed severe hypotension (65/25 mmHg) after intentionally ingesting an unknown amount of citalopram with alcohol. She presented to the emergency department (time of ingestion unknown) with altered mental status, low O2 saturation. and a heart rate of 77 beats/min. After treatment with activated charcoal, she was admitted to ICU where a prolonged QT interval of 0.44 seconds (QTc 0.62 seconds) with left bundle branch block was noted on ECG after an episode of ventricular fibrillation (VF). Sustained ventricular tachycardia with VF episodes occurred 10 hours after ICU admission and continued for 48 hour, requiring repeated defibrillation. She recovered fully and was discharged 16 days after admission (Liotier & Coudore, 2011).
    c) CASE REPORT: ESCITALOPRAM: A 32-year-old woman developed nausea, vertigo, confusion, palpitation, drowsiness, slight leukocytosis, mild fever, tachycardia (pulse 120 beats/min), and mild hypotension (blood pressure 100/70 mmHg) after ingesting escitalopram 190 mg. Following supportive care, she recovered and was discharged 24 hours after hospitalization (Yuksel et al, 2005).
    d) CASE REPORT: A 21-year-old man presented to the emergency department (ED) unresponsive and with seizures after an overdose ingestion of citalopram and olanzapine, with potential maximum ingestions of 11.6 g and 600 mg, respectively. An ECG indicated wide complex tachycardia. Peak serum citalopram concentration, measured within 30 minutes post-presentation, was 522 ng/mL. Approximately 2.5 hours after arrival to the ED, following a second seizure, the patient developed hypotension that worsened over the next several hours, requiring an epinephrine infusion to maintain a mean arterial pressure of 60 mmHg. Bradycardia (less than 40 beats/min) developed approximately 6.5 hours post-presentation, and was unresponsive to atropine and isoproterenol, necessitating placement of a transvenous pacemaker. Approximately 10 hours post-presentation, a third seizure occurred. Because of progressive cardiac and neurotoxicity, IV lipid emulsion therapy was administered, with a total dose of 10% lipid emulsion at 21 mL/hour (0.005 mL/kg/min for a total of 17 hours). This is a 50-fold lower dose than what is recommended. Following the infusion, the patient's condition stabilized with no further clinical worsening. The pacemaker was discontinued 24 hours post-presentation and the epinephrine infusion was titrated down and discontinued 5 hours later (Lung et al, 2013).
    C) TACHYARRHYTHMIA
    1) WITH POISONING/EXPOSURE
    a) Sinus tachycardia has been reported in 4 cases of nonfatal citalopram overdose (range 400 to 5200 mg) (Grundemar et al, 1997).
    b) CASE REPORT: Persistent bidirectional wide-complex tachycardia, QRS prolongation, QTc widening, left bundle-branch block, and seizures were reported in a 22-year-old woman who ingested citalopram 1200 mg in a suicide attempt. She reverted to normal sinus rhythm within 5 hours of presentation (Sztajnkryeer et al, 2003).
    c) CASE SERIES: In a retrospective review of reported citalopram overdoses, tachycardia occurred in 15 of 26 patients. Among the 26 patients, the estimated amounts ingested ranged from 200 to 4960 mg (median 1190 mg) (Jimmink et al, 2008).
    d) ESCITALOPRAM: A 32-year-old woman developed nausea, vertigo, confusion, palpitation, drowsiness, slight leukocytosis (12.5 K/ul), mild fever (37.4 degrees C), tachycardia (pulse 120 beats/min), and mild hypotension (blood pressure 100/70 mmHg) after ingesting escitalopram 190 mg. ECG revealed an increased QT interval and sinus tachycardia. Following supportive care, she recovered and was discharged 24 hours after hospitalization (Yuksel et al, 2005).
    e) CASE REPORT: A 24-year-old woman developed progressive hypotension, tonic-clonic seizures, QTc interval prolongation, and pulseless ventricular tachycardia, unresponsive to aggressive resuscitative measures including defibrillation, external pacing, and overdrive chemical pacing, resulting in death approximately 19 hours after a suspected overdose ingestion of citalopram, bupropion, clonazepam, and trazodone. Potentially, the maximum doses ingested were 1.8 g of citalopram, 13.5 g of bupropion, 90 mg of clonazepam, and 4.5 g of trazodone. Post-mortem serum concentrations revealed supratherapeutic concentrations of citalopram (400 ng/mL) and bupropion (440 ng/mL); citalopram, clonazepam, and trazodone serum concentrations were within the normal therapeutic ranges (Lung et al, 2012).
    D) SUPRAVENTRICULAR TACHYCARDIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 23-year-old man developed supraventricular tachycardia after intentionally ingesting citalopram 920 mg. Heart rate reached the 160s approximately 3 hours after exposure and approximately 20 minutes after he had a seizure; tachycardia was successfully treated with adenosine 6 mg. A repeat ECG was normal, with no QRS widening or QTc prolongation; no further episodes were reported. The patient was discharged within 24 hours for further psychiatric evaluation (Cuenca et al, 2004).
    E) TORSADES DE POINTES
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 36-year-old woman developed palpitation, weakness, nausea, and numbness 36 hours after taking citalopram 1000 mg with alcohol. On day 3, her vital signs were blood pressure, 84/44 mmHg; temperature, 99.3 degrees F; pulse, 102 to 160 beats/min; and respiratory rate, 17 breaths/min. ECG revealed intermittent runs of wide-complex tachycardia with a QTc of 600 msec. Despite saline and magnesium sulfate infusions, she developed intermittent bigeminy, and despite IV lidocaine, intermittent torsade de pointes occurred. After receiving potassium chloride, transvenous pacemaker insertion, and isoproterenol infusion, she converted to normal sinus rhythm, and QTCs interval narrowed to 442 msec after 2 days. Her plasma citalopram level was 477 ng/mL (therapeutic range 40 to 110 ng/mL), with a corresponding desmethylcitalopram level of 123.2 ng/mL (therapeutic range 14 to 40 ng/mL) (Tarabar et al, 2003).
    F) BRADYCARDIA
    1) WITH THERAPEUTIC USE
    a) In patients who had pretreatment heart rates of 48 to 60 beats/min, further declines in heart rate have occurred infrequently following therapeutic citalopram use (Nyth et al, 1992; Nyth & Gottfries, 1990).
    b) CASE REPORT: A 60-year-old woman developed bradycardia and had a presyncopal episode 2 weeks after starting citalopram 20 mg/day. Other medications included omeprazole 20 mg/day and alprazolam 1 mg as needed for anxiety. Heart rate was 30 to 50 beats/min on cardiac monitoring, and a 12-lead ECG indicated sinus bradycardia (39 beats/min) with a QTc of 370 msec. Following drug cessation, heart rate returned to baseline within 48 hours and Holter monitoring showed no significant abnormalities (Isbister et al, 2001).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: An 82-year-old woman developed a generalized tonic-clonic seizure approximately 12 hours after ingesting citalopram 1.6 g in a suicide attempt. Although her cardiovascular and pulmonary examinations were unremarkable, an initial ECG showed sinus bradycardia with a ventricular rate of 58 beats/min, left axis deviation, and left bundle-branch block, which were consistent with her previous ECGs. In addition, the QRS duration, QT interval, and QTCs interval were 146 msec, 544 msec, and 534 msec, respectively. Three hours after she was transferred to telemetry service, marked bradycardia was observed. A 12-lead ECG showed a junctional escape rhythm with a ventricular rate of 40 beats/min, retrograde ventriculoatrial activation, and an underlying left bundle-branch block. The QRS duration, QT interval, and QTc interval were 152 msec, 646 msec, and 527 msec, respectively. The administration of sodium bicarbonate solution 50 mEq (50 mL of 8.4% solution) by direct intravenous injection on 2 different occasions caused a dramatic but temporary impact on the ECG findings. These effects quickly reverted to the previous junctional bradycardia. Approximately 3 hours after starting a continuous infusion of sodium bicarbonate 150 mEq in 1 liter of 5% dextrose in water (a rate of 75 mL/hr or 11.25 mEq/hr), normal sinus rhythm with a rate of 63 beats/min, a QT interval of 510 msec, and a QTc interval 504 msec were noted. After 36 hours, the infusion was stopped, and no new ECG abnormalities were observed (Brucculeri et al, 2005).
    b) CASE REPORT: A 32-year-old woman developed severe sinus bradycardia (41 beats/min), hypotension, and episodes of syncope approximately 4 hours after ingesting citalopram 800 mg in a suicide attempt. ECG showed normal QRS morphology and QT interval values. The bradycardia, hypotension, and syncopal episodes resolved following insertion of a temporary pacemaker (Rothenhausler et al, 2000).
    c) CASE REPORT: A 21-year-old man presented to the emergency department (ED) unresponsive and with seizures after an overdose ingestion of citalopram and olanzapine, with potential maximum ingestions of 11.6 g and 600 mg, respectively. An ECG indicated wide complex tachycardia. Peak serum citalopram concentration, measured within 30 minutes post-presentation, was 522 ng/mL. Approximately 2.5 hours after arrival to the ED, following a second seizure, the patient developed hypotension that worsened over the next several hours, requiring an epinephrine infusion to maintain a mean arterial pressure of 60 mmHg. Bradycardia (less than 40 beats/min) developed approximately 6.5 hours post-presentation, and was unresponsive to atropine and isoproterenol, necessitating placement of a transvenous pacemaker. Approximately 10 hours post-presentation, a third seizure occurred. Because of progressive cardiac and neurotoxicity, IV lipid emulsion therapy was administered, with a total dose of 10% lipid emulsion at 21 mL/hour (0.005 mL/kg/min for a total of 17 hours). This is a 50-fold lower dose than what is recommended. Following the infusion, the patient's condition stabilized with no further clinical worsening. The pacemaker was discontinued 24 hours post-presentation and the epinephrine infusion was titrated down and discontinued 5 hours later (Lung et al, 2013).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A chest x-ray, performed on a 45-year-old man who had intentionally ingested citalopram hydrobromide 3000 mg (citalopram 2400 mg free base), showed bilateral basal shadowing of the lungs. Approximately 10 hours postingestion, the patient became hypoxic and was placed on continuous positive airway pressure (CPAP) ventilation and broad-spectrum antibiotics. A repeat chest x-ray showed bilateral diffuse alveolar shadowing. Twenty-four hours postingestion, the patient's condition worsened and required mechanical ventilation. The patient appeared to spontaneously improve and was extubated 10 days postingestion, although he continued to be hypoxic with persistent bilateral diffuse alveolar infiltrates seen on chest x-ray. With continued improvement, the patient was eventually discharged to psychiatric care 16 days postingestion (Kelly et al, 2003).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) In a review of 108 cases of citalopram overdose, seizures were reported at doses greater than 600 mg (Personne et al, 1997). Generalized seizures were reported following a nonfatal exposure of 400 mg in a 26-year-old woman (Grundemar et al, 1997).
    1) In a similar case, a 21-year-old man developed a seizure 14 hours after an intentional ingestion of citalopram 1800 mg (serum level 10 times the normal therapeutic range of 840 ng/mL), fluoxetine 1000 mg (normal serum level), and acetaminophen 2500 mg. The patient recovered without sequelae (Grover & Caravati, 2001).
    b) INCIDENCE: At doses of 600 to 1900 mg, 18% of patients (6 of 34 patients) developed seizures; the frequency of seizures increased to 47% (9 of 19 patients) at doses of 1900 to 5200 mg (Personne et al, 1997). A study of 88 citalopram overdose cases reported seizures in 5 patients who had ingested amounts ranging from 400 to 3000 mg (Kelly et al, 2004).
    c) CASE SERIES: In a retrospective review of reported citalopram overdoses, seizures occurred in 4 of 26 patients. Among the 26 patients, the estimated amounts ingested ranged from 200 to 4960 mg (median 1190 mg) (Jimmink et al, 2008).
    d) A population-based nested case control study was conducted over an 11-year period, evaluating elderly (65 years and greater) residents of Ontario, Canada who were hospitalized with their first seizure within 60 days of filling a prescription for citalopram, escitalopram, venlafaxine, paroxetine, sertraline, fluoxetine, duloxetine, fluvoxamine, or bupropion. A total of 2,987 patients were identified. Compared to bupropion, escitalopram monotherapy (odds ratio (OR) 1.93; 95% CI 1.48 to 2.53) and citalopram use (OR 1.89; 95% CI 1.48 to 2.40) were associated with the highest risk of seizures (Finkelstein et al, 2015).
    e) CASE REPORTS
    1) A 54-year-old woman with psychotic depression treated with citalopram 20 mg/day and zopiclone 7.5 mg/day intentionally ingested an unknown amount of citalopram with alcohol. She presented to the emergency department (time of ingestion unknown) with a Glasgow coma scale score of 8, severe hypotension, and low O2 saturation. A positive bilateral Babinski (without sensory motor deficit) and jugular vein distension were noted upon examination. She was treated with 50 g activated charcoal and admitted to ICU. Upon admission to ICU, she seized twice and developed ventricular fibrillation (VF) which was treated successfully with midazolam and defibrillation. Sustained ventricular tachycardia with VF episodes occurred 10 hours after ICU admission and continued for 48 hours, requiring repeated defibrillation. She recovered fully and was discharged 16 days after admission (Liotier & Coudore, 2011).
    2) Generalized tonic-clonic seizures occurred in 2 patients who developed severe hypoglycemia following overdose ingestions of citalopram (2760 mg in one patient and an unknown amount in the other patient). Both patients recovered with supportive care (Duncan et al, 2008).
