MOBILE VIEW  | 

ANTIDEPRESSANTS, TRICYCLIC

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Tricyclic antidepressants possess a 3-ring molecular structure. They inhibit the neuronal reuptake of norepinephrine and serotonin (5HT) in the CNS, and possess affinity for muscarinic and histamine H(1) receptors to varying degrees.

Specific Substances

    A) CONSTITUENTS OF THE GROUP
    1) Amitriptyline (synonym)
    2) ClomiPRAMINE (synonym)
    3) Desipramine (synonym)
    4) Dibenzepin (synonym)
    5) Doxepin (synonym)
    6) Dothiepin (synonym)
    7) Imipramine (synonym)
    8) Lofepramine (synonym)
    9) Nortriptyline (synonym)
    10) Protriptyline (synonym)
    11) Trimipramine (synonym)
    12) Tricyclic, antidepressants

Available Forms Sources

    A) FORMS
    1) In order to determine the clinical effects, identify the structural classification.
    GENERICTRADE NAMESTRUCTURAL CLASSI- FICATIONCNS TOXI- CITYCV TOXI- CITY
    AmitriptylineElavil(R), Amitid(R), Endep(R), Amitril(R)Tricyclic++++++++
    ClomiPRAMINEAnafranil(R)Tricyclic++++++++
    DesipramineNorpramin(R), Pertofrane(R)Tricyclic++++++++
    DoxepinAdapin(R), Sinequan(R)Tricyclic++++++++
    ImipramineTofranil(R), Presamine(R), SK-Pramine(R), Janimine(R)Tricyclic++++++++
    NortriptylineAventyl(R), Pamelor(R)Tricyclic++++++++
    ProtriptylineVivactil(R)Tricyclic++++++++
    TrimipramineSurmontil(R)Tricyclic++++++++

    2) COMBINATION PRODUCTS: Combination products containing tricyclic antidepressants include, but are not limited to, Limbitrol(R) (amitriptyline and chlordiazepoxide); Etrafon(R), Perphenyline(R), Triavil(R), Triptazine(R) (amitriptyline and perphenazine).
    B) USES
    1) Tricyclic antidepressants are used to treat a wide range of disorders such as depression, panic disorder, social phobia, bulimia, narcolepsy, attention deficit disorder, obsessive compulsive disorder, childhood enuresis, and chronic pain syndromes (Nyanda et al, 2000).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Tricyclic antidepressants are commonly used in the treatment of major depression, as well as for insomnia, pain relief in chronic pain syndromes, and have been evaluated for use in numerous other clinical settings.
    B) PHARMACOLOGY: Tricyclic antidepressants are structurally similar to the phenothiazines, and have actions at numerous receptor sites in the body, including anticholinergic, alpha-blocking activities, serotonin, norepinephrine and dopamine reuptake inhibition, anticholinergic activity, sodium and potassium channel blockade, and CNS and respiratory depression. They are highly bound to plasma proteins, as well as bound to intracellular sites. Dulling of depressive ideation may explain their antidepressant efficacy.
    C) TOXICOLOGY: Toxicities are mostly via exaggeration of pharmacologic activities, including CNS depression, seizurogenicity, sodium channel blockade, and alpha-adrenergic blockade.
    D) EPIDEMIOLOGY: Exposures to tricyclic antidepressants are very common and are a major cause of drug-related fatalities in the American Association of Poison Control Center (AAPCC) database.
    E) WITH THERAPEUTIC USE
    1) Therapeutic doses initially may cause drowsiness and difficulty concentrating. Hallucinations, excitement, and confusion may be seen at therapeutic doses. Anticholinergic side effects (dry mouth, blurred vision, urinary retention) may occur.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Drowsiness, sedation, tachycardia, hallucinoses, and other anticholinergic effects may be seen at lower doses.
    2) SEVERE TOXICITY: Coma, seizures, QRS prolongation with ventricular dysrhythmias, respiratory failure, and hypotension are the primary life threats. Slowed GI motility may result in retained GI tract drug, with recurrence of toxicity once initial effects resolve and the GI tract becomes active again.
    0.2.3) VITAL SIGNS
    A) Respiratory depression may occur rapidly after overdose. Both hyperthermia and hypothermia have been reported. Hypotension occurs with severe overdose. Tachycardia is a common anticholinergic and early sympathomimetic effect.
    0.2.20) REPRODUCTIVE
    A) Most tricyclic antidepressants are classified as FDA pregnancy category C or D. Tricyclic antidepressants cross the placental barrier, but the few reports of teratogenicity are insufficient to implicate them as teratogens. Third-trimester antipsychotic drug exposure has been associated with extrapyramidal and/or withdrawal symptoms in neonates. There are case reports of cognitive development impairment and other developmental issues. Tricyclic antidepressants are present in the milk of lactating women taking therapeutic doses.

Laboratory Monitoring

    A) Monitor cardiac rhythm and serial ECGs, serum electrolytes, renal function, hepatic enzymes, and CPK.
    B) Acid-base status should be followed with serial blood gases in severe toxicity when serum alkalinization is being performed or with sustained intubation.
    C) Urinalysis should be followed in those at risk for rhabdomyolysis. False positive urine TCA toxicology screens commonly occur with diphenhydramine, carbamazepine, cyclobenzaprine, quetiapine, and others.
    D) Serum tricyclic levels may be of use in drug-naive individuals and to help establish overdose in patients known to be taking tricyclics, but there is poor correlation between blood levels and clinical effects.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) In isolated exposures, toxicity occurs along a spectrum, and mild to moderate toxicity on presentation may progress to severe toxicity over minutes to hours. For patients with mild to moderate effects on presentation, activated charcoal should be considered in patients presenting less than 2 hours post-ingestion. Aggressive symptomatic and supportive care includes behavioral and temperature control, airway protection, blood pressure support, QRS monitoring and narrowing, and seizure control.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Activated charcoal should be considered in patients presenting less than 2 hours post-ingestion (protect airway first). Aggressive symptomatic and supportive care is essential, including behavioral and temperature control, airway protection, blood pressure support, QRS monitoring and narrowing, and seizure control. Consider intravenous lipid therapy early for patients with ventricular dysrhythmias or hypotension.
    C) DECONTAMINATION
    1) PREHOSPITAL: Prehospital induction of emesis is contraindicated. Prehospital administration of activated charcoal may be considered only if endotracheal intubation can be performed in order to protect the airway should CNS depression and/or seizures occur.
    2) HOSPITAL: Activated charcoal should be given if within 2 hours of exposure. The patient’s ability to protect the airway or the need for intubation should be considered. The role of gastric lavage is unclear, but should be considered for massive ingestions presenting within the first 60 minutes, in patients who can protect their airway or who have been intubated.
    D) AIRWAY MANAGEMENT
    1) Severe toxicity almost always requires intubation and ventilator support.
    E) ANTIDOTE
    1) There are no specific antidotes.
    F) SEIZURE
    1) Seizures are initially managed with benzodiazepines. Phenobarbital can be used for repetitive seizures, followed by propofol, and/or rapid sequence intubation and general anesthesia with continuous EEG monitoring.
    G) WIDE QRS COMPLEX
    1) QRS widening is managed by serum alkalinization with sodium bicarbonate and/or hyperventilation. Both approaches may be used, but sodium bicarbonate appears superior to hyperventilation alone and the increase in extracellular sodium may be related to its efficacy. Bolus administration of sodium bicarbonate allows for larger doses than continuous IV infusion and appears more effective in increasing the serum pH. The goal is a serum pH of 7.45 to 7.55 and pH should be closely monitored so as not to exceed 7.6. Severe QRS widening (>160 msec) not responsive to alkalinization may respond to a bolus of hypertonic saline. Consider infusion of lipid emulsion in patients with refractory dysrhythmias or hypotension.
    H) VENTRICULAR ARRHYTHMIA
    1) Ventricular dysrhythmias should be managed initially by serum alkalinization and, if unresponsive, with antiarrhythmics. Ventricular tachycardia and fibrillation may be treated with lidocaine (often ineffective), amiodarone (additive QTc prolongation), and/or D/C shock. Phenytoin may be used for dysrhythmias unresponsive to other measures. Magnesium, overdrive pacing and D/C shock may be used for Torsades de Pointe. Consider infusion of lipid emulsion in patients with refractory dysrhythmias or hypotension.
    I) FAT EMULSION
    1) Patients who develop significant cardiovascular toxicity may be treated with intravenous lipids. Administer 1.5 mL/kg of 20% lipid emulsion over 2 to 3 minutes as an IV bolus, followed by an infusion of 0.25 mL/kg/min. Evaluate the patient's response after 3 minutes at this infusion rate. The infusion rate may be decreased to 0.025 mL/kg/min (ie, 1/10 the initial rate) in patients with a significant response. This recommendation has been proposed because of possible adverse effects from very high cumulative rates of lipid infusion. Monitor blood pressure, heart rate, and other hemodynamic parameters every 15 minutes during the infusion. If there is an initial response to the bolus followed by the re-emergence of hemodynamic instability during the lowest-dose infusion, the infusion rate may be increased back to 0.25 mL/kg/min or, in severe cases, the bolus could be repeated. A maximum dose of 10 mL/kg has been recommended by some sources. Where possible, lipid resuscitation therapy should be terminated after 1 hour or less, if the patient's clinical status permits. In cases where the patient's stability is dependent on continued lipid infusion, longer treatment may be appropriate.
    J) TACHYCARDIA
    1) Tachycardia does not usually require treatment and the use of bradycardic agents, particularly beta-blockers, may result in hemodynamic collapse.
    K) HYPOTENSIVE EPISODE
    1) Hypotension should be managed with positioning, fluid expansion, and pressors if needed. Dopamine may be ineffective because of reuptake inhibition. Direct pressors (e.g. norepinephrine, epinephrine) may be more effective. Consider infusion of lipid emulsion in patients with refractory dysrhythmias or hypotension. Cardiovascular support (ECMO or cardiopulmonary bypass) may be required for patients unable to sustain oxygenation and/or perfusion.
    L) CENTRAL NERVOUS SYSTEM DEPRESSION
    1) CNS depression is managed with intubation and ventilator support.
    M) DELIRIUM
    1) Benzodiazepines are first line for behavioral control and for termination of seizures.
    N) BODY TEMPERATURE ABOVE REFERENCE RANGE
    1) Control agitation and seizures with benzodiazepines. Aggressive sedation, endotracheal intubation and mechanical ventilation are likely to be required in severe cases. Disrobe patient, apply moist sheets or keep skin damp and use fans to enhance evaporative cooling. Consider immersion in cold water in severe cases, but this may make resuscitation difficult.
    O) CONTRAINDICATED TREATMENT
    1) Contraindicated treatments include flumazenil and physostigmine.
    P) ENHANCED ELIMINATION
    1) There is no role for hemodialysis or other enhanced elimination techniques. Evidence of ongoing drug absorption or prolonged effects should prompt consideration of repeat doses of activated charcoal or whole bowel irrigation.
    Q) PATIENT DISPOSITION
    1) HOME CRITERIA: Asymptomatic patients with inadvertent single substance ingestions can be managed at home with follow-up via telephone if the dose is less than the maximum daily single therapeutic dose for age or the following mg/kg amounts (whichever is less):
    a) AMITRIPTYLINE: 5 mg/kg or less; Maximum daily dose 150 to 300 mg adults, 200 mg adolescents and elderly
    b) CLOMIPRAMINE: 5 mg/kg or less; Maximum daily dose 300 mg adults, 200 mg children 10 to 18 years
    c) DESIPRAMINE: 2.5 mg/kg or less; Maximum daily dose 300 mg adults, 150 mg adolescents and elderly
    d) DOXEPIN: 5 mg/kg or less; Maximum daily dose 300 mg adults
    e) DOXEPIN CREAM: 5 mg/kg or less
    f) IMIPRAMINE: 5 mg/kg or less; Maximum daily dose 300 mg adults; 100 mg adolescents and elderly
    g) NORTRIPTYLINE: 2.5 mg/kg or less; Maximum daily dose 150 mg adults, 50 mg adolescents and elderly
    h) PROTRIPTYLINE: 1 mg/kg or less; Maximum daily dose 60 mg adults; 20 mg adolescents and elderly
    i) TRIMIPRAMINE: 2.5 mg/kg or less; Maximum daily dose 200 mg adults, 100 mg adolescents and elderly
    2) OBSERVATION CRITERIA: All patients with intentional ingestions, or unintentional ingestions of unknown amounts, and all symptomatic patients (eg, weak, drowsy, dizzy, tremors, palpitations) should be referred to a healthcare facility; Patients with unintentional, single substance ingestions of more than the maximum single therapeutic dose (adjusted for age) or the following mg/kg ingestion (whichever is less) should be referred to a healthcare facility for observation:
    a) AMITRIPTYLINE: greater than 5 mg/kg; Maximum daily dose 150 to 300 mg adults, 200 mg adolescents and elderly
    b) CLOMIPRAMINE: greater than 5 mg/kg; Maximum daily dose 300 mg adults, 200 mg children 10 to 18 years
    c) DESIPRAMINE: greater than 2.5 mg/kg; Maximum daily dose 300 mg adults, 150 mg adolescents and elderly
    d) DOXEPIN: greater than 5 mg/kg; Maximum daily dose 300 mg adults
    e) DOXEPIN CREAM: greater than 5 mg/kg
    f) IMIPRAMINE: greater than 5 mg/kg; Maximum daily dose 300 mg adults; 100 mg adolescents and elderly
    g) NORTRIPTYLINE: greater than 2.5 mg/kg; Maximum daily dose 150 mg adults, 50 mg adolescents and elderly
    h) PROTRIPTYLINE: greater than 1 mg/kg; Maximum daily dose 60 mg adults; 20 mg adolescents and elderly
    i) TRIMIPRAMINE: greater than 2.5 mg/kg; Maximum daily dose 200 mg adults, 100 mg adolescents and elderly
    3) ADMISSION CRITERIA: Patients who remain symptomatic at 6 hours should be admitted to an intensive care unit. Serial monitoring of mental/respiratory status, acid-base status, and ECG should be performed. Patients who have resolved all clinical effects, have a normal QRS and QTc and are eating with normal bowel activity and no recurrent effects, may be discharged.
    4) CONSULT CRITERIA: All toxic exposures should be reported to the designated regional poison center. Severe tricyclic toxicity patients should have a consultation by a medical toxicologist.
    R) PITFALLS
    1) Pitfalls include the failure to appreciate the potential for rapid deterioration, failure to anticipate the need for cardiopulmonary support, and the potential for cyclic effects secondary to retained GI tract drug.
    S) PHARMACOKINETICS
    1) Absorption is usually rapid, with effects beginning within 1 hour of exposure, peak plasma levels within 1 to 2 hours, and peak clinical effects between 6 and 12 hours. Binding to serum proteins is high, typically greater than 90%, and the volume of distribution is large, between 7 and 78 L/kg. Elimination is hepatic, with some agents (amitriptyline, nortriptyline) having active metabolites. All tricyclic antidepressants undergo enterohepatic circulation.
    T) TOXICOKINETICS
    1) Absorption may be delayed or prolonged because of anticholinergic effects or the formation of bezoars. Patients who develop significant toxicity generally do so within 6 hours of overdose.
    U) PREDISPOSING CONDITIONS
    1) Combining a tricyclic antidepressant with a serotonin reuptake inhibitor may increase the plasma level of the tricyclic between 4- and 10-fold. The combination of a tricyclic antidepressant and another agent which increases CNS serotonin concentrations may precipitate serotonin syndrome.
    V) DIFFERENTIAL DIAGNOSIS
    1) Other agents with anticholinergic effects may produce a similar clinical appearance.

Range Of Toxicity

    A) TOXICITY: Ingestion of 10 to 20 mg/kg of most antidepressants constitutes a moderate to serious exposure where coma and cardiovascular symptoms are expected. Ingestion of either more than the maximal single dose (for age) or the following mg/kg dose (whichever is less) is considered potentially toxic: AMITRIPTYLINE: greater than 5 mg/kg. CLOMIPRAMINE: greater than 5 mg/kg. DESIPRAMINE: greater than 2.5 mg/kg. DOXEPIN: greater than 5 mg/kg. DOXEPIN CREAM: greater than 5 mg/kg. IMIPRAMINE: greater than 5 mg/kg. NORTRIPTYLINE: greater than 2.5 mg/kg. PROTRIPTYLINE: greater than 1 mg/kg. TRIMIPRAMINE: greater than 2.5 mg/kg . Fatalities have occurred in children following the ingestion of as little as 250 mg of amitriptyline or imipramine.
    B) THERAPEUTIC DOSE: ADULT DOSES include: Amitriptyline 75 to 300 mg/day in divided doses; desipramine 100 to 300 mg/day; doxepin 75 to 300 mg/day; imipramine 100 to 300 mg/day in divided doses; nortriptyline 75 to 150 mg/day in divided doses. PEDIATRIC DOSES include: Amitriptyline 0.1 to 2 mg/kg/dose, 10 to 20 mg/dose in adolescents; desipramine 1 to 5 mg/kg/day, 25 to 150 mg/day in adolescents; imipramine 1 to 4 mg/kg/day, 30 to 100 mg/day in adolescents; nortriptyline 1 to 3 mg/kg/day, 30 to 50 mg/day in adolescents.

Summary Of Exposure

    A) USES: Tricyclic antidepressants are commonly used in the treatment of major depression, as well as for insomnia, pain relief in chronic pain syndromes, and have been evaluated for use in numerous other clinical settings.
    B) PHARMACOLOGY: Tricyclic antidepressants are structurally similar to the phenothiazines, and have actions at numerous receptor sites in the body, including anticholinergic, alpha-blocking activities, serotonin, norepinephrine and dopamine reuptake inhibition, anticholinergic activity, sodium and potassium channel blockade, and CNS and respiratory depression. They are highly bound to plasma proteins, as well as bound to intracellular sites. Dulling of depressive ideation may explain their antidepressant efficacy.
    C) TOXICOLOGY: Toxicities are mostly via exaggeration of pharmacologic activities, including CNS depression, seizurogenicity, sodium channel blockade, and alpha-adrenergic blockade.
    D) EPIDEMIOLOGY: Exposures to tricyclic antidepressants are very common and are a major cause of drug-related fatalities in the American Association of Poison Control Center (AAPCC) database.
    E) WITH THERAPEUTIC USE
    1) Therapeutic doses initially may cause drowsiness and difficulty concentrating. Hallucinations, excitement, and confusion may be seen at therapeutic doses. Anticholinergic side effects (dry mouth, blurred vision, urinary retention) may occur.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Drowsiness, sedation, tachycardia, hallucinoses, and other anticholinergic effects may be seen at lower doses.
    2) SEVERE TOXICITY: Coma, seizures, QRS prolongation with ventricular dysrhythmias, respiratory failure, and hypotension are the primary life threats. Slowed GI motility may result in retained GI tract drug, with recurrence of toxicity once initial effects resolve and the GI tract becomes active again.