    3) A 10-month-old child ingested an unknown amount of her grandmother's citalopram and developed refractory seizures shortly thereafter. Seizures recurred despite treatment with midazolam and fosphenytoin, and she required intubation and paralysis with succinylcholine as well as the addition of a third anticonvulsant, phenobarbital. Initial citalopram plasma level 1 hour after ingestion was 1400 ng/mL. She was discharged 48 hours postingestion without evident sequelae (Masullo et al, 2006).
    4) An 82-year-old woman developed a generalized tonic-clonic seizure approximately 12 hours after ingesting citalopram 1.6 g in a suicide attempt (Brucculeri et al, 2005).
    5) A 23-year-old man ingested citalopram 920 mg and developed a generalized tonic-clonic seizure which lasted approximately 2 minutes and was treated with ativan 1 mg. No further seizures occurred (Cuenca et al, 2004).
    6) A 31-year-old man had a witnessed generalized clonic seizure approximately 13 hours after an intentional ingestion of citalopram 400 mg and alcohol. No recurrence was reported (Engebretsen et al, 2003).
    7) A 45-year-old man developed tonic-clonic seizures approximately 2 hours after ingesting citalopram 3000 mg (citalopram 2400 mg as a racemic mixture). The seizures resolved following administration of IV diazepam (Kelly et al, 2003).
    8) Persistent bidirectional wide-complex tachycardia, QRS prolongation, QTc widening, left bundle-branch block (LBBB), and seizures were reported in a 22-year-old woman who ingested citalopram 1200 mg in a suicide attempt. She reverted to normal sinus rhythm within 5 hours of presentation (Sztajnkryeer et al, 2003).
    9) A 21-year-old man presented to the emergency department (ED) unresponsive and with seizures after an overdose ingestion of citalopram and olanzapine, with potential maximum ingestions of 11.6 g and 600 mg, respectively. An ECG indicated wide complex tachycardia. Peak serum citalopram concentration, measured within 30 minutes post-presentation, was 522 ng/mL. Approximately 2.5 hours after arrival to the ED, following a second seizure, the patient developed hypotension that worsened over the next several hours, requiring an epinephrine infusion to maintain a mean arterial pressure of 60 mmHg. Bradycardia (less than 40 beats/min) developed approximately 6.5 hours post-presentation, and was unresponsive to atropine and isoproterenol, necessitating placement of a transvenous pacemaker. Approximately 10 hours post-presentation, a third seizure occurred. Because of progressive cardiac and neurotoxicity, IV lipid emulsion therapy was administered, with a total dose of 10% lipid emulsion at 21 mL/hour (0.005 mL/kg/min for a total of 17 hours). This is a 50-fold lower dose than what is recommended. Following the infusion, the patient's condition stabilized with no further clinical worsening. The pacemaker was discontinued 24 hours post-presentation and the epinephrine infusion was titrated down and discontinued 5 hours later (Lung et al, 2013).
    B) SEROTONIN SYNDROME
    1) WITH POISONING/EXPOSURE
    a) Three cases of fatal serotonin syndrome (tremor, hyperpyrexia, and seizures) occurred following overdoses involving citalopram and moclobemide. The three males died within 3 to 16 hours after overdose (Neuvonen et al, 1993).
    1) Citalopram serum concentration was at a therapeutic level in one patient and 2 and 5 times therapeutic levels in the other cases.
    2) Moclobemide serum concentrations were 5 times therapeutic levels in one patient and 20 to 50 times higher in the other two cases.
    b) CASE REPORT: A 58-year-old woman developed mild symptoms of serotonin syndrome (anxiety, nausea, tremor, muscle rigidity) following an inadvertent dose of 100 mg. Symptoms began within 2 hours of ingestion; staggering gait ataxia was the only residual symptom reported 5 days after exposure (Moyer et al, 2000).
    c) CASE REPORT: A 19-year-old man developed serotonin syndrome (altered mental status, nystagmus, muscle rigidity, hyperreflexia, hyperthermia, and lower-extremity clonus) following intentional ingestion of an unknown amount of citalopram. His serum citalopram concentration, obtained at time of presentation, was 1500 ng/mL (therapeutic, 9 to 200 ng/mL). The serotonin syndrome resolved with symptomatic and supportive therapy, and the patient was discharged, without sequelae, to a psychiatric facility approximately 10 days post-ingestion (Maddry et al, 2013).
    C) DROWSY
    1) WITH THERAPEUTIC USE
    a) Somnolence has been reported after therapeutic use of citalopram (Grundemar et al, 1997; Personne et al, 1997; Nyth et al, 1992; Milne & Goa, 1991; de Wilde et al, 1985) .
    2) WITH POISONING/EXPOSURE
    a) Somnolence has been reported in overdose and after therapeutic use (Personne et al, 1997; Grundemar et al, 1997; Nyth et al, 1992; Milne & Goa, 1991; de Wilde et al, 1985).
    b) CASE REPORT: A 44-year-old man developed somnolence along with prolonged QTc following an overdose of citalopram 3640 mg (Grundemar et al, 1997).
    c) CASE SERIES: At doses below 600 mg in 41 patients, mild symptoms of drowsiness and somnolence were observed (Personne et al, 1997).
    d) CASE SERIES: In a retrospective review of reported citalopram overdoses, drowsiness or a decrease in consciousness occurred in 15 of 26 patients. Of those 15 patients, 4 patients (3 patients with drowsiness and 1 comatose patient) ingested citalopram as the sole drug. Among the 26 patients, the estimated citalopram amounts ingested ranged from 200 to 4960 mg (median 1190 mg) (Jimmink et al, 2008).
    e) ESCITALOPRAM: A 32-year-old woman developed nausea, vertigo, confusion, palpitation, drowsiness, slight leukocytosis, mild fever, tachycardia, and mild hypotension after ingesting escitalopram 190 mg. Following supportive care, she recovered and was discharged 24 hours after hospitalization (Yuksel et al, 2005).
    D) CENTRAL NERVOUS SYSTEM FINDING
    1) WITH THERAPEUTIC USE
    a) Headache (18% of patients), insomnia (15%), tremor (16%), dizziness (14%), restlessness (10%), and sedation (15%) have been commonly reported during clinical studies with citalopram (Milne & Goa, 1991; Nyth et al, 1992; Bouchard et al, 1987; Gravem et al, 1987; de Wilde et al, 1985).
    2) WITH POISONING/EXPOSURE
    a) In a case series of citalopram overdoses in 108 patients, mild symptoms of dizziness and tremor were observed at doses below 600 mg (Personne et al, 1997).
    b) ESCITALOPRAM: A 32-year-old woman developed nausea, vertigo, confusion, palpitation, drowsiness, slight leukocytosis, mild fever, tachycardia, and mild hypotension after ingesting escitalopram 190 mg. Following supportive care, she recovered and was discharged 24 hours after hospitalization (Yuksel et al, 2005).
    c) CASE REPORT: A 36-year-old woman developed palpitation, weakness, nausea, and numbness 36 hours after taking citalopram 1000 mg with alcohol (Tarabar et al, 2003).
    d) CASE REPORT: Lightheadedness and tremors occurred in a 14-year-old girl after she ingested escitalopram 200 mg (Scharko & Schumacher, 2008).
    E) NEUROLEPTIC MALIGNANT SYNDROME
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 20-year-old man became comatose after ingesting citalopram 1900 mg. He presented to the hospital with fever and mild rigidity of the limbs, neck, and abdomen. His pulse was 90 beats/min, and he was hypotensive (90/60 mmHg). Two days postingestion, the patient developed a pneumothorax; a chest drainage tube was inserted, and he was mechanically ventilated. After 6 days, the patient was extubated; he was still unconscious, and his muscle rigidity increased. On day 7, his serum creatine kinase peaked at 1258 units/L, and neuroleptic malignant syndrome was suspected. The patient gradually recovered following bromocriptine therapy (Ayudin et al, 2000).
    F) SYNCOPE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 32-year-old woman developed severe sinus bradycardia, hypotension, and episodes of syncope approximately 4 hours after ingesting citalopram 800 mg in a suicide attempt. ECG showed normal QRS morphology and QT interval values. The bradycardia, hypotension, and syncopal episodes resolved following insertion of a temporary pacemaker (Rothenhausler et al, 2000).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) NAUSEA AND VOMITING
    1) WITH THERAPEUTIC USE
    a) Nausea has been reported with therapeutic use (Personne et al, 1997).(Milne & Goa, 1991).
    b) INCIDENCE: In a meta-analysis of citalopram therapy, nausea (20% of patients), vomiting (20%), xerostomia (17%), and constipation (13%) were the most frequently reported gastrointestinal effects (Milne & Goa, 1991).
    2) WITH POISONING/EXPOSURE
    a) Nausea and vomiting have been reported following overdose (Tarabar et al, 2003; Personne et al, 1997; Milne & Goa, 1991).
    b) CASE SERIES: In a retrospective review of reported citalopram overdoses, nausea and vomiting occurred in 6 of 26 patients. Among the 26 patients, the estimated amounts ingested ranged from 200 to 4960 mg (median 1190 mg) (Jimmink et al, 2008).
    c) ESCITALOPRAM: A 32-year-old woman developed nausea, vertigo, confusion, palpitation, drowsiness, slight leukocytosis, mild fever, tachycardia, and mild hypotension after ingesting escitalopram 190 mg. Following supportive care, she recovered and was discharged 24 hours after hospitalization (Yuksel et al, 2005).
    B) LOSS OF APPETITE
    1) WITH THERAPEUTIC USE
    a) Anorexia and dyspepsia have been reported less frequently following citalopram therapy (Milne & Goa, 1991; Bouchard et al, 1987).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) ABNORMAL LIVER FUNCTION
    1) WITH THERAPEUTIC USE
    a) Several patients have developed abnormal liver function tests after receiving citalopram during therapeutic use (de Wilde et al, 1985; Pedersen et al, 1982).
    b) LACK OF EFFECT: In a meta-analysis of clinical studies with citalopram, no significant changes in liver enzymes were reported (Milne & Goa, 1991).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) DYSURIA
    1) WITH THERAPEUTIC USE
    a) Urination difficulties have been reported infrequently in clinical studies (Burrows et al, 1988; Pedersen et al, 1982).
    B) RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 45-year-old man developed oliguric renal failure approximately 10 hours after ingesting citalopram hydrobromide 3000 mg (citalopram 2400 mg as a racemic mixture). His creatinine peaked at 492 mcmol/L 4 days postingestion. The patient subsequently recovered following spontaneous improvement of his renal function (Kelly et al, 2003).
    C) PRIAPISM
    1) WITH POISONING/EXPOSURE
    a) Priapism occurred in a 58-year-old man several hours after he ingested 3 citalopram 20 mg tablets in addition to his usual dosage of 20 mg twice daily for treatment of depression. The patient was also taking tamsulosin 0.4 mg daily for treatment of benign prostatic hyperplasia. The priapism gradually resolved following administration of phenylephrine and surgical intervention. The patient recovered fully without any evidence of erectile dysfunction. The patient had a history of trazodone-induced priapism. The authors concluded that patients who have a history of priapism with other drugs may be more susceptible to citalopram-associated priapism (Dent et al, 2002).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) Metabolic acidosis has been reported following 2 cases of nonfatal citalopram overdose (range 1680 to 5200 mg). Both patients developed seizures and hypotension (Grundemar et al, 1997).
    b) CASE REPORT: Metabolic acidosis (pH 7.15) occurred in a 54-year-old woman with psychotic depression treated with citalopram 20 mg/day and zopiclone 7.5 mg/day after she intentionally ingested an unknown amount of citalopram with alcohol. She presented to the emergency department (time of ingestion unknown) with altered mental status, low O2 saturation, severe hypotension, and a heart rate within normal limits. Laboratory analyses showed lactic acidosis (lactate 10 mmol/L) and hypokalemia. After treatment with activated charcoal, she was admitted to ICU where a prolonged QT interval of 0.44 seconds (QTc 0.62 seconds) with left bundle branch block was noted on ECG after an episode of ventricular fibrillation (VF). Sustained ventricular tachycardia with VF episodes occurred 10 hours after ICU admission and continued for 48 hours, requiring repeated defibrillation. She recovered fully and was discharged 16 days after admission (Liotier & Coudore, 2011).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) LEUKOCYTOSIS
    1) WITH POISONING/EXPOSURE
    a) ESCITALOPRAM: A 32-year-old woman developed nausea, vertigo, confusion, palpitation, drowsiness, slight leukocytosis (12.5 K/ul), mild fever, tachycardia, and mild hypotension after ingesting escitalopram 190 mg. Following supportive care, she recovered and was discharged 24 hours after hospitalization (Yuksel et al, 2005).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) EXCESSIVE SWEATING
    1) WITH THERAPEUTIC USE
    a) Diaphoresis has been commonly reported following citalopram therapy (Milne & Goa, 1991; Bouchard et al, 1987; Gravem et al, 1987; de Wilde et al, 1985).
    b) INCIDENCE: Approximately 18% of patients reported diaphoresis, as described in a meta-analysis of adverse effects experienced with citalopram (Milne & Goa, 1991).
    B) ERUPTION
    1) WITH THERAPEUTIC USE
    a) Rash has been reported infrequently following therapeutic citalopram use (Bouchard et al, 1987; Gravem et al, 1987).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) Rhabdomyolysis developed in 2 patients as a result of nonfatal overdose (range 3640 to 5200 mg). The authors provided no further data; 1 patient did experience seizures following overdose (Grundemar et al, 1997).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPOGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) Severe hypoglycemia (blood glucose concentrations ranging from 1.7 to 3.8 mmol/L) was reported in 2 patients following overdose ingestions of citalopram in combination with alcohol ingestion. Both patients recovered following treatment with IV dextrose (Duncan et al, 2008).