Vital Signs

    3.3.1) SUMMARY
    A) Respiratory depression may occur rapidly after overdose. Both hyperthermia and hypothermia have been reported. Hypotension occurs with severe overdose. Tachycardia is a common anticholinergic and early sympathomimetic effect.
    3.3.2) RESPIRATIONS
    A) Respiratory depression is common and may occur rapidly after overdose (Kulig, 1985; Meredith & Vale, 1985). Respiratory depression usually resolves within 24 hours, although patients with complications may require ventilatory support for longer periods (Roy et al, 1989).
    B) Severe hyperventilation occurred with therapeutic use of nortriptyline in one case (Sunderrajan et al,1985).
    3.3.3) TEMPERATURE
    A) HYPERTHERMIA
    1) Hyperthermia has been reported in overdose, usually in patients with recurrent seizures or increased muscular activity (Hantson et al, 1994; Tribble et al, 1989; Crome & Newman, 1979; Greenblatt et al, 1974; Fouron & Chicoine, 1971; Giles, 1963) .
    2) CASE REPORT: A 17-year-old man taking 4.5 mg/kg for one year had a single episode of hyperthermia following 2 hours of strenuous exercise. His core temperature rose to 108 degrees F (Squires, 1992).
    B) Mild hypothermia has been reported in overdose (Hulten & Heath, 1983; Crome & Newman, 1979; Woodhead, 1979) .
    3.3.4) BLOOD PRESSURE
    A) Hypotension occurs with severe overdose (Ellison & Pentel, 1989; Shannon et al, 1988). Orthostatic hypotension is very common during therapeutic use (Goldman et al, 1986; Mortensen, 1984) .
    B) Mild hypertension has been reported in overdose (Woodhead, 1979) and may occur early in the clinical course secondary to tricyclic-induced inhibition of norepinephrine reuptake. Mild hypertension soon after ingestion suggests drug effect and may be followed by hypotension and other toxic effects.
    3.3.5) PULSE
    A) Increased heart rate occurs with therapeutic use (Burckhardt et al, 1978). Sinus tachycardia may be a combined anticholinergic and inhibited norepinephrine reuptake effect, but is an insensitive marker for development of serious toxicity (Frommer et al, 1987).