Reproductive

    3.20.1) SUMMARY
    A) Citalopram and escitalopram are classified as FDA pregnancy category C. Limited data on the use of citalopram during human pregnancy have not demonstrated apparent teratogenic risk; however, SSRI administration lasting more than 30 days during the second or third lunar month of pregnancy was associated with a significant 80% increased risk of clubfoot occurrence in infants. In addition, third trimester exposure has been associated with an increased risk for nonteratogenic effects (pulmonary hypertension of the newborn (PPHN) and findings consistent with serotonin syndrome) and neonatal intensive care. Animal studies demonstrated fetal abnormalities, reduced birth weights, death, and mild maternal toxicity with citalopram and escitalopram use during pregnancy; however, these occurred at doses considerably greater than the maximum recommended human dose. The manufacturer suggests considering a dose reduction of citalopram or escitalopram during the third trimester of pregnancy. Citalopram and escitalopram are excreted in human breast milk and long-term effects of exposure to SSRIs via breast milk on the cognitive development of the infant have not been determined. In fertility studies of citalopram use in male and female rats, reduced fertility and increased duration of pregnancy were observed at doses 5 and 8 times higher than the recommended human dose, respectively. An increased risk for social-behavioral abnormalities at 2 to 6 years of age was reported in children exposed to SSRIs or SNRIs in utero who developed neonatal abstinence syndrome (NAS) at birth.
    3.20.2) TERATOGENICITY
    A) CONGENITAL MALFORMATIONS
    1) Data from the case-controlled National Birth Defects Prevention Study (NBDPS), which included data from 13,714 infants born between 1997 and 2002, indicated that early maternal exposure (defined as treatment with any SSRI from 1 month before to 3 months after conception) to SSRIs was associated with anencephaly in 9 exposed infants out of 214 (adjusted odds ratio (OR), 2.4; 95% confidence interval (CI), 1.1 to 5.1; p=0.02), craniosynostosis in 24 exposed infants out of 432 (adjusted OR, 2.5; 95% CI, 1.5 to 4; p less than 0.001), and omphalocele in 11 exposed infants out of 181 (adjusted OR, 2.8; 95% CI, 1.3 to 5.7; p=0.005). However, early exposure did not significantly increase the risks of congenital heart defects or most other birth defects. The most commonly used SSRIs reported by control mothers were sertraline, fluoxetine, paroxetine, and citalopram (Alwan et al, 2007).
    2) SSRI administration lasting more than 30 days during the second or third lunar month of pregnancy was associated with a significant 80% increased risk of clubfoot occurrence in infants. Escitalopram administration had a significant 190% increased risk, paroxetine administration had a non-significant 820% increased risk, sertraline administration had a non-significant 60% increased risk, fluoxetine administration had a non-significant 30% increased risk, and citalopram administration had a non-significant 10% decreased risk. Because of small numbers of subjects exposed to these SSRIs, the estimated odds ratios were unstable for these agents, especially for paroxetine (Yazdy et al, 2014).
    B) ANIMAL STUDIES
    1) Fetal structural abnormalities was reported in the offspring of rats and rabbits treated with either oral escitalopram or racemic citalopram during pregnancy at doses considerably greater than the maximum recommended human dose (Prod Info CELEXA(R) oral tablets, solution, 2008; Prod Info LEXAPRO(R) oral tablets, solution, 2008).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) The manufacturer has classified citalopram and escitalopram as FDA pregnancy category C (Prod Info CELEXA(R) oral tablets, solution, 2008; Prod Info LEXAPRO(R) oral tablets, solution, 2008).
    2) In a prospective longitudinal study of 201 women with a history of major depression and no signs of depression at the beginning of pregnancy, there was a greater likelihood of relapse of major depression in those who discontinued antidepressant drugs during pregnancy compared with those who continued antidepressant drugs throughout the pregnancy (Prod Info CELEXA(R) oral tablets, solution, 2008; Prod Info LEXAPRO(R) oral tablets, solution, 2008).
    B) SPONTANEOUS ABORTION
    1) A nested case-controlled study showed that citalopram, sertraline, fluoxetine, fluvoxamine, or combined use of 2 or more SSRIs during pregnancy did not correspond with a significantly increased risk of spontaneous abortion. However, paroxetine or venlafaxine use alone did increase the spontaneous abortion risk. Data collected from the Quebec Pregnancy Registry between January 1998 and December 2003 on women who filled at least 1 antidepressant prescription during pregnancy and had a clinically detected spontaneous abortion by the twentieth week of gestation (n=284) showed an increased risk of spontaneous abortion (adjusted odds ratio (OR), 1.68; 95% confidence interval (CI), 1.38 to 2.06) when compared with randomly selected registry controls (4 matched controls per case) without antidepressant use. Tracked antidepressant categories included SSRIs, tricyclic antidepressants, serotonin-norepinephrine reuptake inhibitors, combined use of 2 or more antidepressant classes, or others. Paroxetine use (adjusted OR 1.75; 95% CI, 1.31 to 2.34) or venlafaxine use (adjusted OR 2.11; 95% CI, 1.34 to 3.3) alone were independently associated with a higher risk of spontaneous abortion. The highest daily doses of paroxetine or venlafaxine during pregnancy were associated with the greatest spontaneous abortion risk; of the women taking paroxetine (n=84) or venlafaxine (n=33) who spontaneously aborted, an adjusted analysis showed 25.5% averaged daily doses of more than 25 mg of paroxetine and 50% averaged daily doses greater than 150 mg of venlafaxine (Nakhai-Pour et al, 2010).
    C) INFANT PLASMA CITALOPRAM
    1) One prospective study of 11 mothers taking citalopram during pregnancy and 10 matched pregnant women not on the medication found that the trough plasma citalopram and its desmethyl metabolite concentrations in the infants were 64% and 66% of the maternal concentrations, respectively. The delivery outcomes and neurodevelopmental progress of the infants at one year of age were normal (Heikkinen et al, 2002).
    D) CNS EFFECTS
    1) Infants exposed to citalopram, escitalopram, and other SSRIs or serotonin-norepinephrine reuptake inhibitors (SNRIs) late in the third trimester have developed neonatal complications. Epidemiologic studies showed an increased risk of persistent pulmonary hypertension of the newborn (PPHN) with prenatal exposure to citalopram, escitalopram, and other SSRIs during pregnancy, a condition associated with considerable neonatal morbidity and mortality. Prenatal exposure late in the third trimester has been linked to complications that required intensive care (eg, prolonged hospitalization, tube feeding, respiratory support), sometimes immediately after delivery. Symptoms have been consistent with either a direct toxic effect of the agent or a possible drug discontinuation syndrome (eg, constant crying; irritability; jitteriness or tremor; hyperreflexia; hypertonia or hypotonia; hypoglycemia; vomiting; feeding difficulties; temperature instability; seizures; or respiratory distress, cyanosis, or apnea). In some cases, clinical findings were consistent with serotonin syndrome (Prod Info Celexa(R) oral tablets, oral solution, 2012; Prod Info Lexapro(R) oral tablets, oral solution, 2012).
    2) In a prospective, single-blind, cohort study, full-term infants who developed neonatal abstinence syndrome (NAS) at birth had similar cognitive abilities compared with full term infants without NAS at birth when reevaluated at 2 to 6 years of age. However, infants with NAS at birth were at an increased risk for social-behavioral abnormalities at 2 to 6 years of age. The study was designed to assess the long-term neurodevelopment of children exposed in utero to fluoxetine, paroxetine, citalopram, sertraline, fluvoxamine, or venlafaxine. Children with NAS at birth (n=30; Finnegan score of 4 or greater) were compared to children without NAS (n=52; Finnegan score 0 to 3); both groups were similar in mean cognitive ability (106.9 +/- 14 versus 100.5 +/- 14.6, respectively; p=0.12) and developmental scores (98.9 +/- 11.4 versus 95.7 +/- 9.9, respectively; p=0.21). Cognitive ability was based on scores from the Wechsler Preschool and Primary Scale of Intelligence II, the Stanford-Binet Intelligence Scales, or the Bayley Scale of Infant Development II. The NAS infants had an increased risk of social-behavior abnormalities (odds ratio (OR) 3.03, 95% CI, 1.07 to 8.6, p=0.04) based on the Denver Developmental Screening Test II (DDST-II) and NAS after birth was associated with advanced maternal age (OR 1.12, 95% CI, 1 to 1.25, p=0.04). In addition, there was a trend towards small head circumference in the NAS group when compared with the children without NAS (n=6 (20%) versus n=3 (6%), respectively; p=0.068) (Klinger et al, 2011).
    3) An increased risk for CNS serotonergic symptoms was observed in 4-day-old infants whose mothers were taking SSRIs during the third trimester of pregnancy. In a controlled, prospective study, pregnant women received 20 to 40 mg/day of either citalopram (n=10), fluoxetine (n=10), or did not receive an SSRI (n=20). Exposure to SSRI therapy ranged from 7 to 41 weeks. Newborns in the SSRI group had lower Apgar scores at 15 minutes compared with the control group (8.8 vs 9.4; p=0.02). At 2 weeks, a higher heart rate was observed in the SSRI group compared with the control group (mean, 153 vs 141 beats per minute; p=0.049). Serotonergic symptom scores in the first 4 days after birth were significantly higher in the SSRI group compared with the control group (121 vs 30; p=0.008). Tremor, restlessness, and rigidity were the most prominent symptoms. Myoclonus was reported in one infant exposed to fluoxetine. Significantly lower cord blood 5-hydroxyindoleacetic acid (5-HIAA) concentrations were seen in the SSRI group compared with the control group (mean, 63 mmol/L vs 77 mmol/L; p=0.02). Additionally, a significant inverse correlation was observed between the serotonergic symptom score and the umbilical vein 5-HIAA concentrations in the SSRI group compared with the control group (p=0.007). Although not statistically significant, mean umbilical cord serum prolactin concentrations were 29% lower in SSRI-exposed infants than in control infants at the time of birth (Laine et al, 2003).
    E) AUTISM SPECTRUM DISORDER
    1) A cohort study of prospectively collected data demonstrated an increased risk of autism spectrum disorder (ASD) in children whose mothers used antidepressants during the second or third trimesters of pregnancy; the risk was even greater with second or third trimester exposure to SSRIs. Thirty-one infants who were exposed to antidepressants during the second or third trimester were diagnosed with ASD. After adjusting for potential confounders, second or third trimester exposure to antidepressants was associated with a significant 87% increased risk of ASD, while first trimester exposure or use of antidepressants in the year before pregnancy was not associated with any such risk. Use of SSRIs during the second or third trimester was associated with a significant more than 2-fold increased risk of ASD (22 exposed infants), while other classes of antidepressants were not associated with an increased risk. Even after restricting the sample size to those children whose mothers had a history of depression and used antidepressants during the second or third trimester, the risk of ASD still persisted. In addition, use of more than 1 class of antidepressants during the second or third trimester was associated with a significant more than 4-fold increased risk of ASD (Boukhris et al, 2016).
    F) PULMONARY HYPERTENSION
    1) Infants exposed to citalopram, escitalopram, and other SSRIs or serotonin-norepinephrine reuptake inhibitors (SNRIs) late in the third trimester have developed neonatal complications. Epidemiologic studies showed an increased risk of persistent pulmonary hypertension of the newborn (PPHN) with prenatal exposure to citalopram, escitalopram, and other SSRIs during pregnancy, a condition associated with considerable neonatal morbidity and mortality (Prod Info Celexa(R) oral tablets, oral solution, 2012; Prod Info Lexapro(R) oral tablets, oral solution, 2012).
    2) In a case control study of women who delivered infants with pulmonary hypertension of the newborn (PPHN; n=377) and women who delivered healthy infants (n=836), the risk for developing PPHN was approximately 6-fold higher in infants exposed to SSRIs after week 20 of gestation compared with infants not exposed to SSRIs during gestation. This study demonstrates a potential increased risk of PPHN, associated with considerable neonatal morbidity and mortality, in infants exposed to SSRIs later in the pregnancy. Because this is the first study regarding PPHN with SSRI use during pregnancy and there are not enough cases with exposure to individual SSRIs, it cannot be determined if all SSRIs posed similar levels of PPHN risk. In the general population, PPHN occurs in 1 to 2 per 1000 live births (Prod Info CELEXA(R) oral tablets, solution, 2008; Prod Info LEXAPRO(R) oral tablets, solution, 2008).(Chambers et al, 2006).
    G) NEONATAL INTENSIVE CARE
    1) Infants exposed to citalopram, escitalopram, and other SSRIs or serotonin-norepinephrine reuptake inhibitors (SNRIs) late in the third trimester have developed neonatal complications. Epidemiologic studies showed an increased risk of persistent pulmonary hypertension of the newborn (PPHN) with prenatal exposure to citalopram, escitalopram, and other SSRIs during pregnancy, a condition associated with considerable neonatal morbidity and mortality. Prenatal exposure late in the third trimester has been linked to complications that required intensive care (eg, prolonged hospitalization, tube feeding, respiratory support), sometimes immediately after delivery. Symptoms have been consistent with either a direct toxic effect of the agent or a possible drug discontinuation syndrome (eg, constant crying; irritability; jitteriness or tremor; hyperreflexia; hypertonia or hypotonia; hypoglycemia; vomiting; feeding difficulties; temperature instability; seizures; or respiratory distress, cyanosis, or apnea). In some cases, clinical findings were consistent with serotonin syndrome (Prod Info Celexa(R) oral tablets, oral solution, 2012; Prod Info Lexapro(R) oral tablets, oral solution, 2012).
    2) A prospective comparative study of citalopram exposure during pregnancy demonstrated no association with apparent major teratogenicity; however, exposure late in pregnancy was associated with an increased risk for neonatal intensive care. Study participants (n=396) were selected from a group of women who had contacted a Canadian teratogen information center with concerns regarding citalopram and other drugs during pregnancy. There were 132 women in each of 3 groups that were matched for maternal age at time of conception and gestational stage of pregnancy: exposed group (women who took citalopram during pregnancy), a disease-matched group (pregnant women with similar psychiatric issues but treated with other SSRIs), and a non-teratogen group. Of the exposed group, 125 took citalopram at least in the first trimester and 71 continued the drug throughout pregnancy. There were 114 live births, 14 spontaneous abortions, 2 elective terminations, and 2 stillbirths in this group. Citalopram exposure late in pregnancy was associated with a relative risk of 4.2 (95% confidence interval, 1.71 to 10.26) compared with non-exposure. Complications included pneumothorax, fetal distress, decreased heart rate, heart rate variability, and meconium staining, and aspiration. The 3 groups did not show statistically significant differences in fetal survival rates, mean birth weights or duration of pregnancy or the rate of perinatal complications in the third trimester. Only one case of major malformation, umbilical and scrotal hernia requiring surgery, occurred among infants exposed to citalopram in the first trimester (Sivojelezova et al, 2005).
    3) A population-based study of 1782 pregnant women exposed to SSRIs demonstrated no increased risk of adverse perinatal outcome; however, a higher incidence of neonatal intensive or special care unit was noted, particularly with third trimester exposure. Using 1996 to 2001 data derived from a government project involving 4 birth or medication registries in Finland, women who had at least one purchase (a 3-month supply) of an SSRI during the period of 1 month before pregnancy and the day pregnancy ended were compared with 1782 controls with no reimbursed drug purchases during the same peripartum period. The mean age of both cohorts was 30 years (range, 23 to 37 years). There were more than twice as many smokers and six times as many pregnancies induced by artificial reproductive techniques in the SSRI group compared with controls (p less than 0.001). Mean length of gestation and birth weight were lower (p less than 0.001) in the SSRI group. Malformations were not more common in the SSRI group (p = 0.4). Purchases of SSRIs (citalopram, fluoxetine, paroxetine, sertraline and fluvoxamine) were more common in the first trimester than later in pregnancy, with 554 women purchasing citalopram during the first trimester, 219 during the second trimester, 228 during the third, and 94 throughout pregnancy. Compared with first trimester exposure, special or intensive care unit visits were more common for the infants exposed during the third trimester (11.2% vs 15.7%; p=0.009). This difference remained statistically significant even after adjusting for confounding variables (OR 1.6; 95% confidence interval (CI), 1.1 to 2.2) (Malm et al, 2005).
    H) QT PROLONGATION
    1) A study of prospectively collected data suggests antenatal use of selective serotonin-reuptake inhibitor (SSRI) antidepressants is associated with QTc interval prolongation in exposed neonates. Between January 2000 and December 2005, researchers compared 52 neonates exposed to SSRI antidepressants (paroxetine (n=25), citalopram (n=13), fluoxetine (n=12), fluvoxamine (n=1), and venlafaxine (n=1)) in the immediate antenatal period to 52 matched neonates with no exposure. Prolonged QTc is defined as an interval of greater than 460 milliseconds (msec) (the widely used upper limit cited by authorities in both pediatric cardiology and neonatology). A pediatric cardiologist blinded to drug exposure interpreted all electrocardiograms (ECGs) using standard statistical analyses. ECG recordings revealed markedly prolonged mean QTc intervals in exposed neonates compared to unexposed neonates (mean; 409 +/- 42 msec vs. 392 +/- 29 msec, p=0.02). The mean uncorrected QT interval was 7.5% longer among exposed neonates (mean; 280 +/- 31 msec vs. 261 +/- 25 msec, p less than 0.001). Ten percent (n=5) of exposed neonates had a notable increase in QTc interval prolongation (greater than 460 msec) compared to none of the unexposed neonates. The longest QTc interval observed was 543 msec (Dubnov-Raz et al, 2008).
    I) ANIMAL STUDIES
    1) RATS, RABBITS: Reduced fetal body weight, growth retardation, and death were reported in the offspring of rats and rabbits treated with either oral escitalopram or racemic citalopram during pregnancy at doses considerably greater than the maximum recommended human dose. Mild maternal toxicity was also reported in rat studies of escitalopram use during pregnancy (Prod Info CELEXA(R) oral tablets, solution, 2008; Prod Info LEXAPRO(R) oral tablets, solution, 2008).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) CITALOPRAM
    a) In one study, serum levels were either undetectable (n=4) or low (n=6) in breast-fed infants of citalopram-treated mothers. Serum drug level was still low in an infant of a citalopram-treated mother with CYP2C19 poor metabolizer status (Berle et al, 2004).
    b) One study reported a citalopram milk/serum ratio of 1.16 to 1.88 (Spigset et al, 1997); a higher milk/serum ratio of 3 was reported in another study. The first study estimated that the infant would ingest 4.3 mcg/kg/day with a relative citalopram dose of 0.7% to 5.9% of the weight-adjusted maternal dose.(Jensen et al, 1997). There have been 2 case reports of excessive somnolence, weight loss, and decreased feeding in nursing infants whose mothers were taking citalopram. One infant reportedly recovered completely upon maternal citalopram discontinuation; follow-up information was not available for the other infant (Prod Info CELEXA(R) oral tablets, solution, 2008; Prod Info LEXAPRO(R) oral tablets, solution, 2008; Anon, 1998).
    c) A newborn infant, who was exposed prenatally to citalopram 40 mg daily throughout the pregnancy and during lactation, developed irregular breathing, sleep disorders, and hypotonia alternating with hypertonia within 3 weeks of birth. Maternal serum citalopram concentration ranged from 348 to 493 nM (therapeutic range 95 to 620 nM), coinciding with citalopram concentrations in the breast milk ranging from 709 to 986 nM. Infant serum concentration ranged from 3.7 to 7.1 nM. Based on the daily intake of breast milk by the infant and the weight of the infant and mother, the infant's citalopram dose was approximately 9% of the maternal dose. Therefore, it is believed that the infant's symptoms were due to citalopram withdrawal effects and not effects induced by breastfeeding (Franssen et al, 2006).
    2) ESCITALOPRAM
    a) In a study describing 8 lactating women treated with an escitalopram median single daily dose of 10 mg (range, 10 to 20 mg) that began 55 days before the study date, the infant plasma concentrations of escitalopram and its active metabolite, demethylescitalopram, were undetectable (n=4), low (n=1), or not measured (n=3). The total relative infant dose for escitalopram and its metabolite was a mean of 5.3% (95% confidence interval (CI), 4.2% to 6.2%) of the maternal weight-adjusted dose and the absolute doses were 7.6 mcg/kg/day (95% CI, 5.2 to 10) and 3 mcg/kg/day (95% CI, 2.4 to 3.6) for escitalopram and demethylescitalopram, respectively. The mean milk/plasma ratio was 2.2 for both escitalopram (95% confidence interval (CI), 2 to 2.4) and demethylescitalopram (95% CI, 1.9 to 2.5). The authors suggest that escitalopram is safe for use in nursing mothers; however, individual cases should be decided based on a risk/benefit analysis (Rampono et al, 2006).
    3.20.5) FERTILITY
    A) ANIMAL STUDIES
    1) Studies in male and female rats given oral racemic citalopram doses ranging from 32 to 72 mg/kg/day demonstrated reduced fertility at doses greater than or equal to 32 mg/kg/day (approximately 5 times the maximum recommended human dose (MRHD)), increased gestation duration at 48 mg/kg/day (approximately 8 times the MRHD) and decreased mating at all doses (Prod Info CELEXA(R) oral tablets, solution, 2008; Prod Info LEXAPRO(R) oral tablets, solution, 2008).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor vital signs (including temperature) and mental status.
    B) Monitor serum electrolytes (including potassium, bicarbonate) following significant overdose.
    C) Obtain baseline ECG, continuous cardiac monitoring, and serial ECGs following a significant exposure.
    D) Monitor serial blood glucose, especially in patients with depressed mental status; severe hypoglycemia has been reported after citalopram overdose.
    E) Plasma concentration is not readily available and does not correlate well with therapeutic or adverse effects. It is not indicated for the acute management of overdose.
    4.1.2) SERUM/BLOOD
    A) Monitor serum electrolytes (including potassium, bicarbonate) following significant overdose.
    B) Monitor serial blood glucose, especially in patients with depressed mental status; severe hypoglycemia has been reported after citalopram overdose (Duncan et al, 2008).
    4.1.4) OTHER
    A) OTHER
    1) ECG
    a) Obtain baseline ECG, institute continuous cardiac monitoring, and repeat serial ECGs following a significant exposure, to evaluate for QTc prolongation and dysrhythmia.

Methods

    A) CHROMATOGRAPHY
    1) High-performance liquid chromatographic determination has been used for citalopram and its metabolites in plasma and urine samples (Oyehaug & Ostensen, 1984).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients with mild to moderate toxicity after intentional overdose need to be monitored for prolonged QTc for at least 13 hours. Patients with severe toxicity need to be admitted to an intensive care unit. For isolated escitalopram ingestions, patients with significant clinical effects including seizures or persistent lethargy, hypotension, tachycardia, QTc prolongation, or dysrhythmia need to be admitted (Forrester, 2007).
    6.3.1.2) HOME CRITERIA/ORAL
    A) Children and adults with mild symptoms (eg, vomiting, mydriasis, diaphoresis, mild somnolence) following an inadvertent ingestion of up to 100 mg citalopram or 50 mg escitalopram can be managed at home with instructions to call the poison center back, if symptoms develop. For patients already on citalopram or escitalopram, those with inadvertent ingestions of up to 5 times their own single therapeutic dose can be observed at home with instructions to call the poison center back, if symptoms develop (Nelson et al, 2007).
    B) A case review, involving inadvertent pediatric ingestions of citalopram from 2000 to 2014, identified 335 patients (6 years of age or less) who ingested citalopram as the sole drug. Estimated doses were reported in 119 cases, with unknown weights estimated at the 50th percentile weight for age. Doses ranged from 0.2 to 9.57 mg/kg and 1 40-mg/kg dose. Minor effects (ie, hyperactivity, drowsy, vomiting, abdominal pain) occurred at doses of 0.41 to 2.5 mg/kg, moderate effects of drowsiness and fever occurred at 1.67 mg/kg, and a seizure was reported with a 5 mg/kg dose. Based on these results, it is suggested that pediatric ingestions of less than 5 mg/kg will not cause serious toxicity and may be safely managed outside of a healthcare facility (Herrington et al, 2015).
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity or an unclear diagnosis.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Patients with deliberate ingestions, those with more than mild symptoms, and ingestions of more than 100 mg citalopram or 50 mg escitalopram should be sent to a health care facility. Patients on chronic citalopram or escitalopram therapy should be sent to a healthcare facility if they ingest more than 5 time their own single therapeutic dose. Ingestion of more than 600 mg citalopram or more than 300 mg escitalopram requires cardiac monitoring for 8 hours (11 hours if the patient did not receive activated charcoal within 4 hours of ingestion). Ingestion of more than 1000 mg citalopram or more than 500 mg escitalopram requires cardiac monitoring for 13 hours. At the end of the observation period, if the QTc is less than 450 msec, the monitoring can be discontinued, and the patient may be discharged if asymptomatic. Patients with symptoms of toxicity or a QTc of greater than 450 msec at the end of the observation period should be admitted for continued cardiac monitoring (Friberg et al, 2006; Isbister et al, 2006).