Heent

    3.4.3) EYES
    A) Pupils may be dilated but usually respond to light; blurred vision may occur secondary to loss of accommodation reflexes (Chakrabarti et al, 2010; Frommer et al, 1987). Miosis may be seen with coma.
    B) Nystagmus may occur (Frommer et al, 1987; Pulst & Lombroso, 1983) .
    C) A retrospective review of amitriptyline poisoning cases in pediatric patients (mean age 4.6 +/- 2.9 years) reported the occurrence of mydriasis in 17.3% of patients (n=52). The mean dose ingested was 9.4 +/- 5.8 mg/kg (Olgun et al, 2009).
    3.4.6) THROAT
    A) Dry mouth may occur as an anticholinergic effect (Frommer et al, 1987).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) ELECTROCARDIOGRAM ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) ECG changes may include sinus tachycardia, prolonged PR interval, widening of the QRS complex, QTc prolongation, rightward shift in the axis of the terminal 40 milliseconds of the QRS complex, T-wave flattening or inversion, ST-segment depression, right bundle branch block, junctional rhythm, and atrioventricular block (Clark et al, 2015; Hon et al, 2015; Grover et al, 2012; Blaber et al, 2012; Kiberd & Minor, 2012; Frank & Kierdorf, 2000; Keller et al, 2000; Mehta & Alexandrou, 2000; James & Kearns, 1995; Caravati & Bossart, 1991; Foulke & Albertson, 1986) Neimann et al, 1986; (Goldberg et al, 1985; Fasoli & Glauser, 1981; Langou et al, 1980) .
    1) CASE REPORT: A 44-year-old woman developed abnormal atrial and ventricular repolarization resembling acute infarction following amitriptyline intoxication (serum amitriptyline 4880 ng/mL at presentation). In addition, ST-segment elevation with abnormally inflected T wave (mimicking subepicardial injury) was noted (Zakynthinos et al, 2000).
    2) CASE REPORT: A 41-year-old man developed symmetrical T wave repolarization abnormalities mimicking an acute anteroseptal myocardial infarction approximately 9 hours after ingesting 40 dothiepin pills (25 mg each). In addition, minimal broadening of the QRS was noted on admission. The authors suggested that the changes in conduction were caused by either the quinidine-like activity of dothiepin, or by an alteration in membrane permeability allowing differences in potassium concentrations between different areas of the myocardium, rather than by any myocardial ischemic damage (Steeds & Muthusamy, 2000).
    b) Buckley et al (2003) conducted a retrospective review to identify the utility of using ECG findings for determining subsequent risk of psychotropic drug-induced dysrhythmias The study compared 39 patients with significant dysrhythmias (supraventricular tachycardia, ventricular tachycardia, cardiac arrest) following either tricyclic antidepressant overdose or thioridazine overdose with 117 controls with matched overdose ingestions but without the presence of dysrhythmias. The ECG findings of QRS of at least 100 ms (82% vs 76%), a QTc interval greater than 500 (74% vs 50%) and an R/S ratio in aVR greater than 0.7 (28% vs 3%) were more common in the dysrhythmia group as compared to the controls, respectively. However, in general, the association was weak, as evidenced by the following positive (PPV) and negative (NPV) predictive value:
    1) QRS >/= 100 ms: PPV 7.5%; NPV 94.7%
    2) QTc >500: PPV 10%; NPV 96.2%
    3) R/S ratio in aVR >0.7: PPV 41.2%; NPV 94.7%
    c) CASE REPORT (CHILD): A 6-year-old girl developed seizures and ECG abnormalities after taking 300 mg (15 mg/kg) of amitriptyline every night for 1 month instead of the prescribed 30 mg nightly for treatment of insomnia secondary to ADHD. Initial ECG revealed sinus tachycardia with QRS widening, QTc interval prolongation, and right bundle branch block. Her total amitriptyline/nortriptyline concentration, obtained at hospital admission, was 1676 ng/mL (normal therapeutic, 50 to 300 ng/mL). With continued administration of sodium bicarbonate, the patient gradually recovered with resolution of ECG findings, and was subsequently discharged approximately 7 days post-admission (Clement et al, 2012).
    d) CASE REPORT: A 56-year-old woman was found unresponsive after ingesting an unknown amount of amitriptyline tablets. At presentation to the ED, 45 minutes later, the patient's Glasgow Coma Scale was 3, and she developed seizures and hypotension. An initial ECG demonstrated sinus bradycardia with bigeminal premature ventricular contractions, first degree AV block, widening of the QRS complex and a prolonged QTC. With supportive therapy, including intranasal midazolam, IV sodium bicarbonate, and norepinephrine for refractory hypotension, the patient gradually recovered and was discharged without sequelae (Clark et al, 2015).
    e) QRS WIDENING
    1) COMPLICATIONS: QRS duration of greater than 0.10 seconds has been associated with an increased incidence of seizures and a QRS duration of 0.16 seconds or longer has been associated with an increased incidence of ventricular dysrhythmias (Boehnert & Lovejoy, 1985).
    a) QRS duration of greater than 0.10 seconds has been associated with an increased incidence of serious toxicity including coma, need for intubation, hypotension, seizures, and dysrhythmias (Frank & Kierdorf, 2000; Caravati & Bossart, 1991; Foulke & Albertson, 1986; Hulten & Heath, 1983) .
    b) In a retrospective series of 45 children and adolescents with tricyclic antidepressant overdose, the mean QRS and QTc duration were significantly longer in patients who developed seizures than in those who did not (James & Kearns, 1995).
    2) DURATION: In 17 patients with a QRS interval greater than 0.10 seconds, presenting within 3 hours of tricyclic antidepressant overdose, the mean duration of QRS prolongation was 12 to 20 hours. No change in the duration of QRS prolongation was reported when sodium bicarbonate was administered to 7 of these patients (Shannon, 1992).
    3) PEDIATRIC: A retrospective review of amitriptyline poisoning cases in pediatric patients reported QRS widening of 100 ms or greater in 4 of 52 patients, with all 4 patients becoming comatose and 2 of the 4 patients developing seizures. In this small study, the positive predictive value of a QRS interval of 100 ms or longer was 100% for development of coma and 50% for development of seizures in pediatric patients following amitriptyline poisoning (Olgun et al, 2009).
    f) TERMINAL QRS AXIS
    1) Wolfe et al (1989) found that rightward terminal 40 millisecond (ms) frontal plane QRS vector was a more sensitive indicator of tricyclic antidepressant overdose than was QRS duration, but this finding was not useful in predicting complications or outcome (Wolfe et al, 1989).
    a) Mehta & Alexandrou (2000) found that more rightward terminal 40 ms frontal plane axis (130 to 270 degrees) is an ECG finding that correlated with TCAs overdose (Mehta & Alexandrou, 2000).
    2) DEFINITION: A T40-ms axis falling between 120 degrees and 270 degrees requires a negative T40-ms deflection (terminal S wave) in lead I and a positive T40-ms deflection (terminal R wave) in lead AVR.
    3) CASE SERIES: In one study, tricyclic overdose patients were 8.6 times more likely to have a T40-ms axis of more than 120 degrees than were nontricyclic overdose patients. However, 17% of the tricyclic antidepressant overdose patients in the study did not demonstrate this finding. Of these, 50% were either comatose, seizing, or hypotensive secondary to the tricyclic overdose (Wolfe et al, 1989).
    4) Sensitivity and specificity of the T40 axis was 29% and 83%, respectively, in another study (Lavoie et al, 1990).
    5) Liebelt et al (1995) compared the ability of the amplitude of the R wave in AVR, the R-wave/S-wave ratio in AVR, and the maximum limb lead QRS duration to predict the development of seizures and dysrhythmias in a nonconsecutive prospective cohort study of patients with tricyclic antidepressant overdose (Liebelt et al, 1995).
    a) An R wave in AVR of 3mm or more had a sensitivity of 81% and a specificity of 73% in predicting seizures or dysrhythmias, while a QRS duration of 100 milliseconds or more had a sensitivity of 82% and a specificity of 58%.
    b) The positive predictive value of an R wave in AVR of 3 mm or more, an R/S in AVR of 0.7 or more, and a QRS interval of 100 milliseconds or more had positive predictive values of 43%, 46% and 35% respectively in predicting seizures or dysrhythmias. The negative predictive value of all 3 parameters were similar (92% to 94%).
    6) CASE SERIES: Berkovitch et al (1995) compared the QRS duration, and terminal 40 millisecond axis in 35 children younger than 11 years with TCA ingestion and 35 controls. There was no difference in QRS duration or terminal 40 millisecond axis between children with TCA ingestion and controls. This finding did not change when only children who developed significant TCA toxicity were included (Berkovitch et al, 1995).
    a) There was a great deal of variability in the terminal 40 millisecond axis in both the overdose and control children, suggesting that this may not be useful in predicting significant TCA ingestion in children.
    7) ADULT: A retrospective chart review of patients with intentional tricyclic antidepressant overdose (n=100), demonstrated that, when using a multivariable logistic regression model, the terminal 40-ms frontal plane QRS vector (T40) was the only significant ECG parameter that could be used as a sole predictive parameter (OR 1.70; 95%CI 1.02 to 2.84; p=0.041) in determining the occurrence of serious clinical events (seizures, dysrhythmias, or death). (Eyer et al, 2009).
    8) PEDIATRIC CASE SERIES: A retrospective review of amitriptyline poisoning cases in pediatric patients reported the R wave in aVR of 3 mm or longer in 15 of 52 patients, with 4 patients developing seizures. However, none of the patients with an R wave in aVR that was shorter than 3 mm developed seizures. In this small study, the positive predictive value of an R wave in aVR of 3 mm or greater was 26.6% for the development of seizures, and the negative predictive value was 100% (Olgun et al, 2009).
    B) BRUGADA SYNDROME
    1) WITH POISONING/EXPOSURE
    a) The Type I Brugada electrocardiographic pattern, characterized by ECG findings of coved, downsloping ST-segment elevations of 2 mm or greater in the V1-V2 precordial leads, followed by T-wave inversions, has been reported following tricyclic antidepressant overdose. The Brugada ECG pattern may be accompanied by right bundle branch block, not associated with ischemia, electrolyte abnormalities, or structural heart defect (Akhtar & Goldschlager, 2006; Monteban-Kooistra et al, 2006).
    b) CASE REPORT: A 48-year-old man, with a past medical history of hypertension and noncardiac chest pain, ingested an unknown amount of nortriptyline, diazepam, zopiclone, and paroxetine and, approximately 40 hours postingestion, developed typical ECG changes of Brugada syndrome, characterized by J point elevation with downsloping ST-segment elevation in V1-V2 leads and T wave inversion. A serum tox screen was positive only for tricyclic antidepressants. The patient's ECG showed a normal PR interval, a QRS duration of 0.12 seconds and a QTc of 0.46 sec. His troponin I level remained within normal limits (0.9 mcg/L; normal 0 to 2 mcg/L). The patient's ECG abnormalities spontaneously resolved over the next 4 days and he was discharged to psychiatric care (Bigwood et al, 2005).
    C) CONDUCTION DISORDER OF THE HEART
    1) Sinus tachycardia is the most common dysrhythmia following tricyclic overdose (Magdalan et al, 2013; Caksen et al, 2006).
    a) Patients with more severe overdoses may develop supraventricular tachycardia, atrial fibrillation, atrioventricular block, multifocal PVCs, junctional rhythm, ventricular tachycardia, torsades de pointes, ventricular fibrillation and cardiopulmonary arrest (Clark et al, 2015; Blaber et al, 2012; Kiberd & Minor, 2012; Alter et al, 2001; Roberge & Krenzelok, 2001; Lappa et al, 2000; Zakynthinos et al, 2000; Zell-Kanter et al, 2000; Buckley et al, 1994; Peters et al, 1992; Goldberg et al, 1985; Kresse-Hermsdorf et al, 1985; Stern et al, 1985; Carpenter et al, 1982; Fasoli & Glauser, 1981; Langou et al, 1980; Freeman et al, 1969) . Bradycardia and atrial flutter have also been reported (Chakrabarti et al, 2010; Winrow, 1999).
    1) CASE REPORT/ADULT: Third degree atrioventricular block was induced by therapeutic doses of amitriptyline (75 milligrams daily for 4 weeks) in a 58-year-old woman who required temporary cardiac pacing. Amitriptyline was withdrawn and activated charcoal and cathartic were given. Sinus rhythm was restored 4 days later (Lappa et al, 2000).
    2) CASE REPORT/ADULT: Torsade de pointes tachycardia due to the long QT syndrome was reported in a 50-year-old woman after taking 60 doxepin tablets (6 grams); maximum serum level of doxepin was 2845 nmol/L 12 hours after admission. Within 30 minutes of treatment with physostigmine (a loading dose of 4 mg and a maintenance dose of 2 mg/hr for 36 hours), the patient's dysrhythmias completely ceased (Alter et al, 2001).
    3) CASE REPORT/CHILD: A 5-year-old girl developed stupor and tachycardia after being treated with a 30 gram tube of doxepin cream (1329 milligrams of doxepin) for an eczematous rash involving 50% of her body surface area (Toerne et al, 1997). Serum tricyclic antidepressant level was 60 nanograms/milliliter. Absorption was enhanced due to the eczema (diminished skin integrity).
    4) CASE REPORT/CHILD: A 2.5-year-old child, who lived with a parent chronically taking dosulepin, presented to the emergency department comatose with involuntary jerky movements. Examination revealed tachycardia (180 bpm), hypotension (80/50 mmHg), mydriasis, hyperreflexia, and an initial Glasgow Coma Scale score of 5. An initial ECG demonstrated a narrow complex tachycardia. Due to the persistence of his tachycardia, intravenous adenosine was administered to determine the mechanism of his dysrhythmia. A repeat ECG, post-adenosine administration, revealed atrial flutter, that spontaneously resolved within 10 hours post-presentation. With supportive care, the patient's condition continued to improve and he was discharged 2 days post-presentation. The patient's signs and symptoms and spontaneous recovery indicated possible ingestion of tricyclic antidepressants (TCA), which was confirmed with a urine tox screen that was positive for TCA (Chakrabarti et al, 2010).
    b) ONSET: Life-threatening dysrhythmias usually develop within 6 hours of overdose (Dziukas & Vohra, 1991).
    2) A retrospective review of amitriptyline poisoning cases in pediatric patients (mean age 4.6 +/- 2.9 years) reported the occurrence of tachycardia in 57.7% of patients (n=52). The mean dose ingested was 9.4 +/- 5.8 mg/kg (Olgun et al, 2009).
    3) DELAYED DYSRHYTHMIAS
    a) Several case reports have described patients who developed significant dysrhythmias, some of which were fatal, after initial recovery from a tricyclic antidepressant overdose. These patients had clinical evidence of significant tricyclic toxicity prior to the onset of delayed dysrhythmia, and most received inadequate gastrointestinal decontamination (McAlpine et al, 1986; Callaham, 1982; Sedal et al, 1972; Slovis et al, 1971; Barnes et al, 1968; Giles, 1963) .
    b) CASE REPORT/CHILD: A 13-year-old girl presented comatose ingesting an unknown amount of 150-mg amitriptyline tablets. GI decontamination was not performed. She developed pulseless wide-complex dysrhythmias approximately 19 hours after ingestion. Despite aggressive resuscitative efforts, the patient's rhythm degenerated to torsades de pointes. Approximately 30 minutes after initiation of resuscitation, 20% IV lipid emulsion therapy was started. The patient initially received 2 IV boluses of 1.5 mg/kg each, administered over a 3-minute period, with each bolus administered 5 minutes apart, followed by a continuous infusion of 0.25 mg/kg/minute for 30 minutes. Following administration of the second bolus, the patient's cardiac status improved with termination of her dysrhythmias (Levine et al, 2012)
    c) CASE SERIES: In 2 retrospective studies totalling 204 patients, none developed any significant dysrhythmia after maintaining a normal level of consciousness and a normal electrocardiogram for 24 hours (Goldberg et al, 1985; Pentel & Sioris, 1981) .
    d) RECOMMENDATION: In symptomatic patients adequate gastrointestinal decontamination including activated charcoal, and a 24 hour period of inpatient monitoring after normalization of the ECG and mental status is suggested as sufficient follow up provided there is no underlying cardiac disease (Pentel & Sioris, 1981).
    D) CARDIOMYOPATHY
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 54-year-old woman presented with hypertension and a decreased level of consciousness (Glasgow Coma Scale (GCS) score of 8) approximately 8 hours after intentionally ingesting 1000 mg nortriptyline. An ECG at admission revealed sinus tachycardia (114 bpm), and a wide QRS complex. Laboratory data indicated a normal creatine kinase level of 68 international units/L (normal less than 200) but an elevated troponin I level of 0.23 ng/mL (normal less than 0.06). Serum nortriptyline concentration was 858 ng/mL. After intravenous sodium bicarbonate administration, the patient's QRS widening reverted; however, approximately 44 hours postingestion, the patient complained of chest pain. A repeat ECG showed deeply negative T waves and an echocardiography indicated apical akinesia with mildly decreased systolic function. Cardiac catheterization demonstrated left ventricular apical ballooning, characteristic of "tako-tsubo" syndrome. According to repeat laboratory analysis, there appeared to be no increase in creatine kinase or troponin I levels. Following daily administration of 160 mg acetylsalicylic acid and 5 mg bisoprolol, the patient gradually recovered . On hospital day 8, a cardiac MRI demonstrated normal regional wall motion with no evidence of necrosis (De Roock et al, 2008).
    E) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Hypotension is common in severe tricyclic antidepressant overdose (Clark et al, 2015; Hon et al, 2015; Magdalan et al, 2013; Chakrabarti et al, 2010; Engels & Davidow, 2010; Roberge & Krenzelok, 2001; Frank & Kierdorf, 2000; Keller et al, 2000; Winrow, 1999; Toerne et al, 1997; Frommer et al, 1987; Biggs et al, 1977) . Hypotension has been associated with the development of dysrhythmia or pulmonary edema in patients with QRS interval greater than or equal to 0.10 sec, TCA level greater than 500 ng/mL, or grade IV coma.
    b) INCIDENCE: Twenty-two of 64 patients (34%) who presented with acute tricyclic overdose had systolic BP of less than 95 mmHg on admission (Shannon et al, 1988).
    c) ASSOCIATED COMPLICATIONS: In a prospective study of 64 tricyclic antidepressant overdose patients with QRS interval greater than or equal to 0.10 sec, TCA level greater than 500 ng/mL, or grade IV coma, patients with hypotension on admission were more likely to develop dysrhythmias or pulmonary edema than patients who were normotensive on admission (P<0.01) (Shannon et al, 1988).
    d) A retrospective review of amitriptyline poisoning cases in pediatric patients (mean age 4.6 +/- 2.9 years) reported the occurrence of hypotension in 21.2% of patients (n=52). The mean dose ingested was 9.4 +/- 5.8 mg/kg (Olgun et al, 2009).
    F) ORTHOSTATIC HYPOTENSION
    1) WITH THERAPEUTIC USE
    a) Orthostatic hypotension, which may develop as a result of peripheral alpha-receptor blockade (Frommer et al, 1987), is very common during initiation of tricyclic antidepressant therapy (Goldman et al, 1986; Mortensen, 1984) .
    G) HYPERTENSIVE EPISODE
    1) Mild hypertension has been reported in overdose (Woodhead, 1979) and may occur early in the clinical course secondary to tricyclic-induced inhibition of norepinephrine reuptake. Mild hypertension soon after ingestion suggests drug effect and may be followed by hypotension and other toxic effects.
    2) CASE SERIES (CHILDREN): Small, clinically benign increases in diastolic blood pressure, heart rate, and electrocardiographic conduction parameters were associated with desipramine treatment (5 mg/kg daily) in a study of 200 children and adolescents (Biederman, 1991).
    H) MYOCARDIAL INFARCTION
    1) WITH THERAPEUTIC USE
    a) Myocardial infarction has been reported with therapeutic use (Barefoot & Williams, 2000; Smith & Tyznik, 1987).
    b) CASE REPORT: One patient was described as having ECG findings suggestive of a transmural myocardial infarction that resolved when the patient's desipramine was discontinued; coronary angiography was normal (Smith & Tyznik, 1987).
    2) WITH POISONING/EXPOSURE
    a) Myocardial infarction has been reported following overdose (Arya et al, 2004; Cohen et al, 2000; Chamsi-Pasha & Barnes, 1988) .
    b) CASE REPORT: Elevation of creatine kinase (CK) and CK-MB fraction was described in a tricyclic overdose patient who subsequently had a normal coronary angiogram (Guthrie & Lott, 1986).
    c) CASE REPORT: A 22-year-old woman who ingested 300 mg of amitriptyline developed ECG changes and serial cardiac enzyme pattern consistent with an acute anteroseptal myocardial infarction approximately 26 hours postingestion. CK-MB elevation and septal hypokinesia on echocardiogram after an overdose of amitriptyline and diazepam. Coronary angiography was not done (Chamsi-Pasha & Barnes, 1988).
    d) Abnormal ventricular conduction following dothiepin overdose simulating acute myocardial infarction has been reported (Steeds & Muthusamy, 2000).
    e) CASE REPORT: A 33-year-old woman with persistent sinus tachycardia after ingesting 300 mg amitriptyline developed chest pain with diaphoresis approximately 40 hours after ingestion. An ECG showed ST-segment depression and T-wave inversion, and laboratory analysis revealed elevated cardiac enzyme levels (CK 698 units/liter, CK-MB 77.7 ng/mL, troponin I 27.5 ng/mL, myoglobin 80.2 ng/mL), consistent with acute myocardial infarction. Echocardiogram showed no wall motion abnormalities and single photon emission CT showed normal myocardial perfusion; angiography was not performed. The patient recovered with supportive therapy (Kiyan et al, 2006).
    I) ENDOCARDITIS
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: Thrombotic, nonbacterial endocarditis was found on autopsy in 3 patients who died after amitriptyline overdose (Lindstrom et al, 1977). All 3 patients developed thrombocytopenia and ARDS prior to death and had evidence of infarction of multiple organs (kidney, spleen, lung, heart) and emboli/thrombus formation at other sites (lung, radial, coronary and cerebral arteries).
    J) DRUG INTERACTION
    1) Patients who coingest neuroleptic drugs may have an increased incidence of conduction delays.
    2) CASE SERIES: Patients who had coingested a neuroleptic drug showed a higher prevalence of first-degree atrioventricular block (p<0.001), prolongation of the QRS duration (>0.10 second) (p<0.05), and a threefold increase in the prevalence of QTc prolongation (p<0.05) compared to those patients who ingested a TCA alone (Wilens et al, 1990).
    a) Amitriptyline was the TCA ingested in 40 of the 58 TCA alone overdoses. Perphenazine was the neuroleptic drug combined with amitriptyline in 8 of the 12 patients that coingested drugs.
    b) The mean tricyclic serum concentrations were not significantly different between the two groups.
    K) DEAD - SUDDEN DEATH
    1) WITH THERAPEUTIC USE
    a) CASE SERIES: Sudden death has been reported in four 8- to 9-year-old children during routine treatment with desipramine. Two of these children had a family history of cardiac problems (Riddle et al, 1993; Riddle et al, 1991; Tingelstad, 1991; Abramowicz, 1990) .
    b) Using the baseline rate of sudden death in this age group and the numbers of children taking desipramine, there does not appear to be an increased risk of sudden death in children taking therapeutic doses of desipramine (Biederman et al, 1995).
    c) CASE REPORT: Two other cases of sudden death were reported after therapy with tricyclic antidepressants. The first child, aged 9 years, died after 5 weeks of therapy with desipramine 50 mg twice daily. The second child collapsed after 8 months of imipramine therapy plus thioridazine; the imipramine dose had been increased to 150 mg at bedtime 2 months before death (Varley & McClellan, 1997).
    L) CARDIAC ARREST
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 37-year-old man with endogenous depression developed cardiac arrest, severe hypotension, and wide QRS complexes (230-260 ms) after ingestion of at least 5000 milligrams of doxepin. Although fluid load, alkalinization, hypertonic saline and high-dose vasoactive substances did not improve the patient's condition, he improved after treatment with hemoperfusion/hemodialysis (Frank & Kierdorf, 2000).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) ACUTE RESPIRATORY INSUFFICIENCY
    1) Respiratory depression is common with significant overdoses and may develop rapidly (Karaci et al, 2013; Roberge & Krenzelok, 2001; Keller et al, 2000; Kulig, 1985; Meredith & Vale, 1985).
    2) INCIDENCE: Mechanical ventilation was required in 76.8% of 82 patients in one study (Roy et al, 1989).
    B) PNEUMONITIS
    1) Aspiration is a common complication in severe overdoses.
    2) INCIDENCE: Aspiration pneumonitis developed in 10% to 32% of patients with severe TCA poisoning in 3 case series (Roy et al, 1989; Varnell et al, 1989; Shannon & Lovejoy, 1987) .
    3) CHARCOAL ASPIRATION: Of 72 patients who received activated charcoal by NG tube in the emergency department after endotracheal intubation, there were 18 in whom charcoal was recovered from the airway (Roy et al, 1989).
    C) ACUTE LUNG INJURY
    1) May occur in patients with severe TCA poisoning (Varnell et al, 1989).
    2) INCIDENCE: Developed in 8 of 56 (14%) patients with severe TCA toxicity in one series (Shannon & Lovejoy, 1987).
    3) CASE REPORT: Pulmonary edema, possibly a result of decreased left ventricular function, was reported in a woman who ingested 1920 mg dibenzepin delayed-release tablets. She recovered with supportive care including furosemide and mechanical ventilation (Wirtheim & Bloch, 1996).
    D) ADULT RESPIRATORY DISTRESS SYNDROME
    1) Adult respiratory distress syndrome may occur after severe TCA overdose Lipper & Gaynor, 1995; (Guharoy, 1994; Dale & Hole, 1993) Zuckerman & Conway, 1993; (Shannon & Lovejoy, 1987; Flaherty et al, 1986; Lindstrom et al, 1977; Marshall & Moore, 1973) .
    2) INCIDENCE: Of 56 patients with severe tricyclic antidepressant overdose, 5 (9%) developed clinical and radiographic evidence of ARDS (Varnell et al, 1989).
    E) HYPERVENTILATION
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Hyperventilation secondary to nortriptyline therapy was described in a 61-year-old man with end-stage renal disease. Symptoms recurred upon rechallenge with the drug and the patient required intubation, paralysis and mechanical ventilation twice for severe respiratory alkalosis (Sunderragan et al, 1985).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) CENTRAL NERVOUS SYSTEM DEFICIT
    1) CNS depression is common. Coma and seizures may ensue abruptly, but are usually short-lived.
    2) SIGNS/SYMPTOMS: Lethargy, disorientation, ataxia may progress to coma (Magdalan et al, 2013; Mutlu et al, 2011; Chakrabarti et al, 2010; Engels & Davidow, 2010; Roberge & Krenzelok, 2001; Keller et al, 2000; Frommer et al, 1987). Absent brainstem reflexes (absent corneal reflexes, dysconjugate eye movements, absent oculocephalic reflexes, and absent ocular vestibular reflexes), and abnormalities in electroencephalogram and brainstem auditory evoked response may also occur (Roberge & Krenzelok, 2001; White, 1988) Pulst & Lomboroso, 1983.