Monitoring

    A) Monitor vital signs (including temperature) and mental status.
    B) Monitor serum electrolytes (including potassium, bicarbonate) following significant overdose.
    C) Obtain baseline ECG, continuous cardiac monitoring, and serial ECGs following a significant exposure.
    D) Monitor serial blood glucose, especially in patients with depressed mental status; severe hypoglycemia has been reported after citalopram overdose.
    E) Plasma concentration is not readily available and does not correlate well with therapeutic or adverse effects. It is not indicated for the acute management of overdose.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) SUMMARY
    1) Do not induce emesis. Activated charcoal can be considered within the first hour after large ingestion (greater than 600 mg citalopram), if the patient has an appropriate level of consciousness, has a patent airway, and is able to drink the charcoal.
    B) ACTIVATED CHARCOAL
    1) PREHOSPITAL ACTIVATED CHARCOAL ADMINISTRATION
    a) Consider prehospital administration of activated charcoal as an aqueous slurry in patients with a potentially toxic ingestion who are awake and able to protect their airway. Activated charcoal is most effective when administered within one hour of ingestion. Administration in the prehospital setting has the potential to significantly decrease the time from toxin ingestion to activated charcoal administration, although it has not been shown to affect outcome (Alaspaa et al, 2005; Thakore & Murphy, 2002; Spiller & Rogers, 2002).
    1) In patients who are at risk for the abrupt onset of seizures or mental status depression, activated charcoal should not be administered in the prehospital setting, due to the risk of aspiration in the event of spontaneous emesis.
    2) The addition of flavoring agents (cola drinks, chocolate milk, cherry syrup) to activated charcoal improves the palatability for children and may facilitate successful administration (Guenther Skokan et al, 2001; Dagnone et al, 2002).
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    6.5.2) PREVENTION OF ABSORPTION
    A) ACTIVATED CHARCOAL
    1) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    3) A retrospective study was conducted to determine the relationship between decontamination with activated charcoal and the risk of developing QT interval prolongation following citalopram overdose ingestions. Of the 254 citalopram overdose admissions, single-dose activated charcoal (SDAC) was administered, at an average of 2.1 hours postingestion (ranging from 0.5 to 6.25 hours postingestion), in 48 cases. Of those cases, QT interval prolongation was observed in only 2 instances (4.2%). In comparison, of the 206 cases of patients who did not receive activated charcoal, QT interval prolongation was observed in 23 instances (11.2%). The relative risk of developing an abnormal QTc, RR combination following decontamination with SDAC, as compared to no SDAC, was 0.28 (95% CI, 0.06 to 0.7), with an estimated relative risk reduction of 72%. Based on the results of this study, the authors conclude that SDAC administration is effective in decreasing the risk of developing a prolonged QT interval following an overdose ingestion of citalopram (Isbister et al, 2007).
    B) GASTRIC LAVAGE
    1) INDICATIONS: Consider gastric lavage with a large-bore orogastric tube (ADULT: 36 to 40 French or 30 English gauge tube {external diameter 12 to 13.3 mm}; CHILD: 24 to 28 French {diameter 7.8 to 9.3 mm}) after a potentially life threatening ingestion if it can be performed soon after ingestion (generally within 60 minutes).
    a) Consider lavage more than 60 minutes after ingestion of sustained-release formulations and substances known to form bezoars or concretions.
    2) PRECAUTIONS:
    a) SEIZURE CONTROL: Is mandatory prior to gastric lavage.
    b) AIRWAY PROTECTION: Place patients in the head down left lateral decubitus position, with suction available. Patients with depressed mental status should be intubated with a cuffed endotracheal tube prior to lavage.
    3) LAVAGE FLUID:
    a) Use small aliquots of liquid. Lavage with 200 to 300 milliliters warm tap water (preferably 38 degrees Celsius) or saline per wash (in older children or adults) and 10 milliliters/kilogram body weight of normal saline in young children(Vale et al, 2004) and repeat until lavage return is clear.
    b) The volume of lavage return should approximate amount of fluid given to avoid fluid-electrolyte imbalance.
    c) CAUTION: Water should be avoided in young children because of the risk of electrolyte imbalance and water intoxication. Warm fluids avoid the risk of hypothermia in very young children and the elderly.
    4) COMPLICATIONS:
    a) Complications of gastric lavage have included: aspiration pneumonia, hypoxia, hypercapnia, mechanical injury to the throat, esophagus, or stomach, fluid and electrolyte imbalance (Vale, 1997). Combative patients may be at greater risk for complications (Caravati et al, 2001).
    b) Gastric lavage can cause significant morbidity; it should NOT be performed routinely in all poisoned patients (Vale, 1997).
    5) CONTRAINDICATIONS:
    a) Loss of airway protective reflexes or decreased level of consciousness if patient is not intubated, following ingestion of corrosive substances, hydrocarbons (high aspiration potential), patients at risk of hemorrhage or gastrointestinal perforation, or trivial or non-toxic ingestion.
    6.5.3) TREATMENT
    A) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    B) MONITORING OF PATIENT
    1) QT prolongation and sinus tachycardia following citalopram overdose have occurred, torsade de pointes is rare. Obtain a baseline ECG, continuous cardiac monitoring, and serial ECGs following significant exposure.
    a) ECG abnormalities including QTc prolongation, sinus tachycardia, and inferolateral repolarization abnormalities have occurred following severe citalopram overdoses (400 to 5200 mg).
    b) According to simulation studies that were conducted using a previously developed pharmacokinetic-pharmacodynamic model that predicted QT interval prolongation, ingestion of citalopram doses greater than 1000 mg should be treated with activated charcoal and cardiac function should be monitored, with a minimum monitoring time of 13 hours. The simulations demonstrated that, in patients with a normal QT interval at 13 hours, the risk of the QT interval increasing at a later time was less than 1%. For those patients who ingested at least 600 mg of citalopram and who did not receive activated charcoal within 4 hours postingestion, a 13-hour cardiac monitoring time was also recommended (Isbister et al, 2006; Friberg et al, 2006).
    2) Monitor serum electrolytes (including potassium and bicarbonate); metabolic acidosis has occurred following significant exposure.
    C) CARDIOTOXICITY
    1) ECG abnormalities including QTc prolongation, sinus tachycardia, and inferolateral repolarization abnormalities have occurred following severe citalopram overdoses (400 to 5200 mg) (Grundemar et al, 1997).
    a) Severe sinus bradycardia, hypotension, and syncope associated with citalopram overdose have been successfully treated with a temporary pacemaker (Rothenhausler et al, 2000).
    2) SODIUM BICARBONATE: Intravenous sodium bicarbonate has been used as an antidote to SSRI-induced bradyarrhythmias (Brucculeri et al, 2005).
    a) An 82-year-old woman developed a generalized tonic-clonic seizure approximately 12 hours after ingesting citalopram 1.6 g in a suicide attempt. Although her cardiovascular and pulmonary examinations were unremarkable, an initial ECG showed sinus bradycardia with a ventricular rate of 58 beats/min, left axis deviation, and left bundle-branch block, which were consistent with her previous ECGs. In addition, the QRS duration, QT interval, and QTCs interval were 146 msec, 544 msec, and 534 msec, respectively. Three hours after she was transferred to telemetry service, marked bradycardia was observed. A 12-lead ECG showed a junctional escape rhythm with a ventricular rate of 40 beats/min, retrograde ventriculoatrial activation, and an underlying left bundle-branch block. The QRS duration, QT interval, and QTc interval were 152 msec, 646 msec, and 527 msec, respectively. The administration of sodium bicarbonate solution 50 mEq (50 mL of 8.4% solution) by direct intravenous injection on 2 different occasions caused a dramatic but temporary impact on the ECG findings. These effects quickly reverted to the previous junctional bradycardia. Approximately 3 hours after starting a continuous infusion of sodium bicarbonate 150 mEq in 1 liter of 5% dextrose in water (a rate of 75 mL/hr or 11.25 mEq/hr), normal sinus rhythm with a rate of 63 beats/min, a QT interval of 510 msec, and a QTc interval of 504 msec were noted. After 36 hours, the infusion was stopped, and no new ECG abnormalities were observed (Brucculeri et al, 2005).
    D) TORSADES DE POINTES
    1) SUMMARY
    a) Withdraw the causative agent. Hemodynamically unstable patients with Torsades de pointes (TdP) require electrical cardioversion. Emergent treatment with magnesium (first-line agent) or atrial overdrive pacing is indicated. Detect and correct underlying electrolyte abnormalities (ie, hypomagnesemia, hypokalemia, hypocalcemia). Correct hypoxia, if present (Drew et al, 2010; Neumar et al, 2010; Keren et al, 1981; Smith & Gallagher, 1980).
    b) Polymorphic VT associated with acquired long QT syndrome may be treated with IV magnesium. Overdrive pacing or isoproterenol may be successful in terminating TdP, particularly when accompanied by bradycardia or if TdP appears to be precipitated by pauses in rhythm (Neumar et al, 2010). In patients with polymorphic VT with a normal QT interval, magnesium is unlikely to be effective (Link et al, 2015).
    2) MAGNESIUM SULFATE
    a) Magnesium is recommended (first-line agent) for the prevention and treatment of drug-induced torsades de pointes (TdP) even if the serum magnesium concentration is normal. QTc intervals greater than 500 milliseconds after a potential drug overdose may correlate with the development of TdP (Charlton et al, 2010; Drew et al, 2010). ADULT DOSE: No clearly established guidelines exist; an optimal dosing regimen has not been established. Administer 1 to 2 grams diluted in 10 milliliters D5W IV/IO over 15 minutes (Neumar et al, 2010). Followed if needed by a second 2 gram bolus and an infusion of 0.5 to 1 gram (4 to 8 mEq) per hour in patients not responding to the initial bolus or with recurrence of dysrhythmias (American Heart Association, 2005; Perticone et al, 1997). Rate of infusion may be increased if dysrhythmias recur. For persistent refractory dysrhythmias, a continuous infusion of up to 3 to 10 milligrams/minute in adults may be given (Charlton et al, 2010).
    b) PEDIATRIC DOSE: 25 to 50 milligrams/kilogram diluted to 10 milligrams/milliliter for intravenous infusion over 5 to 15 minutes up to 2 g (Charlton et al, 2010).
    c) PRECAUTIONS: Use with caution in patients with renal insufficiency.
    d) MAJOR ADVERSE EFFECTS: High doses may cause hypotension, respiratory depression, and CNS toxicity (Neumar et al, 2010). Toxicity may be observed at magnesium levels of 3.5 to 4.0 mEq/L or greater (Charlton et al, 2010).
    e) MONITORING PARAMETERS: Monitor heart rate and rhythm, blood pressure, respiratory rate, motor strength, deep tendon reflexes, serum magnesium, phosphorus, and calcium concentrations (Prod Info magnesium sulfate heptahydrate IV, IM injection, solution, 2009).
    3) OVERDRIVE PACING
    a) Institute electrical overdrive pacing at a rate of 130 to 150 beats per minute, and decrease as tolerated. Rates of 100 to 120 beats per minute may terminate torsades (American Heart Association, 2005). Pacing can be used to suppress self-limited runs of TdP that may progress to unstable or refractory TdP, or for override refractory, persistent TdP before the potential development of ventricular fibrillation (Charlton et al, 2010). In a case series overdrive pacing was successful in terminating TdP associated with bradycardia and drug-induced QT prolongation (Neumar et al, 2010).
    4) POTASSIUM REPLETION
    a) Potassium supplementation, even if serum potassium is normal, has been recommended by many experts (Charlton et al, 2010; American Heart Association, 2005). Supplementation to supratherapeutic potassium concentrations of 4.5 to 5 mmol/L has been suggested, although there is little evidence to determine the optimal range in dysrhythmia (Drew et al, 2010; Charlton et al, 2010).
    5) ISOPROTERENOL
    a) Isoproterenol has been successful in aborting torsades de pointes that was resistant to magnesium therapy in a patient in whom transvenous overdrive pacing was not an option (Charlton et al, 2010) and has been successfully used to treat torsades de pointes associated with bradycardia and drug induced QT prolongation (Keren et al, 1981; Neumar et al, 2010). Isoproterenol may have a limited role in pharmacologic overdrive pacing in select patients with drug-induced torsades de pointes and acquired long QT syndrome (Charlton et al, 2010; Neumar et al, 2010). Isoproterenol should be avoided in patients with polymorphic VT associated with familial long QT syndrome (Neumar et al, 2010).
    b) DOSE: ADULT: 2 to 10 micrograms/minute via a continuous monitored intravenous infusion; titrate to heart rate and rhythm response (Neumar et al, 2010).
    c) PRECAUTIONS: Correct hypovolemia before using; contraindicated in patients with acute cardiac ischemia (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    1) Contraindicated in patients with preexisting dysrhythmias; tachycardia or heart block due to digitalis toxicity; ventricular dysrhythmias that require inotropic therapy; and angina. Use with caution in patients with coronary insufficiency (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    d) MAJOR ADVERSE EFFECTS: Tachycardia, cardiac dysrhythmias, palpitations, hypotension or hypertension, nervousness, headache, dizziness, and dyspnea (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    e) MONITORING PARAMETERS: Monitor heart rate and rhythm, blood pressure, respirations and central venous pressure to guide volume replacement (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    6) OTHER DRUGS
    a) Mexiletine, verapamil, propranolol, and labetalol have also been used to treat TdP, but results have been inconsistent (Khan & Gowda, 2004).
    7) AVOID
    a) Avoid class Ia antidysrhythmics (eg, quinidine, disopyramide, procainamide, aprindine), class Ic (eg, flecainide, encainide, propafenone) and most class III antidysrhythmics (eg, N-acetylprocainamide, sotalol) since they may further prolong the QT interval and have been associated with TdP.
    E) ACIDOSIS
    1) METABOLIC ACIDOSIS: Treat severe metabolic acidosis (pH less than 7.1) with sodium bicarbonate, 1 to 2 mEq/kg is a reasonable starting dose(Kraut & Madias, 2010). Monitor serum electrolytes and arterial or venous blood gases to guide further therapy.
    2) Repeat doses of no more than one-half the original amount may be given no more often than every 10 minutes if required.
    3) Iatrogenic hypernatremia and alkalemia greater than 7.65 can be associated with life-threatening CNS complications.
    F) HYPOKALEMIA
    1) Monitor potassium following significant exposure; replace as indicated.
    G) RHABDOMYOLYSIS
    1) SUMMARY: Early aggressive fluid replacement is the mainstay of therapy and may help prevent renal insufficiency. Diuretics such as mannitol or furosemide may be added if necessary to maintain urine output but only after volume status has been restored as hypovolemia will increase renal tubular damage. Urinary alkalinization is NOT routinely recommended.
    2) Initial treatment should be directed towards controlling acute metabolic disturbances such as hyperkalemia, hyperthermia, and hypovolemia. Control seizures, agitation, and muscle contractions (Erdman & Dart, 2004).
    3) FLUID REPLACEMENT: Early and aggressive fluid replacement is the mainstay of therapy to prevent renal failure. Vigorous fluid replacement with 0.9% saline (10 to 15 mL/kg/hour) is necessary even if there is no evidence of dehydration. Several liters of fluid may be needed within the first 24 hours (Walter & Catenacci, 2008; Camp, 2009; Huerta-Alardin et al, 2005; Criddle, 2003; Polderman, 2004). Hypovolemia, increased insensible losses, and third spacing of fluid commonly increase fluid requirements. Strive to maintain a urine output of at least 1 to 2 mL/kg/hour (or greater than 150 to 300 mL/hour) (Walter & Catenacci, 2008; Camp, 2009; Erdman & Dart, 2004; Criddle, 2003). To maintain a urine output this high, 500 to 1000 mL of fluid per hour may be required (Criddle, 2003). Monitor fluid input and urine output, plus insensible losses. Monitor for evidence of fluid overload and compartment syndrome; monitor serum electrolytes, CK, and renal function tests.
    4) DIURETICS: Diuretics (eg, mannitol or furosemide) may be needed to ensure adequate urine output and to prevent acute renal failure when used in combination with aggressive fluid therapy. Loop diuretics increase tubular flow and decrease deposition of myoglobin. These agents should be used only after volume status has been restored, as hypovolemia will increase renal tubular damage. If the patient is maintaining adequate urine output, loop diuretics are not necessary (Vanholder et al, 2000).
    5) URINARY ALKALINIZATION: Alkalinization of the urine is not routinely recommended, as it has never been documented to reduce nephrotoxicity, and may cause complications such as hypocalcemia and hypokalemia (Walter & Catenacci, 2008; Huerta-Alardin et al, 2005; Brown et al, 2004; Polderman, 2004). Retrospective studies have failed to demonstrate any clinical benefit from the use of urinary alkalinization (Brown et al, 2004; Polderman, 2004; Homsi et al, 1997).
    H) HYPOGLYCEMIA
    1) Administer 50% dextrose 50 mL IV to adults with severe hypoglycemia. Monitor serum glucose frequently; recurrent hypoglycemia requiring dextrose infusion has been reported after citalopram overdose (Duncan et al, 2008).
    I) SEROTONIN SYNDROME
    1) SUMMARY
    a) Benzodiazepines are the mainstay of therapy. Cyproheptadine, a 5-HT antagonist, is also commonly used. Severe cases have been managed with benzodiazepine sedation and neuromuscular paralysis with non-depolarizing agents(Claassen & Gelissen, 2005).
    2) HYPERTHERMIA
    a) Control agitation and muscle activity. Undress patient and enhance evaporative heat loss by keeping skin damp and using cooling fans.
    b) MUSCLE ACTIVITY: Benzodiazepines are the drug of choice to control agitation and muscle activity. DIAZEPAM: ADULT: 5 to 10 mg IV every 5 to 10 minutes as needed, monitor for respiratory depression and need for intubation. CHILD: 0.25 mg/kg IV every 5 to 10 minutes; monitor for respiratory depression and need for intubation.
    c) Non-depolarizing paralytics may be used in severe cases.
    3) CYPROHEPTADINE
    a) Cyproheptadine is a non-specific 5-HT antagonist that has been shown to block development of serotonin syndrome in animals (Sternbach, 1991). Cyproheptadine has been used in the treatment of serotonin syndrome (Mills, 1997; Goldberg & Huk, 1992). There are no controlled human trials substantiating its efficacy.
    b) ADULT: 12 mg initially followed by 2 mg every 2 hours if symptoms persist, up to a maximum of 32 mg in 24 hours. Maintenance dose 8 mg orally repeated every 6 hours (Boyer & Shannon, 2005).
    c) CHILD: 0.25 mg/kg/day divided every 6 hours, maximum dose 12 mg/day (Mills, 1997).
    4) HYPERTENSION
    a) Monitor vital signs regularly. For mild/moderate asymptomatic hypertension, pharmacologic intervention is usually not necessary.
    5) HYPOTENSION
    a) Administer 10 to 20 mL/kg 0.9% saline bolus and place patient supine. Further fluid therapy should be guided by central venous pressure or right heart catheterization to avoid volume overload.
    b) Pressor agents with dopaminergic effects may theoretically worsen serotonin syndrome and should be used with caution. Direct acting agents (norepinephrine, epinephrine, phentolamine) are theoretically preferred.
    c) NOREPINEPHRINE
    1) PREPARATION: Add 4 mL of 0.1% solution to 1000 mL of dextrose 5% in water to produce 4 mcg/mL.
    2) INITIAL DOSE
    a) ADULT: 2 to 3 mL (8 to 12 mcg)/minute.
    b) ADULT or CHILD: 0.1 to 0.2 mcg/kg/min. Titrate to maintain adequate blood pressure.
    3) MAINTENANCE DOSE
    a) 0.5 to 1 mL (2 to 4 mcg)/minute.
    6) SEIZURES
    a) DIAZEPAM
    1) MAXIMUM RATE: Administer diazepam IV over 2 to 3 minutes (maximum rate: 5 mg/min).
    2) ADULT DIAZEPAM DOSE: 5 to 10 mg initially, repeat every 5 to 10 minutes as needed. Monitor for hypotension, respiratory depression and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after diazepam 30 milligrams.
    3) PEDIATRIC DIAZEPAM DOSE: 0.2 to 0.5 mg/kg, repeat every 5 minutes as needed. Monitor for hypotension, respiratory depression and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after diazepam 10 milligrams in children over 5 years or 5 milligrams in children under 5 years of age.
    4) RECTAL USE: If an intravenous line cannot be established, diazepam may be given per rectum (not FDA approved), or lorazepam may be given intramuscularly.
    b) LORAZEPAM
    1) MAXIMUM RATE: The rate of IV administration of lorazepam should not exceed 2 mg/min (Prod Info Ativan(R), 1991).
    2) ADULT LORAZEPAM DOSE: 2 to 4 mg IV. Initial doses may be repeated in 10 to 15 minutes, if seizures persist (Prod Info ATIVAN(R) injection, 2003).
    3) PEDIATRIC LORAZEPAM DOSE: 0.1 mg/kg IV push (range: 0.05 to 0.1 mg/kg; maximum dose 4 mg); may repeat dose in 5 to 10 minutes if seizures continue. It has also been given rectally at the same dose in children with no IV access (Sreenath et al, 2009; Chin et al, 2008; Wheless, 2004; Qureshi et al, 2002; De Negri & Baglietto, 2001; Mitchell, 1996; Appleton, 1995; Giang & McBride, 1988).
    c) RECURRING SEIZURES
    1) If seizures cannot be controlled with diazepam or recur, give phenobarbital or propofol.
    d) PHENOBARBITAL
    1) SERUM LEVEL MONITORING: Monitor serum levels over next 12 to 24 hours for maintenance of therapeutic levels (15 to 25 mcg/mL).
    2) ADULT PHENOBARBITAL LOADING DOSE: 600 to 1200 mg of phenobarbital IV initially (10 to 20 mg/kg) diluted in 60 mL of 0.9% saline given at 25 to 50 mg/minute.
    3) ADULT PHENOBARBITAL MAINTENANCE DOSE: Additional doses of 120 to 240 mg may be given every 20 minutes.
    4) MAXIMUM SAFE ADULT PHENOBARBITAL DOSE: No maximum safe dose has been established. Patients in status epilepticus have received as much as 100 mg/min until seizure control was achieved or a total dose of 10 mg/kg.
    5) PEDIATRIC PHENOBARBITAL LOADING DOSE: 15 to 20 mg/kg of phenobarbital intravenously at a rate of 25 to 50 mg/min.
    6) PEDIATRIC PHENOBARBITAL MAINTENANCE DOSE: Repeat doses of 5 to 10 mg/kg may be given every 20 minutes.
    7) MAXIMUM SAFE PEDIATRIC PHENOBARBITAL DOSE: No maximum safe dose has been established. Children in status epilepticus have received doses of 30 to 120 mg/kg within 24 hours. Vasopressors and mechanical ventilation were needed in some patients receiving these doses.
    8) NEONATAL PHENOBARBITAL LOADING DOSE: 20 to 30 mg/kg IV at a rate of no more than 1 mg/kg/min in patients with no preexisting phenobarbital serum levels.
    9) NEONATAL PHENOBARBITAL MAINTENANCE DOSE: Repeat doses of 2.5 mg/kg every 12 hours may be given; adjust dosage to maintain serum levels of 20 to 40 mcg/mL.
    10) MAXIMUM SAFE NEONATAL PHENOBARBITAL DOSE: Doses of up to 20 mg/kg/min up to a total of 30 mg/kg have been tolerated in neonates.
    11) CAUTION: Adequacy of ventilation must be continuously monitored in children and adults. Intubation may be necessary with increased doses.
    7) CHLORPROMAZINE
    a) Chlorpromazine is a 5-HT2 receptor antagonist that has been used to treat cases of serotonin syndrome (Graham, 1997; Gillman, 1996). Controlled human trial documenting its efficacy are lacking.
    b) ADULT: 25 to 100 mg intramuscularly repeated in 1 hour if necessary.
    c) CHILD: 0.5 to 1 mg/kg repeated as needed every 6 to 12 hours not to exceed 2 mg/kg/day.
    8) NOT RECOMMENDED
    a) BROMOCRIPTINE: It has been used in the treatment of neuroleptic malignant syndrome but is NOT RECOMMENDED in the treatment of serotonin syndrome as it has serotonergic effects (Gillman, 1997). In one case the use of bromocriptine was associated with a fatal outcome (Kline et al, 1989).
    J) EXPERIMENTAL THERAPY
    1) IV LIPID EMULSION THERAPY
    a) CASE REPORT: A 21-year-old man presented to the emergency department (ED) unresponsive and with seizures after an overdose ingestion of citalopram and olanzapine, with potential maximum ingestions of 11.6 g and 600 mg, respectively. An ECG indicated wide complex tachycardia. Peak serum citalopram concentration, measured within 30 minutes post-presentation, was 522 ng/mL. Approximately 2.5 hours after arrival to the ED, following a second seizure, the patient developed hypotension that worsened over the next several hours, requiring an epinephrine infusion to maintain a mean arterial pressure of 60 mmHg. Bradycardia (less than 40 beats/min) developed approximately 6.5 hours post-presentation, and was unresponsive to atropine and isoproterenol, necessitating placement of a transvenous pacemaker. Approximately 10 hours post-presentation, a third seizure occurred. Because of progressive cardiac and neurotoxicity, IV lipid emulsion (ILE) therapy was initiated, at a total dose of 10% lipid emulsion at 21 ml/hour (0.0005 mL/kg/min administered for a total of 17 hours). There was no bolus dose administered prior to the infusion, and the infusion dose administered was 50-fold lower than the recommended dose. Following the infusion, the patient's condition stabilized with no further clinical worsening. The pacemaker was discontinued 24 hours post-presentation and the epinephrine infusion was titrated down and discontinued 5 hours later. Because the requirements for pacing and epinephrine infusion administration remained unchanged for 12 hours following initiation of ILE and there was no significant increase in the patient's serum triglyceride levels, it is believed that low-dose ILE had no significant effect on the patient's clinical status (Lung et al, 2013).