    3) ONSET: Coma and seizures may ensue abruptly, but usually develop within 6 hours of presentation (Zakynthinos et al, 2000; Frommer et al, 1987; Stern et al, 1985).
    4) DURATION: Coma is usually short-lived with most patients awake within 24 hours (Frommer et al, 1987); however, a 39-year-old woman experienced prolonged coma (5 days) and loss of brainstem reflexes following amitriptyline overdose (Roberge & Krenzelok, 2001).
    5) PROGNOSIS: A Glasgow Coma Scale of <8 or coma grade III or IV using the Matthew-Lawson scale is associated with an increased risk of complications such as dysrhythmias, hypotension, or the need for intubation and ventilation (Hulten et al, 1992; Emerman et al, 1987) .
    6) CASE REPORT: Topical administration of 30 grams of doxepin 5% cream in 24 hours for a generalized eczematous rash resulted in altered mental status in a 5-year-old girl. ECG revealed sinus tachycardia (HR 120 bpm) with a normal axis and a QRS duration of 72 ms. Serum concentrations of doxepin and desmethyldoxepin (major active metabolite) were 11.95 nanograms per milliliter (ng/mL) and 17.71 ng/mL, respectively. Eighteen hours following skin decontamination with soap and water, a full recovery was made and the patient was discharged (Zell-Kanter et al, 2000).
    7) A retrospective study of acute amitriptyline intoxication in children reported the development of lethargy in 20 of 44 patients (45.4%) (Caksen et al, 2006).
    8) A retrospective review of amitriptyline poisoning cases in pediatric patients (mean age 4.6 +/- 2.9 years) reported the occurrence of lethargy and coma in 76.9% and 48.1%, respectively, of patients (n=52). The mean dose ingested was 9.4 +/- 5.8 mg/kg (Olgun et al, 2009).
    9) CASE REPORT: Coma (Glasgow Coma Scale (GCS) score of 5) was reported in a 15-year-old girl following intentional ingestion of 22 mg/kg of amitriptyline. Plasmapheresis was performed for 4 hours and, at the end of the fourth hour, the patient became responsive with her GCS score improving to 13. She recovered uneventfully and was discharged on hospital day 5 without sequelae (Karaci et al, 2013).
    10) CASE REPORT: An 8-year-old girl presented to the emergency department lethargic with seizure-like movements of all extremities. Approximately 90 minutes prior to presentation, the patient was found unresponsive with "shaking movements". At presentation, An ECG demonstrated sinus tachycardia (157 beats/minute) with a QRS of 98 msec, and laboratory values were normal except for slight hypokalemia (3.2 mmol/L [reference range 3.5 to 5.5 mmol/L]). The patient was intubated, transferred to an intensive care setting approximately 5.5 hours post-presentation, and sedated. A repeat ECG demonstrated normal sinus rhythm with a QRS of 96 msec. An EEG indicated no evidence of seizure activity, and a MRI was negative for any structural lesions. A urine drug screen was positive for tricyclic antidepressants (TCA). The patient's sedation was discontinued and after 34 hours post-presentation, she was alert and was extubated. Following extubation, interview of the patient revealed that she had taken one 25-mg (0.8 mg/kg) amitriptyline tablet, prescribed to her mother for depression, prior to onset of signs and symptoms. A TCA level, obtained 18 hours post-presentation, revealed an amitriptyline concentration of 121 ng/mL (therapeutic range 50 to 300 ng/mL) and a nortriptyline concentration of 79 ng/mL (therapeutic range 70 to 170 ng/mL). With supportive care, the patient's mental status normalized and she was discharged approximately 48 hours post-presentation (Grover et al, 2012).
    11) CASE REPORT: An 18-month-old child developed facial twitching, up-rolling of eyes, and lost consciousness for 5 minutes after a suspected ingestion of approximately 7 25-mg imipramine tablets. Intravenous diazepam (2 mg) was administered due to a suspected seizure, and the symptoms resolved (Hon et al, 2015).
    12) CASE REPORT: A 56-year-old woman was found unresponsive after ingesting an unknown amount of amitriptyline tablets. At presentation to the ED, 45 minutes later, the patient's Glasgow Coma Scale was 3, and she developed seizures and hypotension. An initial ECG demonstrated sinus bradycardia with bigeminal premature ventricular contractions, first degree AV block, widening of the QRS complex and a prolonged QTC. With supportive therapy, including intranasal midazolam, IV sodium bicarbonate, and norepinephrine for refractory hypotension, the patient gradually recovered and was discharged without sequelae (Clark et al, 2015).
    B) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) Generalized seizures are a major complication that may result in hemodynamic instability, brain damage, hyperthermia, metabolic acidosis, rhabdomyolysis, and myoglobinuria if uncontrolled. Seizures generally occur within 8 hours of ingestion and are usually short lived; however status epilepticus has been reported.
    b) COMPLICATIONS: Abrupt hemodynamic deterioration has been reported following seizures (Engels & Davidow, 2010; Lipper et al, 1994; Taboulet P, Michard F & Muszynski J et al, 1994; Ellison & Pentel, 1989) . Generalized seizures may result in brain damage, hyperthermia, metabolic acidosis, rhabdomyolysis, and myoglobinuria if uncontrolled.
    c) ONSET: Patients who develop seizures after TCA overdose generally do so within 3 hours of ingestion (28 of 30 patients (93%) in one study and 10 of 10 in another) (Shannon, 1992; Ellison & Pentel, 1989). Onset may be very abrupt, with seizures developing in patients who are awake and alert (Ellison & Pentel, 1989). A single tonic-clonic seizure occurred approximately 16 hours after ingestion of 100 x 50-mg modified-release amitriptyline tablets (O'Connor et al, 2006).
    d) DELAYED-ONSET: A 13-year-old girl presented comatose ingesting an unknown amount of 150-mg amitriptyline tablets. GI decontamination was not performed. She experienced a generalized tonic-clonic seizure approximately 19 hours after ingesting an unknown amount of 150-mg amitriptyline tablets. The seizure continued intermittently for 1 hour and resolved with administration of IV lorazepam and phenobarbital (Levine et al, 2012).
    e) DURATION: Single seizures lasted 2 minutes or less in 57% (17 of 30) cyclic antidepressant overdose patients (serum drug concentration 639 to 9,530 ng/mL). Six patients had more than 2 seizures and one was in status epilepticus for 104 minutes (Ellison & Pentel, 1989). Seizures generally cease by 8 hours after ingestion (Shannon, 1992).
    f) STATUS EPILEPTICUS: Status epilepticus has been reported after overdose with doxepin, imipramine, clomiPRAMINE, desipramine and amitriptyline (Ellison & Pentel, 1989; Sawyer et al, 1984; Flechter et al, 1983; Walter & Kauffman, 1980) Sunshine & Yaffe, 1963.
    g) CASE REPORT: A 29-year-old woman had a generalized tonic-clonic seizure following chronic ingestion of greater than 800 mg amitriptyline per day for 3 years (Wohlreich & Welch, 1993).
    h) CASE REPORT: A 56-year-old woman was found unresponsive after ingesting an unknown amount of amitriptyline tablets. At presentation to the ED, 45 minutes later, the patient's Glasgow Coma Scale was 3, and she developed seizures and hypotension. An initial ECG demonstrated sinus bradycardia with bigeminal premature ventricular contractions, first degree AV block, widening of the QRS complex and a prolonged QTC. With supportive therapy, including intranasal midazolam, IV sodium bicarbonate, and norepinephrine for refractory hypotension, the patient gradually recovered and was discharged without sequelae (Clark et al, 2015).
    i) CASE REPORT (CHILD): Generalized tonic-clonic seizures were reported in a 6-year-old child who had been receiving 300 mg (15 mg/kg) amitriptyline every night for 1 month instead of the prescribed 30 mg nightly for treatment of insomnia secondary to ADHD (Clement et al, 2012).
    j) CASE REPORT (CHILD): Seizure-like movements and altered mental status were reported in an 8-year-old child who ingested one 25-mg (0.8 mg/kg) amitriptyline tablet (Grover et al, 2012).
    k) CASE REPORT (CHILD): An 18-month-old child developed facial twitching, up-rolling of eyes, and lost consciousness for 5 minutes after a suspected ingestion of approximately 7 25-mg imipramine tablets. Intravenous diazepam (2 mg) was administered due to a suspected seizure, and the symptoms resolved (Hon et al, 2015).
    l) CASE REPORT (MUNCHAUSEN-BY-PROXY): A 2-year-old boy who was initially diagnosed with idiopathic seizure disorder after presenting on multiple occasions with generalized tonic-clonic seizures was actually being given amitriptyline 1 to 2 times daily by the patient's grandmother (Mullins et al, 1999).
    1) In another Munchausen-by-proxy case, a child experienced right focal seizures which rapidly progressed to generalized seizures and coma after being given tricyclic antidepressants (the specific drug was not reported). In addition, the child experienced bradycardia, refractory hypotension, facial myoclonus, and a left parietooccipital cerebral infarction (Winrow, 1999).
    m) A retrospective study of acute amitriptyline intoxication in children reported the development of seizures in 11 of 44 patients (25%) (Caksen et al, 2006).
    n) A retrospective review of amitriptyline poisoning cases in pediatric patients (mean age 4.6 +/- 2.9 years) reported the occurrence of seizures in 7.7% of patients (n=52). The mean dose ingested was 9.4 +/- 5.8 mg/kg (Olgun et al, 2009).
    o) PROGNOSTIC INDICATOR: A retrospective case control study was conducted, involving 25 patients with tricyclic antidepressant (TCA) poisoning who did not survive and 72 TCA-poisoned patients who did survive. Ten of the 25 non-survived patients (40%) exhibited advanced ECG changes, including ventricular fibrillation, torsades de pointes, or asystole, as compared to the control group who did not show any advanced ECG changes. The primary causes of death were refractory hypotension (n=6), ventricular tachycardia leading to cardiac arrest (n=13), and seizure or status epilepticus followed by cardiac arrest (n=2 and n=4, respectively). A statistically significant difference occurred between the two groups with respect to the pulse rate and respiratory rate at presentation, the occurrence of seizures (before or after presentation, or both), the height of the R wave in a lead aVR, T40-ms frontal plane QRS axis, and the occurrence of premature ventricular contractions. However, further statistical analysis, using forward logistic regression, showed that only the occurrence of seizures after hospital presentation was a predictive indicator for death in these patients (odds ratio = 40.88; 95% confidence interval (CI) = 9.93 to 168.39; p<0.001) (Sanaei-Zadeh et al, 2011).
    C) NEUROPATHY
    1) Acute polyradiculopathy, thought to represent an acute toxic reaction to amitriptyline, has been reported (Leys et al, 1987; LeWitt & Forno, 1985; Casarino, 1977; Isaacs & Carlish, 1963; Nimmo Smith & Grieve, 1963) .
    2) SIGNS/SYMPTOMS: The syndrome consists of bulbar and truncal weakness, flaccid tetraplegia, diminished deep tendon reflexes, and painless paresthesias.
    3) DIAGNOSIS: The electromyogram shows signs of denervation and nerve conduction velocity is decreased.
    4) CASE REPORT: Painful neuropathy, preceded by bilateral, distal paresthesias, was reported in a 55-year-old woman taking desipramine and nortriptyline (Benazzi & Ciucci, 1997).
    D) CENTRAL STIMULANT ADVERSE REACTION
    1) CNS excitation may progress to acute organic brain syndrome in tricyclic antidepressant overdose (Frommer et al, 1987).
    E) EXTRAPYRAMIDAL DISEASE
    1) Ataxia, choreoathetosis, dysarthria, nystagmus, myoclonus, and tardive dyskinesia may occur.
    2) FINDINGS: Ataxia, dysarthria, nystagmus and tardive dyskinesia have been reported (Frommer et al, 1987; Gangat et al, 1986).
    3) CASE REPORTS (OVERDOSE): Choreoathetosis and myoclonus have been reported following imipramine and amitriptyline overdosage (Burks et al, 1974).
    4) CASE REPORT (THERAPEUTIC USE): Myoclonus was associated with therapeutic use of nortriptyline (75 mg/day) (Patterson, 1990).
    F) DELIRIUM
    1) Delirium has been reported in patients taking therapeutic doses of tricyclics.
    2) RISK FACTORS: TCA concentration in plasma, age, and female gender were all identified as risk factors for the development of delirium in one study of 36 cases and another study of 10 cases (Livingston et al, 1983) Preskorn & Jerkovich, 1990).
    3) ONSET: A prodrome (worsening of depression, development of psychosis) may precede the development of delirium by about 2 weeks (Kutcher & Shulman, 1985) Preskorn & Jerkovich, 1990).
    4) CASE REPORT (THERAPEUTIC USE): A 68-year-old woman developed significant delirium while taking 50 mg of desipramine per day; plasma desipramine level was 112 ng/mL and her delirium resolved upon discontinuation of the drug (Kutcher & Shulman, 1985).
    G) NEUROLEPTIC MALIGNANT SYNDROME
    1) Neuroleptic malignant syndrome (NMS) has been reported in patients taking therapeutic doses of TCAs and in patients taking combinations of tricyclic antidepressants and either monoamine oxidase inhibitors or other psychopharmaceutical agents.
    2) THERAPEUTIC USE
    a) NMS has been reported in patients receiving therapeutic doses of trimipramine and desipramine (Langlow & Alarcon, 1989) Baca & Martinelli, 1990.
    b) NMS may be caused by a central imbalance between dopamine and norepinephrine rather than by dopamine depletion alone (Baca & Martinelli, 1990).
    c) CASE REPORT: A 62-year-old man developed neuroleptic malignant syndrome possibly associated with nortriptyline use (dose and therapy duration not specified). He presented with a temperature of 107.1 degrees F, pulse 114, blood pressure 80/30 mmHg, and blood sugar 400. His creatine kinase and creatine kinase MB were 1046 IU/L and 2.3 IU/L, respectively. The patient developed disseminated intravascular coagulation and expired 2 days later (June et al, 1999).
    3) DRUG COMBINATIONS -
    a) NMS has been reported in several patients taking therapeutic doses of tricyclic antidepressants in combination with neuroleptics or monoamine oxidase inhibitors (Heyland & Sauve, 1991; Corrigan & Coulter, 1988; Horn et al, 1988; Merriam, 1987; Ansseau et al, 1986; Eiser et al, 1982) .
    1) Inhibition of amine reuptake by tricyclics combined with the sympathomimetic properties of MAOIs may predispose to NMS (Heyland & Sauve, 1991).
    2) It has been proposed that the combination of long term tricyclic antidepressant therapy and short term neuroleptic therapy may induce a hypodopaminergic state predisposing to NMS (Corrigan & Coulter, 1988).
    H) CEREBELLAR ATAXIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT/CHILD: A 4-year-old child presented with dysequilibrium, impaired speech, and fainting after a suspected ingestion of 20 amitriptyline tablets (unknown strength). At presentation, her Glasgow Coma Scale score was 9, blood pressure was 90/50 mmHg, and heart rate was 125 beats/min. Approximately 15 minutes after presentation, ECG changes developed, blood pressure decreased to 50/10 mmHg, and she experienced 2 tonic-clonic seizures. An initial brain CT was normal; however, a second brain CT and MRI showed bilateral hypodense edema in the cerebellar hemispheres and an increased signal and mass with diffusion restrictions, indicative of cerebellitis. A urine toxicology panel revealed an elevated tricyclic antidepressant concentration. Following symptomatic therapy and 2 courses of hemoperfusion, the patient gradually recovered and was discharged on hospital day 13 (Tatli et al, 2013).
    I) CEREBROVASCULAR ACCIDENT
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 42-year-old man presented to the emergency department after having been found unconscious. At presentation, Glasgow Coma Scale score was 9. Neurological examination revealed dysconjugate gaze, bilaterally unresponsive to visual threat, left arm weakness, and upper and lower limb drifts. A brain CT scan revealed a hyperdense basilar artery indicative of a thrombus, suggesting a diagnosis of acute basilar artery ischemic stroke. The patient completely recovered with a normal MRI and angiogram approximately 12 hours after receiving 0.9 mg/kg alteplase 3 hours after symptom-onset. Subsequent interview of the patient revealed that he had ingested an unknown quantity of amitriptyline tablets several hours prior to presentation, confirming a final diagnosis of amitriptyline toxicity (Birns et al, 2013).
    J) DRUG DEPENDENCE
    1) Tricyclic abuse for the euphoric effects is rare but has been associated with seizures and conduction defects (Wohlreich & Welch, 1993).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) DRUG-INDUCED ILEUS
    1) The anticholinergic action of the TCAs frequently results in decreased GI motility and delayed gastric emptying (Frommer et al, 1987), and may cause adynamic ileus (Milner & Hills, 1966).
    B) DRUG-INDUCED GASTROINTESTINAL DISTURBANCE
    1) Bowel ischemia has been reported.
    2) CASE REPORTS (OVERDOSE): Bowel ischemia occurred in 2 patients who overdosed on cyclic antidepressants. Resuscitative efforts (MAST trousers, norepinephrine infusion) may have contributed to tissue ischemia by decreasing mesenteric blood flow (Wallace, 1989).
    3) TOXIC MEGACOLON: A 35-year-old woman who ingested 4500 mg imipramine developed abdominal distension, with X-ray showing a dilated large bowel, interstitial edema, and free air. Upon exploratory laparotomy, perforation of the mid-transverse colon was observed and emergency colectomy performed. The authors suggested that the patient developed toxic megacolon due to anticholinergic effects of imipramine on the bowel (Ross et al, 1998)
    C) INTESTINAL OBSTRUCTION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 44-year-old man with no past history of altered bowel habit developed intestinal pseudoobstruction and spontaneous cecal perforation after an overdose of 750 mg of amitriptyline. Longstanding hydrocephalus may have contributed to this complication. (McMahon, 1989).
    D) DIFFUSE SPASM OF ESOPHAGUS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Diffuse esophageal spasm without lesions occurred in a 45-year-old man following ingestion of 8 grams of amitriptyline (Rinder et al, 1988).
    E) DISORDER OF PANCREAS
    1) ACUTE TOXICITY
    a) CASE REPORT: Acute pancreatitis leading to prolonged ileus has been reported in a 48-year-old woman following ingestion of 750 to 1500 mg of clomiPRAMINE. No other drugs or alcohol were found on a drug screen. Serum lipase level on day 3 was 350 IU, and rose to 2546 IU on day 5 with a corresponding serum amylase level of 112 IU. Abdominal studies showed no evidence of biliary tract, renal, or pancreatic disease. Total parenteral nutrition was required as the patient recovered over a 2 week period (Roberge et al, 1994).
    b) CASE REPORT: A 30-year-old woman who ingested 800 mg amitriptyline in a suicide attempt developed elevated pancreatic enzymes, with serum amylase and lipase of 823 IU/L and 1054 IU/L, respectively and no associated clinical symptoms. The pancreas appeared normal on ultrasonography. Pancreatic enzymes continued to increase, peaking on the fourth day, and then progressively decreased (Pezzilli et al, 1998).
    F) BEZOAR
    1) WITH POISONING/EXPOSURE
    a) CASE REPORTS: Bezoars developed in 2 patients following overdose ingestions of slow-release clomiPRAMINE. The first patient, a 28-year-old woman, intentionally ingested 60 75-mg tablets of slow-release clomiPRAMINE (total dose 4.5 grams). Following unsuccessful retrieval of tablets using gastric lavage and activated charcoal, a pharmacobezoar was detected with an abdominal radiograph, and was successfully removed endoscopically. The second patient, a 25-year-old woman, intentionally ingested at least 6 grams of slow-release clomiPRAMINE and 120 mg oxazepam, and subsequently developed sinus tachycardia and a decreased level of consciousness. Following an inability to pass a gastric tube through the esophagus for gastric lavage, a gastroscopy was performed. A esophageal pharmacobezoar was detected and was successfully removed with an endoscope, pliers, flushing, and suction. Both patients completely recovered without complications (Hojer & Personne, 2008).
    b) CASE REPORT: A 42-year-old woman presented comatose (Glasgow Coma Scale score of 5), hypotensive (90/50 mmHg), and tachycardic (120 bpm) approximately 14 hours after intentionally ingesting 60 75-mg sustained-release clomiPRAMINE tablets and 30 25-mg doxepin capsules. Gastric lavage was initiated, with no evidence of a tablet mass, followed by activated charcoal, and over the next 12 hours, the patient was responsive with normalization of blood pressure and heart rate. Approximately 10 hours later, the patient's condition deteriorated with development of respiratory insufficiency, seizures, severe hypotension, and coma. A chest radiograph showed a large round white mass and a CT scan confirmed the presence of a tablet mass in the gastric lumen, necessitating surgical removal. Following removal, crushing of the pharmacobezoar showed that it was comprised entirely from tablets, which had the appearance of clomiPRAMINE tablets. Despite removal of the bezoar, the patient's condition continued to deteriorate and, 32 hours later, she died from multi-organ failure (Magdalan et al, 2013).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) TOXIC HEPATITIS
    1) WITH THERAPEUTIC USE
    a) Drug induced hepatitis has been described in a 39-year-old woman, first from the tricyclic amineptine and then from clomiPRAMINE (Larrey et al, 1986).
    2) WITH POISONING/EXPOSURE
    a) Moderate elevations in hepatic enzymes developed in a patient with fatal amitriptyline overdose (Lindstrom et al, 1977).
    B) HEPATIC FAILURE
    1) WITH THERAPEUTIC USE
    a) Fulminant hepatic failure developed in an 82-year-old woman taking therapeutic doses of nortriptyline (Berkelhammer et al, 1995).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) RETENTION OF URINE
    1) Urinary retention may result from the anticholinergic action of the tricyclic antidepressants (McMahon, 1989; Frommer et al, 1987) .
    B) CRUSH SYNDROME
    1) Acute tubular necrosis may develop in patients with significant rhabdomyolysis or hypotension (Greenblatt et al, 1974).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 44-year-old woman developed abnormal atrial and ventricular repolarization resembling acute infarction following amitriptyline intoxication (serum amitriptyline 4880 ng/mL at presentation). In addition, a slight metabolic acidosis was observed (Zakynthinos et al, 2000).
    b) MODIFIED-RELEASE PREPARATION: A 40-year-old woman experienced metabolic acidosis up to 98 hours following ingestion of 100 x 50-mg modified-release amitriptyline, 13 x 50-mg clomiPRAMINE, and 10 x 20-mg fluoxetine. Her initial serum amitriptyline concentration, obtained 17.5 hours postingestion, was 1100 ng/mL and her peak serum amitriptyline concentration, obtained 42 hours postingestion, was 2100 ng/mL. She gradually recovered following supportive therapy and was hospital discharged approximately 9 days postingestion (O'Connor et al, 2006).
    c) CASE REPORT: Metabolic acidosis, hypotension, and ECG abnormalities were reported in an 18-month-old child following a suspected ingestion of 7 25-mg imipramine tablets. The patient gradually recovered following supportive therapy, including IV sodium bicarbonate and dopamine infusions (Hon et al, 2015).
    2) Significant metabolic acidosis may develop in patients with prolonged seizures or hypotension (Frommer et al, 1987).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) THROMBOCYTOPENIC DISORDER
    1) Thrombocytopenia and thrombotic, nonbacterial endocarditis developed in 3 women with fatal amitriptyline overdose (Lindstrom et al, 1977).
    2) Thrombocytopenia, probably mediated by anti-desipramine antibodies, was reported in 2 patients taking therapeutic doses of imipramine and desipramine (Rachmilewitz et al, 1968).
    3) CASE REPORT: A 68-year-old woman developed thrombocytopenia (platelet count of 31,000//mm(3)) and a pruritic erythematous rash while on amitriptyline 60 mg and flunitrazepam 2 mg for 4 months. She was treated with prednisone 40 mg daily and fluocinonide cream. A patch and photopatch testing were done with amitriptyline and flunitrazepam and both were negative. Amitriptyline 10 mg was restarted orally and after 2 days the rash reappeared and thrombocytopenia developed (Taniguchi & Hamada, 1996).
    B) DISSEMINATED INTRAVASCULAR COAGULATION
    1) Low grade DIC has been reported in patients with severe overdose and in a patient who developed profound hyperthermia with therapeutic use (Squires, 1992; Greenblatt et al, 1974)
    C) AGRANULOCYTOSIS
    1) CASE REPORT: Agranulocytosis occurred in a 49-year-old woman 1 month after starting on clomiPRAMINE (Gravenor et al, 1986).
    2) CASE REPORT: Agranulocytosis developed in a 64-year-old man 5 weeks after starting imipramine therapy (Rothenberg & Hall, 1960).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) DISORDER OF SKIN
    1) CASE REPORT: A 46-year-old woman developed a vesicular eruption on her trunk 5 days after beginning amitriptyline for her fibromyalgia (Garcia-Doval et al, 1999). The vesicular lesions appeared to be grouped to form annular and arciform patterns. Amitriptyline was discontinued and the lesions resolved over several days. Upon rechallenge with amitriptyline, she developed lesions after 2 days.
    2) CASE REPORT: A 37-year-old man developed blistering secondary to his amitriptyline which required skin grafting (Fogarty & Khan, 1999). He ingested a toxic dose of amitriptyline 48 hours prior to admission to the hospital. His liver function tests were elevated. The blisters were treated with silver sulfadiazine dressings for 2 weeks. Surgical excision and split skin grafting of necrotic areas on the inner arm and chest wall were eventually required.
    B) ERUPTION
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 68-year-old woman developed thrombocytopenia (platelet count of 31,000//mm(3)) and a pruritic erythematous rash while on amitriptyline 60 mg and flunitrazepam 2 mg for 4 months. She was treated with prednisone 40 mg daily and fluocinonide cream. A patch and photopatch testing were done with amitriptyline and flunitrazepam and both were negative. Amitriptyline 10 mg was restarted orally and after 2 days the rash reappeared and thrombocytopenia developed (Taniguchi & Hamada, 1996).
    C) ACNE
    1) Severe acne-type lesions have been reported in 5 patients following chronic self administration of high doses of amineptine. In 4 cases, the severity of the cutaneous lesions seemed to be dose-related (Vexiau et al, 1988).
    D) PHOTOSENSITIVITY
    1) CASE REPORT: Severe photodermatitis was reported in an adult therapeutically dosed with clomiPRAMINE and carbamazepine. Patch testing revealed a contact allergy to carbamazepine and a photo and contact allergy to clomiPRAMINE (Ljunggren & Bojs, 1991).
    E) DISCOLORATION OF SKIN
    1) CHRONIC TOXICITY
    a) CASE REPORT: A slate-gray discoloration of sun exposed skin developed in a woman taking desipramine (Steele & Ashby, 1993).
    b) CASE SERIES: Ming et al (1999) reported 4 cases of golden-brown or slate-gray skin hyperpigmentation on the face, arms, and backs of the hands of women taking imipramine for at least 2 years. In 2 patients, discontinuation of imipramine resulted in nearly complete clearing of hyperpigmentation (Ming et al, 1999).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) RHABDOMYOLYSIS
    1) Mild elevations in CPK are common after tricyclic overdose (Shannon, 1989). More severe elevations may develop in patients with prolonged seizures or coma (Lindstrom et al, 1977; Greenblatt et al, 1974) .

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) PHEOCHROMOCYTOMA
    1) A 53-year-old man developed pheochromocytoma crisis after taking clomiPRAMINE 25 mg/day for 3 weeks. The patient presented with confusion, weakness, hypertension (200/120 mm Hg), tachycardia (140 bpm), and fever. Ultrasound revealed a suprarenal mass; plasma norepinephrine and epinephrine levels were grossly elevated. The authors implicated clomiPRAMINE in causing hyperactivity of the preexisting tumor (Korzets et al, 1997).
    2) An 8-year-old girl developed symptoms consistent with a pheochromocytoma (abdominal discomfort, diaphoresis, and tachycardia) 5 hours after taking her first dose of imipramine 25 mg/day for nocturnal enuresis. The authors suggest that patients with classic symptoms of a pheochromocytoma after ingestion of medications that potentiate catecholamines should be suspected of having a latent pheochromocytoma (Brown et al, 2001).
    B) HYPERGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) A retrospective study of acute amitriptyline intoxication in children reported the development of hyperglycemia in 14 of 44 patients (31.8%) (Caksen et al, 2006).
    b) A retrospective review of amitriptyline poisoning cases in pediatric patients (mean age 4.6 +/- 2.9 years) reported the occurrence of hyperglycemia in 21.2% of patients (n=52). The mean dose ingested was 9.4 +/- 5.8 mg/kg (Olgun et al, 2009).