Enhanced Elimination

    A) HEMODIALYSIS
    1) Hemodialysis and hemoperfusion are NOT of value due to the large volume of distribution.

Summary

    A) Acute ingestion of up to 100 mg citalopram or 50 mg escitalopram is not expected to result in toxicity. Serotonin toxicity may develop at therapeutic doses, particularly if another medication that increases CNS serotonin is used concomitantly. Minor toxicity develops with overdose less than 600 mg citalopram. Ingestion of more than 600 mg citalopram or more than 300 mg escitalopram requires cardiac monitoring for 8 hours (11 hours if the patient did not receive activated charcoal within 4 hours of ingestion). Ingestion of more than 1000 mg citalopram or more than 500 mg escitalopram requires cardiac monitoring for 13 hours. At the end of the observation period, if the QTc is less than 450 msec, the monitoring can be discontinued, and the patient may be discharged if asymptomatic. Patients with symptoms of toxicity or a QTc of greater than 450 msec at the end of the observation period should be admitted for continued cardiac monitoring. Seizures have been reported after ingestions of more than 600 mg citalopram, and serious toxicity after ingestion of 800 mg or more. PEDIATRIC: QTc prolongation has been reported after ingestion of escitalopram 200 mg.
    B) THERAPEUTIC DOSE: ADULT: Citalopram: Initially, 20 mg/day orally; max dose 40 mg/day. Escitalopram: 10 to 20 mg/day orally; max dose 20 mg/day. PEDIATRIC: Safety and effectiveness in children have not been established.

Therapeutic Dose

    7.2.1) ADULT
    A) CITALOPRAM
    1) The usual recommended dose is 20 mg orally once daily initially, and increased, in 20-mg increments at intervals at least a week, to a maximum dose of 40 mg orally once daily (Prod Info CITALOPRAM HYDROBROMIDE oral solution, 2015)
    B) ESCITALOPRAM
    1) MAJOR DEPRESSIVE DISORDER: The usual recommended dose is 10 mg orally once daily. The dose may be increased up to a maximum of 20 mg once daily after a minimum of week (Prod Info Lexapro(R) oral tablets, oral solution, 2014).
    2) GENERALIZED ANXIETY DISORDER: The usual recommended dose is 10 mg orally once daily. After at least one week at this dosage regimen, the dose may be increased to 20 mg once daily (Prod Info Lexapro(R) oral tablets, oral solution, 2014).
    7.2.2) PEDIATRIC
    A) CITALOPRAM
    1) Safety and efficacy have not been established in pediatric patients (Prod Info CITALOPRAM HYDROBROMIDE oral solution, 2015).
    B) ESCITALOPRAM
    1) 12 TO 17 YEARS: For major depressive disorder, the recommended dose is 10 mg orally once daily. The dose may be increased up to a maximum of 20 mg orally once daily after a minimum of 3 weeks(Prod Info Lexapro(R) oral tablets, oral solution, 2014).
    a) Safety and efficacy have not been established for the treatment of generalized anxiety disorder in adolescent patients (Prod Info Lexapro(R) oral tablets, oral solution, 2014).
    2) LESS THAN 12 YEARS: Safety and efficacy have not been established for the treatment of depression or generalized anxiety disorder in pediatric patients less than 12 years of age (Prod Info Lexapro(R) oral tablets, oral solution, 2014).

Minimum Lethal Exposure

    A) ADULT
    1) CASE REPORT: A 53-year-old woman died following a probable overdose of citalopram 840 mg; coingestants included diazepam (0.1 mcg/g). At necropsy, citalopram concentration was 6.1 mcg/g femoral vein blood (Ostrom et al, 1996).
    2) CASE REPORT: A 56-year-old man died following an intentional ingestion of citalopram 3920 mg (equivalent to 2 bottles of 100 tablets); no other coingestants were found on necropsy (Glassman, 1997; Ostrom et al, 1996).
    3) CASE REPORT: A 22-year-old woman died following an intentional citalopram ingestion of 2880 mg (approximately 40 times the maximum daily dose). No other drugs or ethanol were detected in her urine or blood. Toxicologic analysis of her blood showed a citalopram concentration of 11.6 mg/L (Luchini et al, 2005).
    4) CASE REPORT: A 24-year-old woman developed progressive hypotension, tonic-clonic seizures, QTc interval prolongation, and pulseless ventricular tachycardia, unresponsive to aggressive resuscitative measures including defibrillation, external pacing, and overdrive chemical pacing, resulting in death approximately 19 hours after a suspected overdose ingestion of citalopram, bupropion, clonazepam, and trazodone. Potentially, the maximum doses ingested were 1.8 g of citalopram, 13.5 g of bupropion, 90 mg of clonazepam, and 4.5 g of trazodone. Post-mortem serum concentrations revealed supratherapeutic concentrations of citalopram (400 ng/mL) and bupropion (440 ng/mL); citalopram, clonazepam, and trazodone serum concentrations were within the normal therapeutic ranges (Lung et al, 2012).
    B) ACUTE
    1) LDLo: (ORAL) HUMAN, Male: 56 mg/kg (RTECS , 2000)

Maximum Tolerated Exposure

    A) SUMMARY
    1) Acute ingestion of up to 100 mg citalopram or 50 mg escitalopram is not expected to result in more than mild symptoms(Nelson et al, 2007).
    2) Ingestion of more than 600 mg citalopram or more than 300 mg escitalopram requires cardiac monitoring for 8 hours (11 hours if the patient did not receive activated charcoal within 4 hours of ingestion). Ingestion of more than 1000 mg citalopram or more than 500 mg escitalopram requires cardiac monitoring for 13 hours. At the end of the observation period, if the QTc is less than 450 msec, the monitoring can be discontinued, and the patient may be discharged if asymptomatic. Patients with symptoms of toxicity or a QTc of greater than 450 msec at the end of the observation period should be admitted for continued cardiac monitoring (Friberg et al, 2006; Isbister et al, 2006).
    3) A case review, involving inadvertent pediatric ingestions of citalopram from 2000 to 2014, identified 335 patients (6 years of age or less) who ingested citalopram as the sole drug. Estimated doses were reported in 119 cases, with unknown weights estimated at the 50th percentile weight for age. Doses ranged from 0.2 to 9.57 mg/kg and 1 40-mg/kg dose. Minor effects (ie, hyperactivity, drowsy, vomiting, abdominal pain) occurred at doses of 0.41 to 2.5 mg/kg, moderate effects of drowsiness and fever occurred at 1.67 mg/kg, and a seizure was reported with a 5 mg/kg dose. Based on these results, it is suggested that pediatric ingestions of less than 5 mg/kg will not cause serious toxicity and may be safely managed outside of a healthcare facility (Herrington et al, 2015).
    B) ADULT
    1) CASE REPORTS
    a) A 23-year-old man ingested citalopram 920 mg alone and developed a generalized tonic-clonic seizure approximately 2.5 hours after ingestion. Supraventricular tachycardia (rate 160s) developed approximately 20 minutes later and was successfully treated with adenosine (Cuenca et al, 2004).
    b) A 41-year-old man tolerated an estimated overdose of citalopram 5200 mg with transient effects of seizures, metabolic acidosis, hypotension for 1 hour, and ECG changes (QTc prolongation and sinus tachycardia); no permanent sequelae were reported (Grundemar et al, 1997).
    c) A 32-year-old woman developed severe sinus bradycardia (41 beats/min), hypotension, and episodes of syncope approximately 4 hours after ingesting citalopram 800 mg in a suicide attempt. The patient recovered following insertion of a temporary pacemaker (Rothenhausler et al, 2000).
    d) A 20-year-old woman developed severe hypoglycemia (1.9 mmol/L) and generalized tonic-clonic seizures after ingesting citalopram 2760 mg. The patient recovered following supportive treatment (Duncan et al, 2008).
    e) A 21-year-old man developed seizures, ECG abnormalities, bradycardia, and hypotension after an overdose ingestion of citalopram and olanzapine, with potential maximum ingestions of 11.6 g and 600 mg, respectively. He gradually recovered with supportive therapy (Lung et al, 2013).
    2) CASE SERIES
    a) In prospective study by the National Poison Control Centre (the Netherlands) conducted during a 1-year period, 42 cases of acute citalopram exposure occurred with no serious toxicity reported. Seven patients were asymptomatic (5 with citalopram alone and 2 as a mixed ingestion). The overdose ranges were 100 to 780 mg in citalopram alone and 100 to 2240 mg in a mixed exposure. Drowsiness, tremor, tachycardia, and gastrointestinal symptoms were the most frequent effects observed; 2 patients developed minor cardiac effects (ie, chest pain and tachycardia) and 1 patient had a seizure. The authors concluded that citalopram alone exposures are generally mild, but cardiovascular effects and seizures may occur (Wijnands-Kleukers et al, 2002).
    b) In a series of 108 patients with citalopram overdose, patients who ingested less than 600 mg (n=41) developed mild effects (nausea, dizziness, tachycardia, tremor, drowsiness, somnolence). Doses of 600 mg to 1.9 g were associated with seizures in 6 of 34 patients (18%). Seizures developed in 9 of 19 patients (47%) who ingested 1.9 to 5.2 g (Personne et al, 1997).
    c) Seizures developed following overdoses which exceeded 600 mg. Five nonfatal cases of citalopram overdose (up to 5200 mg) resulted in seizures developing in 4 patients. All 5 patients had QT prolongation, sinus tachycardia, and inferolateral repolarization disturbances (Grundemar et al, 1997).
    d) Persistent lethargy was the only reported adverse outcome in a retrospective analysis of charts from 28 isolated escitalopram ingestions during a 1-year period. The average amount of escitalopram ingested was 62.5 mg (ranging from 5 to 300 mg in 21 cases) (LoVecchio et al, 2006).
    e) In a retrospective review involving 26 patients, nausea and vomiting, tachycardia, QTc interval prolongation, seizures, drowsiness, and coma were reported following citalopram overdose ingestions in amounts ranging from 200 to 4960 mg (median 1190 mg) (Jimmink et al, 2008).
    3) LACK OF EFFECT: Doses as high as 2000 mg have been taken without observable cardiovascular abnormalities (Milne & Goa, 1991).
    C) PEDIATRIC
    1) ESCITALOPRAM/CASE REPORT: A 14-year-old girl ingested escitalopram 200 mg and subsequently developed lightheadedness, tremors, and upper abdominal discomfort. An ECG, performed approximately 24 hours postingestion, showed normal sinus rhythm with QTc interval prolongation (450 msec). The next day, a repeat ECG showed a worsening of the QTc interval (469 msec) that then spontaneously normalized (Scharko & Schumacher, 2008).
    2) ESCITALOPRAM/CASE REPORT: A 16-year-old girl developed incomplete right bundle branch block with QRS complex widening (110 ms) and QTc interval prolongation (509 ms) after intentionally ingesting 500 mg escitalopram, 25 tablets of tramadol/acetaminophen, and an unknown amount of hydrocodone/acetaminophen. With supportive care, including IV administration of sodium bicarbonate and magnesium sulfate, the patient recovered. Her serum escitalopram concentration, obtained approximately 8 hours post-ingestion, was 45 mcg/L (Schreffler et al, 2013).