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) ACUTE ALLERGIC REACTION
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Three weeks after starting amitriptyline 25 mg twice daily for mild depression, a 24-year-old woman developed a pruritic morbilliform rash on the trunk. She was admitted to the hospital with fever and elevated pulse and respiration rates. Serum liver enzymes were elevated, and eosinophils were highly elevated. Her condition was diagnosed as hypersensitivity syndrome, a specific severe idiosyncratic drug reaction. Amitriptyline was withdrawn, and 5 days later the patient was afebrile. One month later, skin lesions and blood tests had normalized (Milionis et al, 2000).

Reproductive

    3.20.1) SUMMARY
    A) Most tricyclic antidepressants are classified as FDA pregnancy category C or D. Tricyclic antidepressants cross the placental barrier, but the few reports of teratogenicity are insufficient to implicate them as teratogens. Third-trimester antipsychotic drug exposure has been associated with extrapyramidal and/or withdrawal symptoms in neonates. There are case reports of cognitive development impairment and other developmental issues. Tricyclic antidepressants are present in the milk of lactating women taking therapeutic doses.
    3.20.2) TERATOGENICITY
    A) PLACENTAL BARRIER
    1) Tricyclic antidepressants cross the placental barrier, but the few reports of teratogenicity are insufficient to implicate them as teratogens.
    B) CONGENITAL ANOMALY
    1) AMITRIPTYLINE/PERPHENAZINE: A mother ingested a combination of amitriptyline and perphenazine in a suicide gesture at 8 days gestation. The infant was born with multiple congenital defects such as microcephaly, "cotton- like" hair with pronounced shedding, cleft palate, micrognathia, ambiguous genitalia, and dermal ridges (Wertelecki et al, 1980).
    2) DOXEPIN: CASE REPORT: Poor sucking and swallowing, muscle hypotonia, drowsiness, vomiting, and jaundice occurred in a neonate whose mother used doxepin in her third trimester and during the postpartum period. The doxepin dose had been 75 mg/day, but was tapered in the last weeks of pregnancy and was 35 mg/day at parturition. The amount of doxepin and desmethyldoxepin (active metabolite) ingested by the nursing infant was 10 to 20 mcg/kg/day (2.5% of the weight-adjusted dose of the mother) (Frey et al, 1999).
    C) LACK OF EFFECT
    1) CASE SERIES: No congenital abnormalities were found in infants of 9 women who were taking tricyclics therapeutically during the first 60 days of their pregnancies (Misri & Sivertz, 1991).
    2) CLOMIPRAMINE: CASE SERIES: In four cases involving maternal use of clomiPRAMINE 75 to 250 mg daily during pregnancy, the infants were born at term without evidence of congenital malformation. Two of the neonates exhibited mild hypotonia or transient tachypnea during the first 2 to 7 days following delivery (Schimmell et al, 1991).
    D) ANIMAL STUDIES
    1) DOXEPIN: RATS: Increased rates of fetal structural abnormalities and decreased fetal body weight were reported at doses of 100 mg/kg/day or greater when pregnant rats were given oral doxepin (30, 100 and 150 mg/kg/day) during the period of organogenesis. In rats, the AUC at the no-effect dose for embryo-fetal developmental toxicity is 30 mg/kg/day (approximately 6 and 3 times the plasma AUCs for doxepin and nordoxepin at the maximum recommended human dose (MRHD)) (Prod Info SILENOR(R) oral tablets, 2010).
    3.20.3) EFFECTS IN PREGNANCY
    A) EXTRAPYRAMIDAL AND/OR WITHDRAWAL SYNDROME
    1) Maternal use of antipsychotic drugs during the third trimester of pregnancy has been associated with an increased risk of neonatal extrapyramidal and/or withdrawal symptoms (eg, agitation, hypertonia, hypotonia, tremor, somnolence, respiratory distress, and feeding disorder) following delivery. Severity of these adverse effects have ranged from cases that are self-limiting to cases that required prolonged periods of hospitalization and ICU care (Prod Info perphenazine and amitriptyline HCl oral tablet, 2010).
    2) IMIPRAMINE: CASE REPORT: A woman with agoraphobia was treated with imipramine (400 mg/day) throughout her pregnancy and delivered a female baby who exhibited withdrawal effects of imipramine characterized by rapid breathing, irritability, restlessness, and insomnia for one month (Shrand, 1982).
    3) CLOMIPRAMINE: CASE REPORT: A term male infant born to a mother treated with clomiPRAMINE 125 mg daily throughout pregnancy presented with low Apgar scores and developed jitteriness, respiratory distress, and hypotonia which resolved spontaneously at 6 days of age. Clinically significant clomiPRAMINE plasma concentrations were detected in the infant the first few days following delivery. The elimination half-life of clomiPRAMINE in the neonate was 92.8 hours. At 7 days of age, breastfeeding was begun and the infant remained asymptomatic despite continued maternal use of clomiPRAMINE. The gradual disappearance of symptoms correlated with the decline in clomiPRAMINE plasma concentrations, suggesting toxicity rather than a withdrawal syndrome (Schimmell et al, 1991).
    4) NORTRIPTYLINE: CASE REPORT: A term male infant born to a mother taking 100 mg of nortriptyline a day (last dose 245 mg 12 hours prior to delivery) developed lethargy at 10 hours of age and urinary retention which resolved at 40 hours of life (Shearer et al, 1972).
    5) NORTRIPTYLINE: CASE REPORT: A term male infant delivered 20 hours after maternal nortriptyline overdose developed decreased muscle tone, sedation, decreased sensitivity to pain, tachycardia (160), widened and split QRS complexes (0.1 seconds) (Sjoqvist et al, 1972). The child appeared clinically normal 40 hours after birth and the ECG was considered normal five days after birth. Nortriptyline half life in the infant was 56 hours.
    B) COGNITIVE DEVELOPMENT
    1) Based on data collected through the Mother Risk Program for a very small number of patients, infants who were exposed to either fluoxetine or tricyclic antidepressants throughout gestation and born to mothers with uncontrolled depressive symptoms showed lower cognitive and language achievements than those born to mothers who were well-controlled (Nulman et al, 2002).
    C) PREGNANCY CATEGORY
    1) The following tricyclic antidepressants have been classified as FDA pregnancy category D (Briggs et al, 1998):
    1) AMITRIPTYLINE
    2) DOTHIEPIN
    3) IMIPRAMINE
    4) NORTRIPTYLINE
    2) The following tricyclic antidepressants have been classified as FDA pregnancy category C (Briggs et al, 1998):
    1) CLOMIPRAMINE HYDROCHLORIDE
    2) DESIPRAMINE
    3) DOXEPIN (Prod Info SILENOR(R) oral tablets, 2010)
    4) PROTRIPTYLINE
    D) LACK OF EFFECT
    1) DOXEPIN: Based on data collected through the Mother Risk Program for a very small number of patients, there appear to be no differences in cognitive function, temperament, and general behavior in children exposed to doxepin throughout gestation as compared with controls (Nulman et al, 2002).
    E) ANIMAL STUDIES
    1) DOXEPIN: RATS: Reduced pup survival and transient growth delay were observed when rats were given oral doxepin 100 mg/kg/day during pregnancy and lactation periods. In this rat study, the AUC at the no-effect dose for embryo-fetal developmental toxicity is 30 mg/kg/day (approximately 3 and 2 times the plasma AUCs for doxepin and nordoxepin at the MRHD) (Prod Info SILENOR(R) oral tablets, 2010).
    2) DOXEPIN: RABBITS: Decreased fetal body weights in the absence of maternal toxicity were reported when oral doxepin doses of 60 mg/kg/day where given to pregnant rabbits during the period of organogenesis. In rabbits, the AUC at the no-effect dose for embryo-fetal developmental toxicity is 30 mg/kg/day (approximately 6 and 18 times the plasma AUCs for doxepin and nordoxepin at the MRHD)(Prod Info SILENOR(R) oral tablets, 2010).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) Tricyclic antidepressants are present in the milk of lactating women taking therapeutic doses, but the amounts have been insufficient to cause immediate symptoms.
    2) AMITRIPTYLINE/PERPHENAZINE: Although the effects on the nursing infant from exposure to the drug have not been determined, the American Academy of Pediatrics considers it to be of concern, especially when given to mothers for long periods of time (Anon, 2001). Existing data indicate that very little amitriptyline and nortriptyline (active metabolite) would be ingested by a breastfeeding infant and that maternal use of amitriptyline during breastfeeding is probably safe. However, the prolonged half-life of nortriptyline in newborns suggests that some infants may be at risk of accumulation. If the patient elects to breastfeed during amitriptyline therapy, the infant should be monitored for adverse effects. The manufacturer of the combination amitriptyline/perphenazine does not recommend its use in nursing women (Prod Info perphenazine and amitriptyline HCl oral tablet, 2010).
    3) DESIPRAMINE: CASE REPORT: Measurements of desipramine and metabolite (2-hydroxydesipramine) were obtained in both plasma and milk of a nursing mother. On day 7 the desipramine and metabolite plasma levels were 257 ng/mL and 234 ng/mL respectively. Corresponding breast milk levels were 316 ng/mL and 381 ng/mL. The tricyclic antidepressant was slightly concentrated in breast milk compared to plasma, however the concentrations of desipramine and metabolite were too small to be detected in the baby's plasma (Stancer & Reed, 1986).
    4) CLOMIPRAMINE: CASE REPORT: A term male infant born to a mother treated with clomiPRAMINE 125 mg daily throughout pregnancy demonstrated symptoms of clomiPRAMINE toxicity that resolved spontaneously in 6 days.
    a) The infant remained asymptomatic despite initiation of breastfeeding at 7 days of age. A milk to plasma ratio of 0.8 to 1.6 was calculated. Maternal and neonatal plasma levels drawn 10, 14, and 35 days after birth demonstrated that the infant achieved approximately 6% of maternal clomiPRAMINE concentrations through nursing (Schimmell et al, 1991).
    5) DOXEPIN: CASE REPORT: Poor sucking and swallowing, muscle hypotonia, drowsiness, vomiting, and jaundice occurred in a neonate whose mother used doxepin in her third trimester and during the postpartum period. The doxepin dose had been 75 mg/day, but was tapered in the last weeks of pregnancy and was 35 mg/day at parturition. The amount of doxepin and desmethyldoxepin (active metabolite) ingested by the nursing infant was 10 to 20 mcg/kg/day (2.5% of the weight-adjusted dose of the mother) (Frey et al, 1999).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor cardiac rhythm and serial ECGs, serum electrolytes, renal function, hepatic enzymes, and CPK.
    B) Acid-base status should be followed with serial blood gases in severe toxicity when serum alkalinization is being performed or with sustained intubation.
    C) Urinalysis should be followed in those at risk for rhabdomyolysis. False positive urine TCA toxicology screens commonly occur with diphenhydramine, carbamazepine, cyclobenzaprine, quetiapine, and others.
    D) Serum tricyclic levels may be of use in drug-naive individuals and to help establish overdose in patients known to be taking tricyclics, but there is poor correlation between blood levels and clinical effects.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Monitor serum electrolytes, renal and hepatic function in patients with significant toxicity. Follow CPK levels in patients with prolonged seizures or coma.
    2) Serum tricyclic levels may be of use in drug-naive individuals and to help establish overdose in patients known to be taking tricyclics, but there is poor correlation between blood levels and clinical effects.
    B) ACID/BASE
    1) Monitor arterial blood gases with special attention to acid base status in patients with seizures, ECG changes and those requiring intubation or alkalinization.
    4.1.3) URINE
    A) URINALYSIS
    1) Follow urinalysis and urine output in patients with hypotension, dysrhythmias and those at risk for rhabdomyolysis.
    4.1.4) OTHER
    A) OTHER
    1) ECG
    a) Monitor serial ECGs and institute continuous cardiac monitoring in all patients with suspected tricyclic overdose. ECG changes may include sinus tachycardia, prolonged PR interval, widening of the QRS complex, QTc prolongation, rightward shift in the axis of the terminal 40 milliseconds of the QRS complex, T-wave flattening or inversion, ST segment depression, right bundle branch block, junctional rhythm and atrioventricular block (Goldberg et al, 1985) Neimann et al, 1986; (Caravati & Bossart, 1991; Foulke & Albertson, 1986; Langou et al, 1980; Fasoli & Glauser, 1981).
    1) QRS duration of greater than 0.10 seconds has been associated with an increased incidence of seizures and a QRS duration of 0.16 seconds or longer has been associated with an increased incidence of ventricular dysrhythmias (Boehnert & Lovejoy, 1985).
    2) QRS duration of greater than 0.10 seconds has been associated with an increased incidence of serious toxicity including coma, need for intubation, hypotension, seizures, and dysrhythmias (Hulten & Heath, 1983; Caravati & Bossart, 1991; Foulke & Albertson, 1986).
    3) Rightward deviation of the mean frontal plane terminal 40-ms QRS axis of 130 to 270 degrees or an R wave in lead aVR of > 3 mm are thought to be a more sensitive and specific indicator of tricyclic antidepressant toxicity than QRS duration (Liebelt et al, 1995; Wolfe et al, 1989), however they have not been correlated with complications or outcome.
    4) A T40-ms axis falling between 120 degree and 270 degrees require a negative T40-ms deflection (terminal S wave) in lead I and a positive T40-ms deflection (terminal R wave) in lead AVR.
    5) The following parameters, looked at individually, could classify correctly 60% to 62% of cyclic antidepressant overdoses in a series of 401 overdose patients: 1) heart rate >100; 2) QRS duration > 0.1 seconds; 3) T40 axis 130 to 270 degrees; 4) prolonged QTc (Lavoie et al, 1990).
    6) In a review of the use of ECG as a diagnostic tool, conflicting results were found regarding use of a QRS interval of 100 msec or more to predict toxicity. One small study reviewed showed QRS widening 100% sensitive for prediction of dysrhythmias and seizures while in another study it was only 53% sensitive. Improperly calibrated electrocardiograms, misinterpretation of tracings, and interobserver disagreement on manually measured QRS intervals were noted as problem areas of ECG monitoring (Caravati, 1999).
    7) In a study of experienced clinical toxicologists and their agreement in the measurement of QRS intervals, clinicians did not unanimously agree on whether the interval was above or below the cutoff of 100 milliseconds in 45 of 231 (19.5%) patients studied, indicating that reliance on the QRS interval alone for determination of seizure and arrhythmia monitoring is unjustified (Buckley et al, 1996).
    8) In a study of 36 patients with TCA overdose, maximum QRS interval and maximum T40-ms axis occurred at the time of presentation in a majority of patients (80% and 86%, respectively). The other patients had a median time to maximum QRS interval and median time to maximum T40-ms axis of 3 hours after presentation to the ED. Resolution of ECG abnormalities was highly variable (Liebelt et al, 1997).

Radiographic Studies

    A) CHEST RADIOGRAPH
    1) Chest x-ray should be obtained in any patient with significant toxicity or pulmonary symptoms (Frommer et al, 1987).

Methods

    A) OTHER
    1) Many laboratories can qualitatively detect the presence of cyclic antidepressants in the urine or gastric contents; however, this procedure will not differentiate between therapeutic and toxic amounts.
    2) Plasma tricyclic antidepressant levels are not available at all hospitals and are rarely available on a stat basis. Tricyclic antidepressant levels are not predictive of clinical complications (Boehnert & Lovejoy, 1985); serum levels are not likely to be useful in the initial assessment of the severity of overdose, unless they are quite elevated.
    B) IMMUNOASSAY
    1) A qualitative EMIT(R) homogeneous enzyme immunoassay is available for detecting the presence of most commonly used tricyclic antidepressants and their metabolites in serum or plasma.
    a) The assay detects amitriptyline, nortriptyline, desipramine, imipramine, clomiPRAMINE, doxepin, protriptyline, and trimipramine. Assay responses are cumulative of most tricyclic antidepressants and structurally similar metabolites present in the sample.
    b) However, the EMIT assay may give false positives for tricyclics in the presence of promethazine (Prod Info, 1993), diphenhydramine (Sorisky & Watson, 1986), cyproheptadine (Wians & Norton, 1993), thioridazine, chlorpromazine, or alimemazine (Benitez et al, 1986), and carbamazepine, cyclobenzaprine, or perphenazine (Labrosse & McCoy, 1988).
    2) The Adx(TM) assay is a competitive binding immunoassay for quantitative determination of tricyclic antidepressants in plasma or serum.
    a) Its sensitivity is 20 ng/mL (Prod Info, 1993g). It does not reliably detect amoxapine, maprotiline, mianserin, nomifensine, or trazodone (Prod Info, 1993g).
    b) Cross-reactivity may occur with cyproheptadine, chlorpromazine, cyclobenzaprine, chlorprothixene (Meenan et al, 1990), perphenazine, and promethazine (Prod Info, 1993g).
    c) Poklis et al (1990) compared the ADx(TM) assay with gas chromatography and showed good correlation with imipramine and desipramine. The assay was not well correlated with amitriptyline, nortriptyline, doxepin, or desmethyldoxepin levels.
    3) TDx(TM) is a fluorescent polarization assay for tricyclic antidepressants in serum.
    a) Significant cross-reactivity occurs with: dixyrazine, alimemazine, perazine, lofepramine, dothiepin, amitriptylinoxide, opipramol, and desmethyltrimipramine (Nebinger & Koel, 1990).
    4) URINE - Heterogeneous immunoassays are available that qualitatively detect tricyclic antidepressants in urine.
    a) In one study, the Biosite Triage(TM) heterogeneous immunoassay system had 100% specificity and sensitivity in detecting TCA in urine using TLC as a gold standard (Schwartz et al, 1994).
    5) DOTHIEPIN - Radioimmunoassay has been used to detect dothiepin and its metabolites in human body fluids and organ samples (Keller et al, 2000).
    6) IMMUNOASSAY INTERFERENCE
    a) The presence of hydroxyzine and cetirizine when using the fluorescence polarization immunoassay for measurement of tricyclic antidepressants (TCA) in serum has resulted in false-positive serum TCA concentrations. Apparent TCA levels may be observed at a hydroxyzine or cetirizine concentration of 500 ng/mL (Dasgupta et al, 2006).
    b) An immunoassay toxicological screen was conducted of the serum of a 5-year-old child who presented to the ED with agitation, visual hallucinations, slurred speech, ataxia, tachycardia, and fixed and dilated pupils. The serum tox screen was positive for tricyclic antidepressants, however, a review of medications available in the home did not indicate any tricyclic antidepressants. Further investigation revealed that the child had been playing with cyproheptadine tablets. High-performance liquid chromatography and gas chromatography/mass spectrometry of the patient's serum and urine, respectively, indicated the presence of cyproheptadine and no evidence of tricyclic antidepressants. The authors conclude that the initial immunoassay screening was cross-reactive from the presence of the nor-metabolite of cyproheptadine (Yuan et al, 2003).
    C) CHROMATOGRAPHY
    1) HAIR - HPLC and GC/MS methods for detecting tricyclic antidepressants in hair have been described (Couper et al, 1994; (Couper et al, 1995).
    2) SERUM OR PLASMA - GC/MS method for detecting tricyclic antidepressants in serum was used in one case report (Roberge & Krenzelok, 2001).
    a) Nyanda et al (2000) evaluated a reversed-phase HPLC method to determine the commonly used tricyclic antidepressant drugs in plasma or serum. It is suggested that this method is rapid (run time of 10 minutes), sensitive, free from interferences, and capable of detection of TCAs and their metabolites. A detection limit of 15 ng/mL was estimated. The recoveries ranged between 80% and 108% (Nyanda et al, 2000).
    3) DOTHIEPIN - TLC, GC, GC-MS, and HPLC methods have been used to detect dothiepin and its metabolites in human body fluids and organ samples. In one fatal overdose case, GC-MS and GC-ECD methods were used to separate and analyze dothiepin and its major metabolite, desmethyldothiepin, in postmortem specimens (Keller et al, 2000).
    D) SPECTROSCOPY/SPECTROMETRY
    1) DOTHIEPIN - UV-spectroscopy and cyclic voltammetry methods have been used to detect dothiepin and its metabolites in human body fluids and organ samples (Keller et al, 2000).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) SUMMARY
    1) Patients who remain symptomatic at 6 hours should be admitted to an intensive care unit. Serial monitoring of mental/respiratory status, acid-base status, and ECG should be performed. Patients who have resolved all clinical effects, have a normal QRS and QTc and are eating with normal bowel activity and no recurrent effects may be discharged.
    2) Patients should have a baseline electrocardiogram and be monitored for a minimum of 6 hours and the ECG repeated at the end of this observation period. Patients with significant symptoms or any ECG changes during this period, or with mild persistent symptoms (sinus tachycardia or lethargy), should be admitted and monitored in the ICU until mental status is baseline and the ECG has returned to normal for 24 hours (Tokarski & Young, 1988; Banahan & Schelkun, 1990).
    B) Several studies have shown that serious ECG and clinical effects occur within 6 hours of the overdose (Boehnert & Lovejoy, 1985; Callaham & Kassel, 1985; Tokarski & Young, 1988).
    C) There have been anecdotal reports of delayed toxicity and death from dysrhythmias after an asymptomatic interval (McAlpine et al, 1986; Sedal et al, 1972). These patients all had severe clinical signs and symptoms prior to apparent improvement and most received inadequate gastrointestinal decontamination. Retrospective studies however, do not support the notion of delayed cardiotoxicity in asymptomatic patients (Pentel & Sioris, 1981; Fasoli & Glauser, 1981; Tokarski & Young, 1988).
    D) Patients with ECG changes should be monitored in the ICU until the mental status is baseline, the patient is asymptomatic, and the ECG has returned to normal for 24 hours (Pentel & Sioris, 1981; Callaham, 1982) .
    E) If the patient remains symptom-free, and there are no anticholinergic signs present including tachycardia, then the patient may be referred for psychiatric clearance.
    6.3.1.2) HOME CRITERIA/ORAL
    A) Asymptomatic patients with inadvertent single substance ingestions can be managed at home with follow-up via telephone if the dose is less than the maximum daily single therapeutic dose for age or the following mg/kg amounts (whichever is less) (Woolf et al, 2007):
    1) AMITRIPTYLINE: 5 mg/kg or less; Maximum daily dose 150 to 300 mg adults, 200 mg adolescents and elderly
    2) CLOMIPRAMINE: 5 mg/kg or less; Maximum daily dose 300 mg adults, 200 mg children 10 to 18 years
    3) DESIPRAMINE: 2.5 mg/kg or less; Maximum daily dose 300 mg adults, 150 mg adolescents and elderly
    4) DOXEPIN: 5 mg/kg or less; Maximum daily dose 300 mg adults
    5) DOXEPIN CREAM: 5 mg/kg or less
    6) IMIPRAMINE: 5 mg/kg or less; Maximum daily dose 300 mg adults; 100 mg adolescents and elderly
    7) NORTRIPTYLINE: 2.5 mg/kg or less; Maximum daily dose 150 mg adults, 50 mg adolescents and elderly
    8) PROTRIPTYLINE: 1 mg/kg or less; Maximum daily dose 60 mg adults; 20 mg adolescents and elderly
    9) TRIMIPRAMINE: 2.5 mg/kg or less; Maximum daily dose 200 mg adults, 100 mg adolescents and elderly
    B) PEDIATRIC REVIEW: According to a retrospective study that evaluated 246 cases of unintentional cyclic antidepressant ingestions in pediatric patients, ingestions of less than 5 milligrams/kilogram resulted in little or no toxic effects, and therefore were able to be managed at home with follow-up via telephone (Spiller et al, 2003).
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) All toxic exposures should be reported to the designated regional poison center. Severe tricyclic toxicity patients should have a consultation by a medical toxicologist.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) All patients with intentional ingestions, or unintentional ingestions of unknown amounts, and all symptomatic patients (eg, weak, drowsy, dizzy, tremors, palpitations) should be referred to a healthcare facility; Patients with unintentional, single substance ingestions of more than the maximum single therapeutic dose (adjusted for age) or the following mg/kg ingestion (whichever is less) should be referred to a healthcare facility for observation (Woolf et al, 2007):
    1) AMITRIPTYLINE: greater than 5 mg/kg; Maximum daily dose 150 to 300 mg adults, 200 mg adolescents and elderly
    2) CLOMIPRAMINE: greater than 5 mg/kg; Maximum daily dose 300 mg adults, 200 mg children 10 to 18 years
    3) DESIPRAMINE: greater than 2.5 mg/kg; Maximum daily dose 300 mg adults, 150 mg adolescents and elderly
    4) DOXEPIN: greater than 5 mg/kg; Maximum daily dose 300 mg adults
    5) DOXEPIN CREAM: greater than 5 mg/kg
    6) IMIPRAMINE: greater than 5 mg/kg; Maximum daily dose 300 mg adults; 100 mg adolescents and elderly
    7) NORTRIPTYLINE: greater than 2.5 mg/kg; Maximum daily dose 150 mg adults, 50 mg adolescents and elderly
    8) PROTRIPTYLINE: greater than 1 mg/kg; Maximum daily dose 60 mg adults; 20 mg adolescents and elderly
    9) TRIMIPRAMINE: greater than 2.5 mg/kg; Maximum daily dose 200 mg adults, 100 mg adolescents and elderly