Serum Plasma Blood Concentrations

    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) ADULT
    a) CASE SERIES
    1) In a series of 30 citalopram deaths, mean postmortem blood concentration in suicide cases (n=20) was 1.83 mcg/mL (range 0.04 to 13.9 mcg/mL), and in accidental deaths (n=8) the average blood level was 1.12 mcg/mL (range 0.09 to 5.72 mcg/mL). In this study, citalopram overdose was not considered the cause of death in subjects with postmortem blood levels of less than 0.35 mcg/mL, when citalopram was taken alone. In addition, citalopram levels between 0.37 and 0.83 mcg/mL did not always produce toxicity in all cases (Segura & Bravo, 2004).
    2) In a retrospective review involving 26 patients, nausea and vomiting, tachycardia, QTc interval prolongation, seizures, drowsiness, and coma were reported following citalopram overdose ingestions in amounts ranging from 200 to 4960 mg (median 1190 mg). Serum citalopram concentrations, obtained from 20 minutes to 8 hours (median 3 hours) postingestion, ranged from 0.21 to 7.5 mg/L (median 1.13 mg/L) (Jimmink et al, 2008).
    b) CASE REPORTS
    1) A serum citalopram level of 5.88 mg/L (therapeutic range 0.01 to 0.2 mg/L) was detected in a 54-year-old woman after she intentionally ingested an unknown amount of citalopram with alcohol. She presented to the emergency department (time of ingestion unknown) with altered mental status, hypotension, and low O2 saturation. Laboratory analysis showed a blood ethanol level of 0.2 g/L and urine screening was negative for opiates, cocaine, benzodiazepines, and tricyclic antidepressants. She was treated with 50 g activated charcoal and admitted to ICU. Sustained ventricular tachycardia with VF episodes occurred 10 hours after ICU admission and continued for 48 hours. External electric shocks and sternum strikes were administered. She recovered fully and was discharged 16 days after admission (Liotier & Coudore, 2011).
    2) An overdose of 840 mg in an adult resulted in death. At necropsy, citalopram concentration was 6.1 mcg/g femoral vein blood (Ostrom et al, 1996).
    3) A combined overdose of trimipramine and citalopram in an adult woman resulted in postmortem femoral blood levels of 2.33 mg/kg and 4.81 mg/kg, respectively (Musshoff et al, 1999).
    4) At autopsy a serum citalopram level of 3,402 ng/mL was thought to be the cause of death in a 38-year-old man who had a history of depression and was found pulseless (Carlton & Hughes, 2001).
    5) A seizure and transient ECG abnormalities occurred in an adult man following an overdose of citalopram (total 1800 mg, with a serum level of 840 ng/mL) and fluoxetine (serum level within normal limits) (Grover & Caravati, 2001).
    6) A maximum serum citalopram concentration of 1.92 mg/L occurred 2 hours following an intentional ingestion of citalopram hydrobromide 3000 mg (citalopram 2400 mg free base). The maximum serum norcitalopram concentration of 0.27 mg/L occurred approximately 24 hours postingestion (Kelly et al, 2003).
    7) A 36-year-old woman developed palpitation, weakness, nausea, and numbness 36 hours after taking citalopram 1000 mg with alcohol. On day 3, her vital signs were blood pressure, 84/44 mmHg; temperature, 99.3 degrees F; pulse, 102 to 160 beats/min; and respiratory rate, 17 breaths/min. ECG revealed intermittent runs of wide-complex tachycardia with a QTc of 600 msec. Despite saline and magnesium sulfate infusions, she developed intermittent bigeminy, and despite IV lidocaine, intermittent torsade de pointes occurred. After receiving potassium chloride, transvenous pacemaker insertion, and isoproterenol infusion, she converted to normal sinus rhythm, and QTCs interval narrowed to 442 msec after 2 days. Her plasma citalopram level was 477 ng/mL (therapeutic range 40 to 110 ng/mL), with a corresponding desmethylcitalopram level of 123.2 ng/mL (therapeutic range 14 to 40 ng/mL) (Tarabar et al, 2003).
    8) A 22-year-old woman died following intentional ingestion of citalopram 2880 mg. Citalopram concentrations in her body fluids and organ tissues were as follows (Luchini et al, 2005):
    1) Blood: 11.6 mg/L
    2) Urine: 149.67 mg/L
    3) Brain: 22.295 mcg/g
    4) Lung: 51.235 mcg/g
    5) Heart: 4.28 mcg/g
    6) Spleen: 32.195 mcg/g
    7) Liver: 36.945 mcg/g

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) HUMAN DATA

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) Alaspaa AO, Kuisma MJ, Hoppu K, et al: Out-of-hospital administration of activated charcoal by emergency medical services. Ann Emerg Med 2005; 45:207-12.
    3) Alwan S, Reefhuis J, Rasmussen SA, et al: Use of selective serotonin-reuptake inhibitors in pregnancy and the risk of birth defects. N Engl J Med 2007; 356(26):2684-2692.
    4) American Heart Association: 2005 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2005; 112(24 Suppl):IV 1-203. Available from URL: http://circ.ahajournals.org/content/vol112/24_suppl/. As accessed 12/14/2005.
    5) Anon: Citalopram for depression. Med Lett 1998; 40:113-114.
    6) Appleton R: Lorazepam vs diazepam in the acute treatment of epileptic seizures and status epilepticus.. Dev Med Child Neuro 1995; 37:682-688.
    7) Ayudin N, Anac E, Caykoylu A, et al: Neuroleptic malignant syndrome due to citalopram overdose (letter). Can J Psychiatry 2000; 45:941-942.
    8) Baettig D, Bondolfi G, Montaldi S, et al: Tricyclic antidepressant plasma levels after augmentation with citalopram: a case study. Eur J Clin Pharmacol 1993; 44:403-405.
    9) Barclay TS & Lee AJ: Citalopram-associated SIADH. Ann Pharmacother 2002; 36:1558-1563.
    10) Berle JO, Steen VM, Aamo TO, et al: Breastfeeding during maternal antidepressant treatment with serotonin reuptake inhibitors: infant exposure, clinical symptoms, and cytochrome P450 genotypes. J Clin Psychiatry 2004; 65:1228-1234.
    11) Bouchard JM, Delaunay J, & Delisle JP: Citalopram versus maprotiline: a controlled, clinical multicentre trial in depressed patients. Acta Psychiatr Scand 1987; 76:583-592.
    12) Boukhris T, Sheehy O, Mottron L, et al: Antidepressant use during pregnancy and the risk of autism spectrum disorder in children. JAMA Pediatr 2016; 170(2):117-124.
    13) Boyer EW & Shannon M: The serotonin syndrome. N Eng J Med 2005; 352(11):1112-1120.
    14) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    15) Brown CV, Rhee P, Chan L, et al: Preventing renal failure in patients with rhabdomyolysis: do bicarbonate and mannitol make a difference?. J Trauma 2004; 56(6):1191-1196.
    16) Brucculeri M, Kaplan J, & Lande L: Reversal of citalopram-induced junctional bradycardia with intravenous sodium bicarbonate. Pharmacother 2005; 25(1):119-122.
    17) Burrows GD, McIntyre IM, & Judd FK: Clinical effects of serotonin reuptake inhibitors in the treatment of depressive illness. J Clin Psychiatry 1988; 49(suppl):18-22.
    18) Camp NE: Drug- and toxin-induced Rhabdomyolysis. J Emerg Nurs 2009; 35(5):481-482.
    19) Caravati EM, Knight HH, & Linscott MS: Esophageal laceration and charcoal mediastinum complicating gastric lavage. J Emerg Med 2001; 20:273-276.
    20) Carlton FB & Hughes MP: Fatal overdose of citalopram (abstract). J Toxicol - Clin Toxicol 2001; 39:491.
    21) Catalano G, Catalano MC, & Epstein MA: QTc interval prolongation associated with citalopram overdose: a case report and literature review. Clin Neuropharmacol 2001; 24:158-162.
    22) Chamberlain JM, Altieri MA, & Futterman C: A prospective, randomized study comparing intramuscular midazolam with intravenous diazepam for the treatment of seizures in children. Ped Emerg Care 1997; 13:92-94.
    23) Chambers CD, Hernandez-Diaz S, VanMarter LJ, et al: Selective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newborn. N Engl J Med 2006; 354(6):579-587.
    24) Charlton NP , Lawrence DT , Brady WJ , et al: Termination of drug-induced torsades de pointes with overdrive pacing. Am J Emerg Med 2010; 28(1):95-102.
    25) Chin RF , Neville BG , Peckham C , et al: Treatment of community-onset, childhood convulsive status epilepticus: a prospective, population-based study. Lancet Neurol 2008; 7(8):696-703.
    26) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    27) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    28) Claassen JAHR & Gelissen HPMM: The serotonin syndrome (letter). N Eng J Med 2005; 352(23):2455.
    29) Criddle LM: Rhabdomyolysis. Pathophysiology, recognition, and management. Crit Care Nurse 2003; 23(6):14-22, 24-26, 28.
    30) Cuenca PJ, Holt KR, & Hoefle JD: Seizure secondary to citalopram overdose. J Emerg Med 2004; 26:177-181.
    31) Dagnone D, Matsui D, & Rieder MJ: Assessment of the palatability of vehicles for activated charcoal in pediatric volunteers. Pediatr Emerg Care 2002; 18:19-21.
    32) Dams R, Benijts TH, Lambert WE, et al: A fatal case of serotonin syndrome after combined moclobemide-citalopram intoxication. J Anal Toxicol 2001; 25:147-151.
    33) De Berardis D, Serroni N, Marini S, et al: Emerging mania following escitalopram withdrawal in a patient with unipolar depression managed with its reintroduction. J Psychiatr Pract 2014; 20(3):228-231.
    34) De Negri M & Baglietto MG: Treatment of status epilepticus in children. Paediatr Drugs 2001; 3(6):411-420.
    35) Dent LA, Brown WC, & Murney JD: Citalopram-induced priapism. Pharmacother 2002; 22:538-541.
    36) Drew BJ, Ackerman MJ, Funk M, et al: Prevention of torsade de pointes in hospital settings: a scientific statement from the American Heart Association and the American College of Cardiology Foundation. J Am Coll Cardiol 2010; 55(9):934-947.
    37) Drewes P, Thijssen I, Mengel H, et alDrewes P, Thijssen I, Mengel H, et al: A single-dose cross-over pharmacokinetic study comparing racemic citalopram (40 mg) with the S-enantiomer of citalopram (escitalopram, 20 mg) in healthy male subjects (poster), Annual Meeting of New Clinical Drug Evaluation Unit, Phoenix, AZ, 2001.
    38) Dubnov-Raz G, Juurlink DN, Fogelman R, et al: Antenatal use of selective serotonin-reuptake inhibitors and QT interval prolongation in newborns. Pediatrics 2008; 122(3):e710-e715.
    39) Duncan RA, Armstrong PAR, & Paterson B: Severe hypoglycaemia in citalopram overdose. Europ J Emerg Med 2008; 15(4):234-235.
    40) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    41) Engebretsen KM, Carson RH, & Wood JE: Cardiotoxicity and late onset seizures with citalopram overdose. J Emerg Med 2003; 25:163-166.
    42) Erdman AR & Dart RC: Rhabdomyolysis. In: Dart RC, Caravati EM, McGuigan MA, et al, eds. Medical Toxicology, 3rd ed. Lippincott Williams & Wilkins, Philadelphia, PA, 2004, pp 123-127.
    43) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    44) Fernando AT III & Schwader P: A case of citalopram withdrawal. J Clin Psychopharmacol 2000; 20:581-582.
    45) Finkelstein Y, Macdonald EM, Hollands S, et al: Antidepressants and the Risk of Seizures: a population-based, nested case control study (abstracts). Clin Toxicol (Phila) 2015; 53(7):673.
    46) Forrester MB: Escitalopram ingestions reported to Texas poison control centers, 2002-2005. Hum Exp Toxicol 2007; 26(6):473-482.
    47) Franssen EJ, Meijs V, Ettaher F, et al: Citalopram serum and milk levels in mother and infant during lactation. Ther Drug Monit 2006; 28(1):2-4.
    48) Friberg LE, Isbister GK, & Duffull SB: Pharmacokinetic-pharmacodynamic modelling of QT interval prolongation following citalopram overdoses. Br J Clin Pharmacol 2006; 61(2):177-190.
    49) Giang DW & McBride MC : Lorazepam versus diazepam for the treatment of status epilepticus. Pediatr Neurol 1988; 4(6):358-361.
    50) Gillman PK: Ecstasy, serotonin syndrome and the treatment of hyperpyrexia (letter). MJA 1997; 167:109-111.
    51) Gillman PK: Successful treatment of serotonin syndrome with chlorpromazine (letter). MJA 1996; 165:345.
    52) Glassman AH: Citalopram toxicity (letter). Citalopram toxicity. Lancet 1997; 350:818.
    53) Goldberg RJ & Huk M: Serotonin syndrome from trazodone and buspirone (letter). Psychosomatics 1992; 33:235-236.
    54) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    55) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    56) Graham PM: Successful treatment of the toxic serotonin syndrome with chlorpromazine (letter). Med J Australia 1997; 166:166-167.
    57) Gravem LF, Amthor KF, & Astrup C: A double-blind comparison of citalopram (Lu 10-171) and amitriptyline in depressed patients. Acta Psychiatr Scand 1987; 75:478-486.
    58) Grover J & Caravati EM: Right bundle branch block and delayed seizure associated with citalopram and fluoxetine ingestion (abstract). J Toxicol - Clin Toxicol 2001; 39:491.
    59) Grover S, Biswas P, Bhateja G, et al: Escitalopram-associated hyponatremia. Psychiatry Clin Neurosci 2007; 61(1):132-133.
    60) Grundemar L, Wohlfart B, & Lagerstedt C: Symptoms and signs of severe citalopram overdose. Lancet 1997; 349:1602.
    61) Guenther Skokan E, Junkins EP, & Corneli HM: Taste test: children rate flavoring agents used with activated charcoal. Arch Pediatr Adolesc Med 2001; 155:683-686.
    62) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    63) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    64) Heikkinen T, Ekblad U, kero P, et al: Citalopram in pregnancy and lactation. Clin Pharmacol Ther 2002; 72(2):184-191.
    65) Herrington L, Miller JJ, Geller RJ, et al: Pediatric ingestion of citalopram: What is a safe dose for home management?: 2015 Annual Meeting of the North American Congress of Clinical Toxicology (NACCT). Clin Toxicol 2015; 53(7):723-724.
    66) Homsi E, Barreiro MF, Orlando JM, et al: Prophylaxis of acute renal failure in patients with rhabdomyolysis. Ren Fail 1997; 19(2):283-288.
    67) Huerta-Alardin AL, Varon J, & Marik PE: Bench-to-bedside review: Rhabdomyolysis -- an overview for clinicians. Crit Care 2005; 9(2):158-169.
    68) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    69) Isbister GK, McGettigan P, & Dawson A: A fatal case of moclobemide-citalopram intoxication. J Anal Toxicol 2001 ; 25:716-717.
    70) Isbister GK, Friberg LE, & Duffull SB: Application of pharmacokinetic-pharmacodynamic modelling in management of QT abnormalities after citalopram overdose. Intensive Care Med 2006; 32(7):1060-1065.
    71) Isbister GK, Friberg LE, Stokes B, et al: Activated charcoal decreases the risk of QT prolongation after citalopram overdose. Ann Emerg Med 2007; 50(5):593-600, 600.
    72) Isbister GK, Prior FH, & Foy A: Citalopram-induced bradycardia and presyncope. Annal Pharmacother 2001; 35:1552-1555.
    73) Jensen PN, Olesen OV, Bertelsen A, et al: Citalopram and desmethylcitalopram concentrations in breast milk and in serum of mother and infant. Ther Drug Monit 1997; 19(2):236-239.
    74) Jimmink A, Caminada K, Hunfeld NG, et al: Clinical toxicology of citalopram after acute intoxication with the sole drug or in combination with other drugs: overview of 26 cases. Ther Drug Monit 2008; 30(3):365-371.
    75) Kelly CA, Dhaun N, Laing WJ, et al: Comparative toxicity of citalopram and the newer antidepressants after overdose. J Toxicol - Clin Toxicol 2004; 42:67-71.
    76) Kelly CA, Upex A, Spencer EP, et al: Adult respiratory distress syndrome and renal failure associated with citalopram overdose. Hum Exp Toxicol 2003; 22:103-105.
    77) Keren A, Tzivoni D, & Gavish D: Etiology, warning signs and therapy of torsade de pointes: a study of 10 patients. Circulation 1981; 64:1167-1174.
    78) Khan IA & Gowda RM: Novel therapeutics for treatment of long-QT syndrome and torsade de pointes. Int J Cardiol 2004; 95(1):1-6.
    79) Kline SS, Mauro LS, & Scala-Barnett DM: Serotonin syndrome versus neuroleptic malignant syndrome as a cause of death. Clin Pharmac 1989; 8:510-514.
    80) Klinger G, Frankenthal D, Merlob P, et al: Long-term outcome following selective serotonin reuptake inhibitor induced neonatal abstinence syndrome. J Perinatol 2011; 31(9):615-620.
    81) Kragh-Sorensen P, Overo KF, & Petersen OL: The kinetics of citalopram: single and multiple dose studies in man. Acta Pharmacol Toxicol 1981a; 48:53-60.
    82) Kragh-Sorensen P, Overo KF, Petersen OL, et al: The kinetics of citalopram: single and multiple dose studies in man. Acta Pharmacol Toxicol 1981; 48:53-60.
    83) Kraut JA & Madias NE: Metabolic acidosis: pathophysiology, diagnosis and management. Nat Rev Nephrol 2010; 6(5):274-285.
    84) Laine K, Heikkinen T, Ekblad U, et al: Effects of exposure to selective sertonin reuptake inhibitors during pregnancy on serotonergic symptoms in newborns and cord blood monoamine and prolactin concentrations. Arch Gen Psychiatry 2003; 60:720-726.
    85) Link MS, Berkow LC, Kudenchuk PJ, et al: Part 7: Adult Advanced Cardiovascular Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S444-S464.
    86) Liotier J & Coudore F: Drug monitoring of a case of citalopram overdosage. Drug Chem Toxicol 2011; 34(4):420-423.
    87) LoVecchio F, Watts D, Winchell J, et al: Outcomes after supratherapeutic escitalopram ingestions. J Emerg Med 2006; 30(1):17-19.
    88) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    89) Luchini D, Morabito G, & Centini F: Case report of a fatal intoxication by citalopram. Am J Forensic Med Pathol 2005; 26(4):352-354.
    90) Lung D, Yeh K, & Kiang C: Delayed, fatal cardiotoxicity associated with bupropion and citalopram overdose. J Clin Psychopharmacol 2012; 32(3):431-434.
    91) Lung DD, Wu AH, & Gerona RR: Cardiotoxicity in a citalopram and olanzapine overdose. J Emerg Med 2013; 45(4):554-558.
    92) Maddry JK, Breyer K, Cook KM, et al: Monomorphic ventricular tachycardia after intentional citalopram overdose. Am J Emerg Med 2013; 31(2):447-448.
    93) Malm H, Klaukka T, & Neuvonen PJ: Risks Associated With Selective Serotonin Reuptake Inhibitors in Pregnancy. Obstet Gynecol 2005; 106(6):1289-1296.
    94) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    95) Masullo LN, Miller MA, Baker SD, et al: Clinical course and toxicokinetic data following isolated citalopram overdose in an infant. Clin Toxicol (Phila) 2006; 44(2):165-168.
    96) Mills KC: Serotonin syndrome: a clinical update. Med Toxicol 1997; 13:763-783.
    97) Milne RJ & Goa KL: Citalopram: a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in depressive illness. Drugs 1991; 41:450-477.
    98) Mitchell WG: Status epilepticus and acute repetitive seizures in children, adolescents, and young adults: etiology, outcome, and treatment. Epilepsia 1996; 37(S1):S74-S80.
    99) Moyer B, Shrading W, & Burkhart KK: Celexa(R) for Celebrex(R)? A case of medication sample error. Int J Med Toxicol 2000; 3:7.
    100) Musshoff F, Schmidt P, & Madea B: Case report: Fatality caused by a combined trimipramine-citalopram intoxication. Forensic Sci International 1999; 106:125-131.
    101) Nakhai-Pour HR, Broy P, & Berard A: Use of antidepressants during pregnancy and the risk of spontaneous abortion. CMAJ 2010; Epub:Epub.
    102) Nelson LS, Erdman AR, Booze LL, et al: Selective serotonin reuptake inhibitor poisoning: An evidence-based consensus guideline for out-of-hospital management. Clin Toxicol (Phila) 2007; 45(4):315-332.
    103) Neumar RW , Otto CW , Link MS , et al: Part 8: adult advanced cardiovascular life support: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2010; 122(18 Suppl 3):S729-S767.
    104) Neuvonen PJ, Pohjola-Sintonen S, & Tacke U: Five fatal cases of serotonin syndrome after moclobemide-citalopram or moclobemide-clomipramine overdoses (letter). Lancet 1993; 342:1419.
    105) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    106) Nyth AL & Gottfries CG: The clinical efficacy of citalopram in treatment of emotional disturbances in dementia disorders. A nordic multicentre study. Br J Psychiatry 1990; 157:894-901.
    107) Nyth AL, Gottfries CG, & Lyby K: A controlled multicenter clinical study of citalopram and placebo in elderly depressed patients with and without concomitant dementia. Acta Psychiatr Scand 1992; 86:138-145.
    108) Ostrom M, Anders E, & Thorson J: Fatal overdose with citalopram (letter). Lancet 1996; 348:340.
    109) Overo KF, Toft B, & Christophersen L: Kinetics of citalopram in elderly patients. Psychopharmacology 1985; 86:253-257.
    110) Overo KF: Kinetics of citalopram in man; plasma levels in patients. Neuro-Psychopharmacol & Biol Psychiat 1982; 6:311-318.
    111) Overo KF: Preliminary studies of the kinetics of citalopram in man. Eur J Clin Pharmacol 1978; 14:69-73.
    112) Oyehaug E & Ostensen ET: High-performance liquid chromatographic determination of citalopram and four of its metabolites in plasma and urine samples from psychiatric patients. J Chromatography 1984; 308:199-298.
    113) Pedersen OL, Kragh-Sorensen P, & Bjerre M: Citalopram, a selective serotonin reuptake inhibitor: clinical antidepressive and long-term effect - a phase II study. Psychopharmacology 1982; 77:199-204.
    114) Personne M, Persson H, & Sjoberg G: Citalopram toxicity (letter). Lancet 1997; 350:519.
    115) Perticone F, Ceravolo R, & Cuccurullo O: Prolonged magnesium sulfate infusion in the treatment of ventricular tachycardia in acquired long QT syndrome. Clin Drug Inverst 1997; 13:229-236.
    116) Polderman KH: Acute renal failure and rhabdomyolysis. Int J Artif Organs 2004; 27(12):1030-1033.
    117) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    118) Power A: Citalopram in overdose may result in serious morbidity and death. BMJ 1998; 316:307-308.
    119) Product Information: ATIVAN(R) injection, lorazepam injection. Baxter Healthcare Corporation, Deerfield, IL, 2003.
    120) Product Information: CELEXA(R) oral tablets, solution, citalopram hydrobromide oral tablets, solution. Forest Pharmaceuticals,Inc, St.Louis, MO, 2008.
    121) Product Information: CELEXA(R) oral tablets, solution, citalopram hydrobromide oral tablets, solution. Forest Pharmaceuticals Inc, St. Louis, MO, 2009.
    122) Product Information: CITALOPRAM HYDROBROMIDE oral solution, citalopram hydrobromide oral solution. Citron Pharma LLC (per DailyMed), East Brunswick, NJ, 2015.
    123) Product Information: Celexa(R) oral tablets, citalopram hydrobromide oral tablets. Forest Pharmaceuticals, Inc. (per manufacturer), St. Louis, MO, 2014.
    124) Product Information: Celexa(R) oral tablets, oral solution, citalopram hydrobromide oral tablets, oral solution. Forest Pharmaceuticals, Inc. (per FDA), St. Louis, MO, 2012.
    125) Product Information: Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, isoproterenol HCl intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection. Hospira, Inc. (per FDA), Lake Forest, IL, 2013.
    126) Product Information: LEXAPRO(R) oral tablets, solution, escitalopram oxalate oral tablets, solution. Forest Pharmaceuticals,Inc., St. Louis, MO, 2008.
    127) Product Information: Lexapro(R) oral tablets, oral solution, escitalopram oxalate oral tablets, oral solution. Forest Pharmaceuticals, Inc. (per FDA), St. Louis, MO, 2012.
    128) Product Information: Lexapro(R) oral tablets, oral solution, escitalopram oxalate oral tablets, oral solution. Forest Pharmaceuticals, Inc. (per FDA), St. Louis, MO, 2014.
    129) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    130) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    131) Product Information: magnesium sulfate heptahydrate IV, IM injection, solution, magnesium sulfate heptahydrate IV, IM injection, solution. Hospira, Inc. (per DailyMed), Lake Forest, IL, 2009.
    132) Qureshi A, Wassmer E, Davies P, et al: Comparative audit of intravenous lorazepam and diazepam in the emergency treatment of convulsive status epilepticus in children. Seizure 2002; 11(3):141-144.
    133) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    134) Rampono J, Hackett LP, Kristensen JH, et al: Transfer of escitalopram and its metabolite demethylescitalopram into breastmilk. Br J Clin Pharmacol 2006; 62(3):316-322.
    135) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    136) Rothenhausler HB, Hoberl C, Ehrentrout S, et al: Suicide attempt by pure citalopram overdose causing long-lasting severe sinus bradycardia, hypotension and syncopes: successful therapy with a temporary pacemaker. Pharmacopsychiatry 2000; 33(4):150-152.
    137) Scharko AM & Schumacher J: Prolonged QTc interval in a 14-year-old girl with escitalopram overdose. J Child Adolesc Psychopharmacol 2008; 18(3):297-298.
    138) Schreffler SM, Marraffa JM, Stork CM, et al: Sodium channel blockade with QRS widening after an escitalopram overdose. Pediatr Emerg Care 2013; 29(9):998-1001.
    139) Scott R, Besag FMC, & Neville BGR: Buccal midazolam and rectal diazepam for treatment of prolonged seizures in childhood and adolescence: a randomized trial. Lancet 1999; 353:623-626.
    140) Segura LJ & Bravo B: Postmortem citalopram concentrations: Alone or along with other compounds. J Forensic Sci 2004; 49(4):814-819.
    141) Sivojelezova A, Shuhaiber S, Sarkissian L, et al: Citalopram use in pregnancy: prospective comparative evaluation of pregnancy and fetal outcome. Am J Obstet Gynecol 2005; 193(6):2004-2009.
    142) Smith WM & Gallagher JJ: "Les torsades de pointes": an unusual ventricular arrhythmia. Ann Intern Med 1980; 93:578-584.
    143) Snider RD: Case report: left bundle branch block - a rare complication of citalopram overdose. J South Carolina Med Assoc 2001; 97:380-382.
    144) Spigset O, Carleborg L, Ohman R, et al: Excretion of citalopram in breast milk. Br J Clin Pharmacol 1997; 44:295-298.
    145) Spiller HA & Rogers GC: Evaluation of administration of activated charcoal in the home. Pediatrics 2002; 108:E100.
    146) Sreenath TG, Gupta P, Sharma KK, et al: Lorazepam versus diazepam-phenytoin combination in the treatment of convulsive status epilepticus in children: A randomized controlled trial. Eur J Paediatr Neurol 2009; Epub:Epub.
    147) Sternbach H: The serotonin syndrome. Am J Psychiatr 1991; 148:705-713.
    148) Sztajnkryeer MD, Scaglione JM, & Goetz RJ: Bidirectional wide complex tachycardia after citalopram overdose (abstract). J Toxicol Clin Toxicol 2003; 41:509-510.
    149) Tarabar AF, Hoffman RS, & Nelson LS: Citalopram overdose: late presentation of Torsades de Pointes (TDP) with cardiac arrest (abstract). J Toxicol Clin Toxicol 2003; 41:676.
    150) Thakore S & Murphy N: The potential role of prehospital administration of activated charcoal. Emerg Med J 2002; 19:63-65.
    151) Unterecker S , Warrings B , Deckert J , et al: Correlation of QTc interval prolongation and serum level of citalopram after intoxication--a case report. Pharmacopsychiatry 2012; 45(1):30-34.
    152) Vale JA, Kulig K, American Academy of Clinical Toxicology, et al: Position paper: Gastric lavage. J Toxicol Clin Toxicol 2004; 42:933-943.
    153) Vale JA: Position Statement: gastric lavage. American Academy of Clinical Toxicology; European Association of Poisons Centres and Clinical Toxicologists. J Toxicol Clin Toxicol 1997; 35:711-719.
    154) Vanholder R, Sever MS, Erek E, et al: Rhabdomyolysis. J Am Soc Nephrol 2000; 11(8):1553-1561.
    155) Walter LA & Catenacci MH: Rhabdomyolysis. Hosp Physician 2008; 44(1):25-31.
    156) Wheless JW : Treatment of status epilepticus in children. Pediatr Ann 2004; 33(6):376-383.
    157) Wijnands-Kleukers APG, Zoelen GV, & de Meulenbelt VI: A prospective study on intoxications with citalopram (abstract). J Toxicol - Clin Toxicol 2002; 40:274-275.
    158) Yazdy MM, Mitchell AA, Louik C, et al: Use of selective serotonin-reuptake inhibitors during pregnancy and the risk of clubfoot. Epidemiology 2014; 25(6):859-865.
    159) Yu BN, Chen GL, He N, et al: Pharmacokinetics of citalopram in relation to genetic polymorphism of CYP2C19. Drug Metab Dispos 2003; 31(10):1255-1259.
    160) Yuksel FV, Tuzer V, & Goka E: Escitalopram intoxication. Eur Psychiatr 2005; 20:82-84.
    161) de Wilde J, Mertens C, & Overo KF: Citalopram versus mianserin: a controlled, double-blind trial in depressed patients. Acta Psychiatr Scand 1985; 72:89-96.