Monitoring

    A) Monitor cardiac rhythm and serial ECGs, serum electrolytes, renal function, hepatic enzymes, and CPK.
    B) Acid-base status should be followed with serial blood gases in severe toxicity when serum alkalinization is being performed or with sustained intubation.
    C) Urinalysis should be followed in those at risk for rhabdomyolysis. False positive urine TCA toxicology screens commonly occur with diphenhydramine, carbamazepine, cyclobenzaprine, quetiapine, and others.
    D) Serum tricyclic levels may be of use in drug-naive individuals and to help establish overdose in patients known to be taking tricyclics, but there is poor correlation between blood levels and clinical effects.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) Prehospital induction of emesis is contraindicated.
    B) ACTIVATED CHARCOAL
    1) Prehospital administration of activated charcoal may be considered only if endotracheal intubation can be performed in order to protect the airway should CNS depression and/or seizures occur.
    2) 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).
    3) 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) Activated charcoal should be given if within 2 hours of exposure. The patient’s ability to protect the airway or the need for intubation should be considered.
    2) Tricyclic antidepressants are significantly adsorbed to activated charcoal. It has been estimated that 4 grams of tricyclic antidepressant will be bound by 100 grams of activated charcoal (Braithwaite et al, 1978).
    a) A single 50 gram dose of activated charcoal (given 5 minutes after amitriptyline) reduced absorption of a 75 milligram amitriptyline dose by 99 percent (Karkkainen & Neuvonen, 1986).
    b) Two single-dose charcoal studies in overdosed patients did not demonstrate a significant clinical benefit in symptoms or elimination kinetics, however most patients were treated several hours postingestion. (Crome, 1983; Hulten et al, 1988).
    c) Another study of single dose activated charcoal administration following amitriptyline overdose found a correlation between time to activated charcoal administration and drug half life; clinical endpoints were not evaluated (Hedges et al, 1987).
    3) 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.
    4) 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).
    B) GASTRIC LAVAGE
    1) The role of gastric lavage is unclear, but should be considered for massive ingestions presenting within the first 60 minutes, in patients who can protect their airway or who have been intubated.
    2) Gastric lavage performed a mean 3.5 hours after ingestion of tricyclic antidepressants resulted in recovery of only 4 to 22 percent. The highest recovery was in patients lavaged 1.5 hours postingestion. Eighty-eight percent of recovered drug was retrieved with the first 5 liters of lavage fluid (Watson et al, 1989).
    3) A prospective study of 55 patients revealed a significant correlation between the tricyclic antidepressant plasma concentration and the decrease in arterial oxygen tension during gastric lavage (Jorens et al, 1991).
    4) In a randomized study of 51 patients with tricyclic antidepressant overdose, there was no difference in outcome between patients who received decontamination with 50 grams of activated charcoal and 10 ounces of magnesium citrate, those who underwent gastric lavage followed by 50 grams of activated charcoal and 10 ounces of magnesium citrate, and those who received 25 grams of activated charcoal followed by gastric lavage followed by a further 50 grams of activated charcoal and 10 ounces of magnesium citrate (Bosse et al, 1995).
    a) Outcome variables examined were length of hospital and ICU stay, duration of sinus tachycardia, duration of mechanical ventilation, seizures, QRS duration >100 milliseconds, hypotension, ventricular dysrhythmias and death.
    5) 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.
    6) 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.
    7) 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.
    8) 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).
    9) 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.
    C) MULTIPLE DOSE ACTIVATED CHARCOAL
    1) RECOMMENDATION - Because of conflicting results, study design flaws, and the potential for adverse effects such as impaction and intestinal infarction, the routine use of multiple dose charcoal is not recommended in tricyclic antidepressant overdose. Administration of a second dose should be considered in patients with serious toxicity because of the possibility of desorption of tricyclic antidepressants from charcoal. It should also be considered in patients who ingest modified release formulations (O'Connor et al, 2006).
    2) TCAs are known to undergo enterohepatic recirculation. Up to 15 percent of metabolized drug is excreted in bile and gastric secretions and then reabsorbed in the intestines, suggesting that multiple dose charcoal might be of benefit.
    3) Multiple dose activated charcoal reduced the half life of therapeutic doses of amitriptyline from 27 to 21 hours compared with no charcoal administration in volunteers (Karkkainen & Neuvonen, 1986).
    4) In overdose patients, multiple dose activated charcoal reduced the apparent half life of amitriptyline to 4 to 40 hours, compared to previously published values of 37 to over 60 hours in overdose (Swartz & Sherman, 1984). Changes in the severity or duration of intoxication were not reported; there is currently no evidence that multiple dose charcoal has any impact on the clinical course of tricyclic overdose.
    5) Multiple dose charcoal reduced the half life of doxepin and desmethyldoxepin compared with single dose charcoal, but not compared with no charcoal administration in volunteers (Scheinin et al, 1985). This suggests that doxepin may desorb from activated charcoal.
    6) Multiple dose charcoal did not reduce half life or increase clearance after intravenous imipramine administration in volunteers (Goldberg et al, 1985).
    7) In 3 patients with dothiepin overdose, multiple dose charcoal was associated with half-lives of 10.6 hours, 12.5 hours, and 13.1 hours (initial levels ranging from 819 mcg/L to 3,851 mcg/L), compared with half-lives of 18 to 24 hours in other cases in the reported literature (Ilett et al, 1991).
    8) COMPLICATIONS
    a) A 39-year-old woman on methadone maintenance received multiple dose activated charcoal after tricyclic antidepressant overdose (Gomez et al, 1994). Four days after admission she developed a charcoal stercolith and colonic perforation.
    b) A 17-year-old boy with anorexia nervosa, laxative and diuretic abuse and surgical gastric reduction was treated with multiple dose activated charcoal after overdose with codeine, temazepam and dothiepin (Merriman & Stokes, 1995). He developed a charcoal bezoar with jejunal impaction that required surgical removal.
    D) ENDOSCOPY
    1) CASE REPORT - Endoscopic removal of pharmacobezoars has been used in patients who overdosed on slow after other methods of decontamination (gastric lavage, activated charcoal) failed (Hojer & Personne, 2008).
    6.5.3) TREATMENT
    A) GENERAL TREATMENT
    1) Aggressive supportive care and serum alkalinization are the mainstays of therapy. Early intubation is advised for patients with CNS depression or ECG changes because of the potential for rapid deterioration. Serum alkalinization to a pH of 7.45 to 7.55 using intravenous boluses of sodium bicarbonate is recommended for patients with dysrhythmias or QRS widening. Intubation and hyperventilation may be used as an adjunct to sodium bicarbonate to achieve serum alkalinization, with careful monitoring of blood gases to avoid profound alkalemia. Conventional antiarrhythmics may also be necessary.
    2) Monitor vital signs, serial ECGs and institute intravenous access and continuous cardiac monitoring in all patients. Secure airway, administer oxygen, dextrose, naloxone, and thiamine in patients with depressed consciousness.
    3) CONDUCTION DEFECTS: Serum alkalinization using intravenous boluses of sodium bicarbonate is recommended for patients with QRS widening. Intubation and hyperventilation may be used as an adjunct to sodium bicarbonate to achieve serum alkalinization, with careful monitoring of blood gases to avoid profound alkalemia. A pH greater than 7.60 or a pCO2 less than 20 mmHg is probably undesirable.
    4) VENTRICULAR DYSRHYTHMIAS: Initial treatment consists of alkalinization of the blood to achieve a pH of 7.45 to 7.55. Intravenous sodium bicarbonate boluses are generally first line therapy. Intubation and hyperventilation may be used as an adjunct to sodium bicarbonate to achieve serum alkalinization, with careful monitoring of blood gases to avoid profound alkalemia. Early intubation is advised in any patient with QRS prolongation because of the potential for abrupt deterioration. Consider infusion of lipid emulsion in patients with refractory dysrhythmias or hypotension.
    a) Dysrhythmias are often torsade de pointes and may respond to alkalinization therapy. For those unresponsive consider magnesium, beta 1-sympathomimetics, or overdrive pacing. Lidocaine, although a type 1b agent may also be tried. Disopyramide, quinidine, procainamide are type 1a and are CONTRAINDICATED.
    5) SUPRAVENTRICULAR DYSRHYTHMIAS: Treatment may be required if the rate exceeds 160 beats per minute and the patient demonstrates signs and symptoms of hemodynamic instability. In these cases propranolol may be used cautiously.
    6) SEIZURES: If seizures cannot be controlled with diazepam, or recur, administer phenobarbital. If phenobarbital is ineffective consider paralysis and/or barbiturate coma.
    7) HYPOTENSION: Dopamine or norepinephrine may be used if alkalinization and volume repletion are ineffective. Intra-aortic balloons have been used successfully when pressors have failed. Hemodynamic interventions may be guided by right-sided heart catheterization. Infusion of lipid emulsion should be considered in patients with refractory hypotension or dysrhythmias.
    8) PHYSOSTIGMINE/CONTRAINDICATED: Use of physostigmine in the setting of tricyclic antidepressant overdose has been associated with the development of seizures and fatal dysrhythmias. It is NOT recommended.
    9) CNS DEPRESSION: Early intubation is advised in patients with mental status changes. Neurologic effects do not respond to serum alkalinization. Flumazenil is CONTRAINDICATED for seriously poisoned patients even if benzodiazepines are known coingestants, as use of flumazenil in the setting of tricyclic antidepressant overdose has been associated with seizures and ventricular dysrhythmias(Prod Info ROMAZICON(R) IV injection, 2004; Thomson et al, 2006).
    B) WIDE QRS COMPLEX
    1) SUMMARY - Increased QRS duration may be the best indication of severity of overdose and risk of serious complications, and should be treated aggressively (Nattel & Mittleman, 1984). According to medical directors from 58 of 73 (79%) regional U.S. Poison Centers, the QRS width for which serum alkalinization is recommended varies from 90 to 160 msec, with 31 (53%) of the centers recommending serum alkalinization following a QRS width of greater than 100 msec (Seger et al, 2003). Serum alkalinization using intravenous boluses of sodium bicarbonate to achieve a pH of 7.45 to 7.55 is generally first line therapy. Intubation and hyperventilation may be used as an adjunct to sodium bicarbonate to achieve serum alkalinization, with careful monitoring of blood gases to avoid profound alkalemia.
    2) SODIUM BICARBONATE
    a) SUMMARY - Sodium bicarbonate administration appears to have a beneficial effect on TCA induced conduction defects and dysrhythmias in humans and in animal models (Nattel et al, 1984; Nattel & Mittleman, 1984; Hedges et al, 1985; Hoffman et al, 1993). Studies have suggested that this effect may be secondary to increased pH (Brown et al, 1973; Brown, 1976; Nattel & Mittleman, 1984; Stone et al, 1995), increased concentration of sodium ion (Pentel & Benowitz, 1984; Hoffman et al, 1993; McCabe et al, 1993; McCabe et al, 1994) or both (Sasyniuk et al, 1986; Shanon & Liebelt, 1998; McCabe et al, 1998).
    1) Serum alkalinization to a pH of 7.45 to 7.55 should be achieved using intravenous boluses of sodium bicarbonate as necessary. Intubation and hyperventilation may be used as an adjunct to sodium bicarbonate to achieve serum alkalinization, with careful monitoring of blood gases to avoid profound alkalemia. Simultaneous hyperventilation and bicarbonate administration may result in profound alkalemia (Wrenn et al, 1992) and should only be done with extreme caution and careful monitoring of pH.
    b) SODIUM BICARBONATE DOSE - 1 to 2 milliequivalents/kilogram as needed to achieve a physiologic pH or slightly above (7.45 to 7.55) (Nattel & Mittleman, 1984). In some cases alkalinization of blood to a pH above physiologic may be necessary to reverse dysrhythmias (Sasyniuk et al, 1986).
    1) Effective alkalinization may not be achievable by using intravenous continuous infusion of sodium bicarbonate with conventional doses (2 ampules per liter). In an animal study, a bolus dose of 2 milliequivalents/kilogram transiently increased blood pH for 40 minutes, while a continuous conventional infusion was ineffective (Schlesinger & Janz, 1989).
    a) Adding bicarbonate to maintenance fluids is of unknown value as prophylaxis.
    2) CASE REPORTS - Several case reports describe reversal of dysrhythmias and improvement in hemodynamic status in patients with severe TCA overdose treated with sodium bicarbonate (Hoffman & McElroy, 1981; Molloy et al, 1984).
    3) STUDY/ANIMAL - Hyperventilation, that increased arterial pH to above 7.50, did not cause QRS narrowing in a rat model of desipramine overdose. Injection of 3 to 6 milliequivalents/kilogram of NaCl or high doses of sodium bicarbonate (3 milliequivalents/kilogram) reduced cardiac toxicity in acidotic and normal animals. The beneficial effects of NaHCO3 may therefore be due to its sodium content, not in its ability to change pH (Pentel & Benowitz, 1984).
    4) In an evidence-based review of the literature, Mackway-Jones (1999) reported that alkalinization to a pH of 7.55 appeared with sodium bicarbonate to be appropriate treatment for treatment of dysrhythmias after tricyclic overdose(Mackway-Jones, 1999).
    c) MECHANICAL HYPERVENTILATION - Induction of respiratory alkalosis by mechanical hyperventilation may be as effective as intravenous sodium bicarbonate (Bessen et al, 1983; Bessen & Niemann, 1985-86). A pH greater than 7.60 or a pCO2 less than 20 mmHg is probably undesirable(Bessen & Niemann, 1985-86).
    1) CASE REPORTS - Several case reports describe reversal of dysrhythmias and improvement in conduction delay in patients with severe TCA overdose treated with hyperventilation (Kingston, 1979; Bessen et al, 1983) Bessen & Niemann, 1986).
    2) STUDY/ANIMAL - In a dog model of amitriptyline overdose treatment with either hyperventilation or sodium bicarbonate reduced dysrhythmias and conduction slowing while infusion of isotonic or hypertonic sodium chloride did not (Nattel & Mittleman, 1984a).
    3) IN VITRO - Alkalinization therapy may work by affecting plasma protein binding of tricyclics (Brown et al, 1973; Levitt et al, 1986).
    3) SODIUM CHLORIDE
    a) CASE REPORT - HYPERTONIC SODIUM CHLORIDE - A 29-year-old woman was comatose and had a widened QRS interval after ingestion of approximately 8 grams nortriptyline. The patient developed refractory hypotension and worsening cardiac conduction despite fluid resuscitation, serum alkalinization, and high-dose catecholamine therapy. After a 200 mL bolus of 7.5% saline infused over 3 minutes, QRS interval narrowed from 139 milliseconds to 120 milliseconds and blood pressure increased; the patient survived without further sequelae (McKinney & Rasmussen, 2003; Rasmussen & McKinney, 1999).
    b) ANIMAL DATA - HYPERTONIC SODIUM CHLORIDE - In a randomized, controlled study of 24 domestic swine, hypertonic sodium chloride solution (15 mEq sodium per kilogram) was more effective in reversing induced severe tricyclic antidepressant toxicity compared to sodium bicarbonate, hyperventilation, or dextrose 5% in water. The authors suggested that in this animal model, sodium loading may be the most important factor in reversing tricyclic toxicity (McCabe et al, 1998). This study has been reported to have methodological flaws, in which the 3 groups may have been treated differently and thus affected survival outcome (Dick & Hack, 1999).
    C) CONDUCTION DISORDER OF THE HEART
    1) SUMMARY: Ventricular dysrhythmias (multifocal PVCs, ventricular tachycardia, flutter and fibrillation) may respond to serum alkalinization therapy to pH 7.45 to 7.55 by intravenous boluses of sodium bicarbonate. Intubation and hyperventilation may be used as an adjunct to sodium bicarbonate to achieve serum alkalinization, with careful monitoring of blood gases to avoid profound alkalemia. Dysrhythmias are often torsade de pointes and more respond to alkalinization therapy. For those unresponsive consider magnesium, beta-sympathomimetics, or overdrive pacing. Lidocaine, although a type 1b agent, may also be tried.
    2) CONTRAINDICATIONS: Quinidine, disopyramide, and procainamide are type 1a and are contraindicated as their effects on myocardial conduction are similar to that of the tricyclic antidepressants.
    3) LIDOCAINE
    a) STUDY/ANIMAL - In a canine model of amitriptyline overdose, 2 milligrams/kilogram of lidocaine was only transiently effective in reducing the frequency of ventricular ectopic complexes(Nattel & Mittleman, 1984a). Significant blood pressure reduction was an adverse effect of lidocaine use in this study.
    b) LIDOCAINE/DOSE
    1) ADULT: 1 to 1.5 milligrams/kilogram via intravenous push. For refractory VT/VF an additional bolus of 0.5 to 0.75 milligram/kilogram can be given at 5 to 10 minute intervals to a maximum dose of 3 milligrams/kilogram (Neumar et al, 2010). Only bolus therapy is recommended during cardiac arrest.
    a) Once circulation has been restored begin a maintenance infusion of 1 to 4 milligrams per minute. If dysrhythmias recur during infusion repeat 0.5 milligram/kilogram bolus and increase the infusion rate incrementally (maximal infusion rate is 4 milligrams/minute) (Neumar et al, 2010).
    2) CHILD: 1 milligram/kilogram initial bolus IV/IO; followed by a continuous infusion of 20 to 50 micrograms/kilogram/minute (de Caen et al, 2015).
    c) LIDOCAINE/MAJOR ADVERSE REACTIONS
    1) Paresthesias; muscle twitching; confusion; slurred speech; seizures; respiratory depression or arrest; bradycardia; coma. May cause significant AV block or worsen pre-existing block. Prophylactic pacemaker may be required in the face of bifascicular, second degree, or third degree heart block (Prod Info Lidocaine HCl intravenous injection solution, 2006; Neumar et al, 2010).
    d) LIDOCAINE/MONITORING PARAMETERS
    1) Monitor ECG continuously; plasma concentrations as indicated (Prod Info Lidocaine HCl intravenous injection solution, 2006).
    4) PHENYTOIN
    a) Phenytoin may be useful in improving cardiac conduction (Hagerman & Hanashiro, 1981; Boehnert & Lovejoy, 1985) and in treating ventricular dysrhythmias (Postlethwaite & Price, 1974; Mayron & Ruiz, 1986).
    b) In a dog model of amitriptyline intoxication phenytoin increased the duration and frequency of ventricular tachycardia (Callaham et al, 1988). Routine use of phenytoin as prophylaxis for seizures or dysrhythmias is not recommended, but it may be useful in treating dysrhythmias unresponsive to other therapy (Hagerman & Hanashiro, 1981; Mayron & Ruiz, 1986).
    c) PHENYTOIN LOADING DOSE (ADULT and CHILD) - Administer 15 milligrams/kilogram, up to 1 gram, intravenously not to exceed a rate of 0.5 milligram/kilogram/minute.
    d) PHENYTOIN MAINTENANCE DOSE (ADULT) - 2 milligrams/kilogram intravenously every 12 hours as needed. Monitor serum phenytoin levels just prior to initiating and during maintenance therapy to assure therapeutic levels of 10 to 20 micrograms/milliliter.
    e) PHENYTOIN MAINTENANCE DOSE (CHILD) - 2 milligrams/kilogram intravenously every 8 hours as needed. Monitor serum phenytoin levels just prior to initiating and during maintenance therapy to assure therapeutic levels of 10 to 20 micrograms/milliliter.
    5) MAGNESIUM
    a) Knudsen & Abrahamson (1997) reported the successful use of magnesium sulfate in the treatment of refractory ventricular fibrillation after TCA overdose(Knudsen & Abrahamsson, 1997). Two doses of 20 mmol magnesium sulfate and defibrillation resulted in successful treatment of amitriptyline-induced refractory ventricular fibrillation in a 44-year-old woman. The patient had not previously responded to sodium bicarbonate, lidocaine, or epinephrine.
    b) RANDOMIZED CONTROLLED TRIAL: A randomized controlled trial was conducted to determine the effectiveness of magnesium sulfate in the treatment of TCA intoxication, involving patients with a history of TCA overdose who presented to the emergency department in Tehran, Iran from March, 2009 to April, 2010. Inclusion criteria consisted of patients ingesting more than 20 mg/kg of a TCA, QRS duration greater than 10 ms, and the development of seizures or a blood pH of less than 7.2 after overdose. Exclusion criteria included any patients with a prior history of cardiovascular disease and the concomitant use of cardiac, anticholinergic, and antipsychotic agents. The patients were randomly divided into two groups: The control group who received sodium bicarbonate infusion and supportive therapy (n=36), and the case group who received IV magnesium sulfate (1 g every 6 hours) in addition to sodium bicarbonate infusion and supportive therapy (n=36). Eighteen patients from the case group (50%) and 19 patients from the control group (52.8%) were admitted to the ICU. Following their respective treatments, the mean ICU stay was 25.63 +/-9.33 hours and 82.67 +/-21.66 hours in the case and control groups, respectively (p<0.001). The mortality rate of the case and control groups was 13.9% and 33.3%, respectively (p=0.052). This suggests that magnesium sulfate may be effective when used with sodium bicarbonate and other supportive therapy in the treatment of TCA poisoning, although further studies are warranted (Emamhadi et al, 2012).
    c) Animal studies of the effects of magnesium on tricyclic-induced dysrhythmias have yielded conflicting results.
    1) In rats treated with norepinephrine prior to the induction of severe amitriptyline poisoning, magnesium infusion was associated with a decrease in heart rate and mean arterial blood pressure and an increased incidence of conversion from ventricular tachycardia to sinus rhythm (9 of 10 animals compared to 1 of 10 animals treated with lidocaine and 1 of 10 control animals) (Knudsen & Abrahamsson, 1994).
    2) In an isolated rat heart model of imipramine toxicity magnesium 6 mEq/L produced significantly decreased heart rate and left ventricular pressure, and increased the incidence of electromechanical dissociation and asystole (Kline et al, 1994).
    6) CARDIAC PACEMAKER
    a) CASE REPORT/ADOLESCENT: A 15-year-old girl presented to the emergency department with vomiting and fatigue approximately 12 hours after intentionally ingesting 12 25-mg imipramine tablets (approximately 6 mg/kg). The patient was hypotensive (70/30 mmHg) and an ECG revealed junctional escape rhythm (46 beats/min), no P waves, QRS widening, right bundle branch-like pattern, and QT interval prolongation. Despite supportive measures (including administration of IV fluids, vasopressors, sodium bicarbonate and atropine), the patient remained hypotensive and bradycardic. Following placement of a temporary pacemaker, as well as hemodialysis and hemoperfusion, the patient's blood pressure normalized and her urine toxicological screening for tricyclic antidepressants was negative. After removal of the pacemaker, a repeat ECG showed sinus bradycardia with QRS widening and a right bundle branch-like pattern; however, she was discharged on day 5 without sequelae, and a follow-up examination indicated no ECG abnormalities (Sert et al, 2011).
    D) FAT EMULSION
    1) Intravenous lipid emulsion (ILE) has been effective in reversing severe cardiovascular toxicity from local anesthetic overdose in animal studies and human case reports. Several animal studies and human case reports have also evaluated the use of ILE for patients following exposure to other drugs. Although the results of these studies are mixed, there is increasing evidence that it can rapidly reverse cardiovascular toxicity and improve mental function for a wide variety of lipid soluble drugs. It may be reasonable to consider ILE in patients with severe symptoms who are failing standard resuscitative measures (Lavonas et al, 2015).
    2) The American College of Medical Toxicology has issued the following guidelines for lipid resuscitation therapy (LRT) in the management of overdose in cases involving a highly lipid soluble xenobiotic where the patient is hemodynamically unstable, unresponsive to standard resuscitation measures (ie, fluid replacement, inotropes and pressors). The decision to use LRT is based on the judgement of the treating physician. When possible, it is recommended these therapies be administered with the consultation of a medical toxicologist (American College of Medical Toxicology, 2016; American College of Medical Toxicology, 2011):
    a) Initial intravenous bolus of 1.5 mL/kg 20% lipid emulsion (eg, Intralipid) over 2 to 3 minutes. Asystolic patients or patients with pulseless electrical activity may have a repeat dose, if there is no response to the initial bolus.
    b) Follow with an intravenous infusion of 0.25 mL/kg/min of 20% lipid emulsion (eg, Intralipid). Evaluate the patient's response after 3 minutes at this infusion rate. The infusion rate may be decreased to 0.025 mL/kg/min (ie, 1/10 the initial rate) in patients with a significant response. This recommendation has been proposed because of possible adverse effects from very high cumulative rates of lipid infusion. Monitor blood pressure, heart rate, and other hemodynamic parameters every 15 minutes during the infusion.
    c) If there is an initial response to the bolus followed by the re-emergence of hemodynamic instability during the lowest-dose infusion, the infusion rate may be increased back to 0.25 mL/kg/min or, in severe cases, the bolus could be repeated. A maximum dose of 10 mL/kg has been recommended by some sources.
    d) Where possible, LRT should be terminated after 1 hour or less, if the patient's clinical status permits. In cases where the patient's stability is dependent on continued lipid infusion, longer treatment may be appropriate.
    3) CASE REPORTS
    a) ADULT
    1) A 51-year-old man presented with tachycardia and a Glasgow coma scale [GCS] score of 3 approximately 1 hour after intentionally ingesting more than 65 50-mg tablets (greater than 43 mg/kg) as well as unknown quantities of quetiapine, citalopram, metoprolol, quinapril, and aspirin. An ECG revealed wide complex tachycardia with QRS widening (180 ms) and a prominent R wave. Following intubation and mechanical ventilation, and administration of sodium bicarbonate, a repeat ECG showed narrowing of the QRS to 96 ms; however, the patient became profoundly hypotensive (70/58 mmHg) despite vasopressor administration. Approximately 115 minutes post-ingestion, 100 mL 20% lipid emulsion was intravenously administered over 1 minute, followed by 400 mL administered over 30 minutes. Following administration, the patient's blood pressure increased to 140/80 mmHg and an ECG revealed further narrowing of the QRS duration to 80 ms. The patient remained hemodynamically stable, was extubated on hospital day 3, and was subsequently discharged to the psychiatry service on hospital day 7 (Harvey & Cave, 2012).
    2) A 36-year-old woman presented to the emergency department comatose (Glasgow Coma Scale score of 4), hypotensive, and tachycardic, approximately 90 minutes after ingesting 2.25 g of dothiepin. An ECG revealed a broad-complex tachycardia with a prolonged QTc interval of 502 ms. Approximately 30 minutes post-presentation, the patient developed cardiac arrest, that was successfully electrically cardioverted, although the patient continued to have broad complex tachycardia, despite administration of IV amiodarone, sodium bicarbonate boluses, and overdrive-pacing. A 100 mL bolus of 20% IV lipid emulsion was then administered over 1 minute, followed by a 400 mL infusion administered over a 15-minute period. During administration, sinus rhythm was restored and remained stable. Although the patient's clinical course was complicated with development of ventilator-associated pneumonia, she recovered with supportive care and was discharged with a repeat ECG demonstrating sinus rhythm with partial right bundle branch block and normal QRS and QTc durations (Blaber et al, 2012).
    3) A 25-year-old woman, who ingested an unknown amount of amitriptyline, developed hypotension, a wide QRS (186 ms), QTc interval prolongation, and episodic pulseless wide-complex tachycardia that recurred despite electrical cardioversion and treatment with lidocaine, magnesium sulfate, and sodium bicarbonate. The patient was then given 150 mL IV bolus of 20% lipids, followed by a continuous infusion at 16 mL/hour over the next 36 hours (receiving a total of 814 mL of 20% lipid (16.3 mL/kg)). During lipid treatment, the patient did not experience any more episodes of wide-complex tachycardia. However, within hours after discontinuing the lipid infusion, the QRS duration widened, the QTc interval continued to be prolonged, and the patient again developed pulseless wide-complex tachycardia twice, requiring electrical cardioversion each time. Following the second cardioversion, the tachycardia resolved, although the patient continued to have a prolonged QRS duration and QTc interval requiring intermittent sodium bicarbonate administration until 8 days post-admission (Kiberd & Minor, 2012).
    4) A 27-year-old man, who intentionally ingested 85 50-mg amitriptyline tablets (total dose 4.25 g) was comatose (Glasgow coma score [GCS] of 3) and developed hypotension, seizures, and persistent pulseless ventricular tachycardia requiring continued resuscitative efforts with CPR and IV boluses of epinephrine and sodium bicarbonate. Intralipid therapy was initiated, along with IV infusions of epinephrine and norepinephrine. The patient received an initial 20% intralipid bolus of 100 mL followed by an infusion of 400 mL, administered over 30 minutes. The epinephrine and norepinephrine infusions were weaned and discontinued, and the next day the patient's GCS was 15 and he was discharged from the ICU (Engels & Davidow, 2010)
    b) ADOLESCENT
    1) A 13-year-old girl developed a delayed-onset generalized tonic-clonic seizure and pulseless wide-complex dysrhythmias approximately 19 hours after ingesting an unknown amount of 150-mg amitriptyline tablets. Despite aggressive resuscitative efforts, the patient's rhythm degenerated to torsades de pointes. Approximately 30 minutes after initiation of resuscitation, 20% IV lipid emulsion therapy was started. The patient initially received 2 IV boluses of 1.5 mg/kg each, administered over a 3-minute period, with each bolus administered 5 minutes apart, followed by a continuous infusion of 0.25 mg/kg/minute for 30 minutes. Following administration of the second bolus, the patient's cardiac status improved with termination of her dysrhythmias (Levine et al, 2012).
    4) ANIMAL DATA
    a) An animal study, involving clomiPRAMINE administration in rabbits, was conducted to determine the efficacy of intralipid infusion as compared with sodium bicarbonate administration in the setting of clomiPRAMINE toxicity. Sedated and mechanically ventilated rabbits were infused with clomiPRAMINE at 320 mg/kg/hour, and subsequently developed severe hypotension. Sodium bicarbonate 8.4% at 3 mL/kg or 20% Intralipid at 12 mL/kg was then administered as rescue therapy. Three and 5 minutes following rescue therapy, the rate of change in mean arterial pressure was greater in the intralipid treated group as compared with the sodium bicarbonate-treated group (6.2 mmHg/min versus -0.25 mmHg/min, respectively, at 3 minutes; 4.4 mmHg/min versus 0.06 mmHg/min, respectively, at 5 minutes).
    1) In the second phase of the experiment, 8 sedated and mechanically ventilated rabbits from the first phase were infused with clomiPRAMINE at 240 mg/kg/hour to a mean arterial pressure of 25 mmHg. The rabbits then received either 2 mL/kg of 8.4% sodium bicarbonate (n=4) or 8 mL/kg of 20% intralipid (n=4) as rescue therapy. All of the intralipid-treated rabbits maintained spontaneous circulation; however, all of the sodium bicarbonate-treated rabbits developed pulseless electrical activity and were refractory to resuscitation(Harvey & Cave, 2007).
    2) In another study, rats received clomiPRAMINE 12.5 mg dissolved in either normal saline or 10% lipid (total volume 2.5 mL). All animals in the saline group died, while 3 of 15 (20%) of the lipid group died (Yoav et al, 2002).
    3) In another rat study, pretreatment with lipid emulsion did not increase mean arterial pressure or prolong survival in amitriptyline-poisoned animals (Bania & Chu, 2006).
    E) TACHYARRHYTHMIA
    1) SUMMARY: Supraventricular tachydysrhythmias may require treatment if the rate exceeds 160 beats per minute and the patient demonstrates signs and symptoms of hemodynamic instability. In these cases beta blockers may be used cautiously, a short acting agent such as esmolol is preferred.
    2) PROPRANOLOL
    a) Animal studies suggest that preventing tachycardia by sinus node destruction (Ansel et al, 1993) or by using bradycardic agents that impede sinus node automaticity without affecting myocardial repolarization or contractility may prevent the development of tricyclic induced ventricular dysrhythmias (Ansel et al, 1994). The use of beta blocking agents in this setting may result in abrupt hemodynamic collapse because of the combined negative inotropic effects of beta blocking agents and tricyclic antidepressants.
    F) SEIZURE
    1) Seizures in the setting of TCA overdose have been associated with abrupt deterioration of hemodynamic status (Ellison & Pentel, 1989) Taboulet et al, 1995) and should be aggressively controlled. Because of animal studies showing increased duration and frequency in ventricular tachycardia following the use of phenytoin in the setting of amitriptyline overdose (Callaham et al, 1988), phenobarbital may be preferable to phenytoin in treating seizures refractory to benzodiazepines.
    2) 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).
    3) 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 .
    4) 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).
    5) 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).
    6) 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).
    7) 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).
    8) RECURRING SEIZURES
    a) If seizures are not controlled by the above measures, patients will require endotracheal intubation, mechanical ventilation, continuous EEG monitoring, a continuous infusion of an anticonvulsant, and may require neuromuscular paralysis and vasopressor support. Consider continuous infusions of the following agents:
    1) MIDAZOLAM: ADULT DOSE: An initial dose of 0.2 mg/kg slow bolus, at an infusion rate of 2 mg/minute; maintenance doses of 0.05 to 2 mg/kg/hour continuous infusion dosing, titrated to EEG (Brophy et al, 2012). PEDIATRIC DOSE: 0.1 to 0.3 mg/kg followed by a continuous infusion starting at 1 mcg/kg/minute, titrated upwards every 5 minutes as needed (Loddenkemper & Goodkin, 2011).
    2) PROPOFOL: ADULT DOSE: Start at 20 mcg/kg/min with 1 to 2 mg/kg loading dose; maintenance doses of 30 to 200 mcg/kg/minute continuous infusion dosing, titrated to EEG; caution with high doses greater than 80 mcg/kg/minute in adults for extended periods of time (ie, longer than 48 hours) (Brophy et al, 2012); PEDIATRIC DOSE: IV loading dose of up to 2 mg/kg; maintenance doses of 2 to 5 mg/kg/hour may be used in older adolescents; avoid doses of 5 mg/kg/hour over prolonged periods because of propofol infusion syndrome (Loddenkemper & Goodkin, 2011); caution with high doses greater than 65 mcg/kg/min in children for extended periods of time; contraindicated in small children (Brophy et al, 2012).
    3) PENTOBARBITAL: ADULT DOSE: A loading dose of 5 to 15 mg/kg at an infusion rate of 50 mg/minute or lower; may administer additional 5 to 10 mg/kg. Maintenance dose of 0.5 to 5 mg/kg/hour continuous infusion dosing, titrated to EEG (Brophy et al, 2012). PEDIATRIC DOSE: A loading dose of 3 to 15 mg/kg followed by a maintenance dose of 1 to 5 mg/kg/hour (Loddenkemper & Goodkin, 2011).
    4) THIOPENTAL: ADULT DOSE: 2 to 7 mg/kg, at an infusion rate of 50 mg/minute or lower. Maintenance dose of 0.5 to 5 mg/kg/hour continuous infusing dosing, titrated to EEG (Brophy et al, 2012)
    b) Endotracheal intubation, mechanical ventilation, and vasopressors will be required (Brophy et al, 2012) and consultation with a neurologist is strongly advised.
    c) Neuromuscular paralysis (eg, rocuronium bromide, a short-acting nondepolarizing agent) may be required to avoid hyperthermia, severe acidosis, and rhabdomyolysis. If rhabdomyolysis is possible, avoid succinylcholine chloride, because of the risk of hyperkalemic-induced cardiac dysrhythmias. Continuous EEG monitoring is mandatory if neuromuscular paralysis is used (Manno, 2003).
    G) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) If alkalinization and volume repletion are ineffective in reversing hypotension, consider the use of pressor or inotropic agents (Frommer et al, 1987). Hemodynamic interventions may be guided by right-sided heart catheterization (Frommer et al, 1987). Dopamine and norepinephrine are the most commonly used agents. Animal data support the use of either agent (Vernon et al, 1991).
    b) Infusion of lipid emulsion should be considered in patients with refractory hypotension or dysrhythmias.
    c) Intra-aortic balloons have been used successfully when pressors have failed.
    d) SUMMARY
    1) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    2) NOREPINEPHRINE
    a) Hypotension may be a result of antidepressant-induced depletion of norepinephrine due to inhibition of neuronal uptake. Theoretically, norepinephrine and phenylephrine may be more effective agents due to their alpha-stimulating effects (Frommer et al, 1987).
    b) Norepinephrine, in doses of 15 and 30 micrograms/minute for 5 and 24 hours respectively, was successful in reversing circulatory shock refractory to dopamine in two adults with tricyclic antidepressant overdose (Teba et al, 1988).
    c) Disadvantages of using norepinephrine include the need for continuous nursing supervision, a central venous line, and the tissue damage caused by extravasation.
    d) NOREPINEPHRINE
    1) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    2) DOSE
    a) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    b) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    c) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    3) DOPAMINE
    a) While there are reports of patients whose TCA-induced shock was refractory to dopamine, but not to other agents (Heath et al, 1984; Teba et al, 1988; Hagerman & Hanashiro, 1981; Tran et al, 1997), most patients respond adequately to dopamine. Animal studies have yielded conflicting results regarding the efficacy of dopamine for TCA-induced hypotension; dosages used and varying experimental conditions limit the usefulness of these studies.
    b) HUMAN DATA
    1) In a retrospective review of patients with hypotension after TCA overdose, 9 of 15 (60%) patients treated with relatively low doses of dopamine (5 to 10 micrograms/kilogram/minute) responded. Six patients has hypotension that did not respond to dopamine at doses ranging from 10 to 20 micrograms/kilogram/minute and subsequently responded to norepinephrine (Tran et al, 1997).
    c) ANIMAL DATA
    1) Vernon et al (1991), using a dog amitriptyline model, found that dosing with dopamine 30 micrograms/kilogram/minute or with norepinephrine 0.25 microgram/kilogram/minute significantly improved cardiac output, heart rate, peak left ventricular pressure change, mean arterial pressure, and mixed venous oxygen saturation(Vernon et al, 1991). They concluded that both norepinephrine and dopamine were efficacious in this study.
    2) Studies performed on a cat model of TCA toxicity showed minimal or even deleterious effects of dopamine (10 to 40 micrograms/kilogram/minute) and dobutamine (Jackson & Banner, 1981).
    3) Follmer & Lum (1982) refuted this finding; in a cat model, they compared several agents and found that dopamine (20 microgram/kilogram/minute) was superior to norepinephrine (0.2 to 0.4 microgram/kilogram/minute) in reversing hypotension and in preventing death(Follmer & Lum, 1982).
    d) DOSE
    1) PREPARATION: Add 200 or 400 milligrams to 250 milliliters of normal saline or dextrose 5% in water to produce 800 or 1600 micrograms per milliliter or add 400 milligrams to 500 milliliters of normal saline or dextrose 5% in water to produce 800 micrograms per milliliter.
    2) DOSE: Begin at 10 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed. Norepinephrine should be added if more than 20 micrograms/kilogram/minute of dopamine is needed.
    3) CAUTION: If VENTRICULAR DYSRHYTHMIAS occur, decrease rate of administration. Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred.
    4) EPINEPHRINE
    a) In a rat model, epinephrine infusion reversed tricyclic-induced hypotension (Knudsen & Abrahamsson, 1993; Knudsen & Abrahamsson, 1994). Another study showed that epinephrine and norepinephrine increased survival in a rat model of tricyclic poisoning; sodium bicarbonate had additive beneficial effects with the most effective treatment being epinephrine and sodium bicarbonate (Knudsen & Abrahamsson, 1997a).
    5) GLUCAGON
    a) CASE REPORT - A 25-year-old woman presented with respiratory arrest, hypotension, widened QRS complexes (129 milliseconds with a rate of 99/minute) and seizures after ingesting 300 imipramine 25 milligrams and unknown amount of propranolol and temazepam (Sener et al, 1995). She was treated with intubation and ventilation, polygeline, diazepam, gastric lavage, activated charcoal, phenytoin, sodium bicarbonate and 1 milligram of glucagon without improvement.
    1) She was then given 10 milligrams of glucagon with improvement in her blood pressure form 70/30 to 110/70. Glucagon infusion of 10 milligrams over 6 hours and isoprenaline infusion were begun. Two hours later the QRS width was 89 milliseconds. Propranolol was never detected on toxicologic screening.
    b) CASE REPORT - A 36-year-old woman who took an overdose of dothiepin (admission serum level of 2.58 mg/L) presented with tachycardia, hypotension (60/30 mmHg), mydriasis, and a Glasgow coma score of 7. She was intubated and ventilated after respiratory arrest. ECG revealed broad complex tachycardia with a QRS interval of 0.16 to 0.2 seconds.
    1) Hypotension was refractory to treatment with conventional inotropic agents including epinephrine, norepinephrine, ephedrine, dobutamine, and aminophylline. After a 10 mg bolus of glucagon was given, an immediate rise in systolic pressure to 90 mmHg was noted. The effect of glucagon lasted 3 hours. Two 1 mg glucagon boluses followed by another 10 mg bolus resulted in increased blood pressure which was then maintained by continuous infusions of dobutamine, epinephrine, and dopamine (Sensky & Olczak, 1999).
    6) VASOPRESSIN
    a) A 56-year-old man presented to the emergency department (ED) unresponsive with tachycardia (105 bpm), hypotension (48/23 mmHg), and seizures after ingesting an unknown quantity of amitriptyline. He was given sodium bicarbonate, lorazepam, and a norepinephrine infusion. Despite administration of norepinephrine titrated to 20 mcg/min, his hypotension persisted. Five hours after presentation to the ED, a vasopressin infusion was initiated at 0.04 units/minute. Over the next 3 hours, his blood pressure improved, and the norepinephrine infusion was decreased. Over the next 30 hours, the patient was weaned off of all vasopressor agents (Barry et al, 2006).
    7) HIGH-DOSE INSULIN
    a) CASE REPORT - A 65-year-old woman developed cardiopulmonary arrest following multi-drug ingestion, including amitriptyline and citalopram. After successful resuscitation, hypotension and bradycardia continued to persist and an ECG demonstrated wide complex bradycardia with QRS widening and QTC interval prolongation, an ejection fraction of 55%, and mild left ventricular hypertrophy. Despite infusions of norepinephrine (40 mcg/minute), vasopressin (4 units/hour), sodium bicarbonate, and insulin (80 units/hour), the patient's condition continued to deteriorate. In order to improve cardiac inotropic function and peripheral perfusion, the insulin infusion was then increased incrementally at doses of 1 unit/kg/hour (100 units/hour increases) over a 5-hour period, with concomitant decreases of norepinephrine and vasopressin. At a total dose of 600 units of insulin/hour (6 units/kg/hour), the vasopressors were discontinued. The patient's hemodynamic status began to stabilize, and after 36 hours of high-dose insulin infusion of 500 units/hour or greater, the infusion was gradually weaned by 50 units/hour increments. The patient gradually recovered with residual confusion and unsteadiness requiring some assistance (Holger et al, 2009).
    H) FLUID/ELECTROLYTE BALANCE REGULATION
    1) Electrolytes should be monitored; potassium replacement should be done with caution as hyperkalemia may aggravate antidepressant-induced cardiac dysrhythmias.
    I) EXTRACORPOREAL MEMBRANE OXYGENATION
    1) Extracorporeal membrane oxygenation was employed for 60 hours for cardiac support in an 18-month-old with severe desipramine intoxication (Goodwin et al, 1993). The patient sustained right frontal and occipital infarcts but was neurologically normal on follow up.
    J) CARDIOPULMONARY BYPASS OPERATION
    1) A 37-year-old woman developed mental status depression, QRS widening and hypotension not responsive to fluids, dopamine or phenylephrine infusion after imipramine overdose (Williams et al, 1994). Approximately 5 hours after ingestion femoral-femoral extracorporeal circulation was initiated. She became hemodynamically stable, pressors were weaned and her QRS returned to normal. She died of multisystem organ failure 4 weeks after admission.
    2) In a swine model of severe amitriptyline overdose 9 of 10 animals treated with cardiopulmonary bypass survived compared with 1 of 10 treated with maximum supportive therapy (iv fluids, bicarbonate, hyperventilation, vasopressors, antiarrhythmics, open chest cardiac massage) (Larkin et al, 1994).
    K) 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).
    L) PHYSOSTIGMINE
    1) Use of physostigmine in the setting of tricyclic antidepressant overdose is controversial and has been associated with the development of seizures and fatal dysrhythmias. It is NOT recommended.
    a) Although coma altered mental status may be reversed dramatically in some cases (Heiser & Wilbert, 1974; Granacher & Baldessarini, 1975; Aquilonius & Hedstrand, 1978), physostigmine should not be used just to reverse coma in cases of tricyclic antidepressant overdose.
    b) Use of physostigmine in patients with acute tricyclic antidepressant overdose has been associated with seizures and intractable cardiac arrest (Stewart, 1979; Newton, 1975; Pentel & Peterson, 1980). Rapid IV administration of physostigmine may cause seizures, bradycardia, and asystole.
    M) FLUMAZENIL
    1) CONTRAINDICATED - Flumazenil should not be used in patients with serious cyclic antidepressant poisoning, as manifested by motor abnormalities (twitching, rigidity, seizure), dysrhythmias (wide QRS, ventricular dysrhythmia, heart block), anticholinergic signs (mydriasis, dry mucosa, hypoperistalsis), or cardiovascular collapse at presentation (Prod Info ROMAZICON(R) IV injection, 2004; Thomson et al, 2006; Burr et al, 1989; Marchant et al, 1989; Mordel et al, 1992; Geller et al, 1991; McDuffee & Tobias, 1995).
    2) Flumazenil should not be used in patients who are benzodiazepine dependent or who have been given benzodiazepines for control of a life-threatening condition(Prod Info ROMAZICON(R) IV injection, 2004; Thomson et al, 2006).
    3) There is no known benefit of treatment with flumazenil in a mixed drug overdose patient who is in critical condition. Flumazenil should NOT be used in cases where seizures are likely, from any cause (Prod Info ROMAZICON(R) IV injection, 2004; Thomson et al, 2006).
    4) STUDY/ANIMAL - Dogs treated with flumazenil 0.2 mg/kg after intoxication with amitriptyline and midazolam or amitriptyline alone developed worsening dysrhythmias whereas intoxicated dogs treated with normal saline did not (Lheureux et al, 1992). Two of the dogs treated with flumazenil died compared with none in the saline treated groups.
    N) EXPERIMENTAL THERAPY
    1) SUMMARY: Antibody Fab fragments, intralipid infusion, and flunarizine, have appeared effective in reducing tricyclic antidepressant toxicity in animal models.
    2) ANTIBODY FAB FRAGMENT
    a) HUMAN STUDY - In a study of 4 patients who overdosed on tricyclic antidepressants with a QRS interval greater than 100 milliseconds, administration of ovine anti-TCA immune Fab fragments resulted in increased total TCA serum and urine levels and inconsistent changes in free TCA levels. The authors speculated that Fab administration resulted in redistribution of TCA's into serum from tissue (Heard et al, 1999).
    b) Drug specific antibody Fab fragments have been shown to reduce tricyclic antidepressant-induced QRS prolongation in rats.
    c) Brunn et al (1992) found the combined treatment with anti-TCA Fab and sodium bicarbonate reduced desipramine-induced QRS prolongation in rats to a greater extent than either treatment alone(Brunn et al, 1992).
    d) In a rat model of severe tricyclic antidepressant toxicity administration of less than equimolar doses of antidesipramine Fab produced greater and more rapid improvement in cardiovascular status than no treatment (Dart et al, 1991).
    e) In a rat model of desipramine toxicity, administration of ovine desipramine-specific Fab antibody fragments resulted in rapid reversal of cardiotoxicity with narrowing of the QRS interval observed in a dose-response relationship (Dart et al, 1996).
    f) Hursting et al (1989), using a rabbit model of desipramine overdose, found that treatment with Fab induced significant changes in the concentrations of free and total desipramine in both serum and urine(Hursting et al, 1989).
    g) Active immunization of rabbits against nortriptyline increased survival and reduced the fraction of unbound drug in plasma while increasing the plasma concentration of total drug after intraperitoneal amitriptyline injection (Sabouraud et al, 1990).
    h) Rats treated with monoclonal anti-imipramine antibodies had reduced brain and heart imipramine concentration and increased serum imipramine concentrations compared with controls, suggesting that antibodies redistribute imipramine and reduce target tissue concentrations (Pentel et al, 1991).
    i) Fab administration reduced QRS duration and increased blood pressure in desipramine poisoned rats (Pentel et al, 1994).
    j) Fab prolonged survival during desipramine infusion by 58% compared with saline or albumin treated controls in a rat model (Pentel et al, 1995). The molar ratio of Fab to desipramine was 0.11.
    k) In a rat model of severe desipramine toxicity, high doses of Fab (4 grams/kilogram) were associated with decreased QRS duration and increased blood pressure but increased mortality (3 of 18 Fab treated rats compared with 0 of 6 controls) (Keyler et al, 1994).
    3) FLUNARIZINE - Simultaneous treatment of flunarizine (a calcium channel blocker) along with toxic doses of imipramine in rats resulted in prolonged survival time. The flunarizine treated animal did not develop seizures or significant cardiotoxicity. Additionally, cardiac output was improved (Trouve et al, 1986).
    4) Fv FRAGMENTS - In a rat model, recombinant monoclonal anti-desipramine Fv fragments were found to redistribute tracer doses of radiolabeled desipramine to serum (Shelver et al, 1995).
    5) ADENOSINE RECEPTOR ANTAGONISTS - A study, involving rats, was conducted to determine if adenosine receptors contribute to the cardiotoxicity induced by amitriptyline. The results of this study showed that the mean arterial pressure of rats, who were given an amitriptyline infusion followed by a continuous infusion of either a selective adenosine (A1) antagonist (DPCPX) or a selective adenosine (A2a) antagonist (CSC), increased significantly more than in the control group (rats who were given a continuous dextrose infusion). The adenosine antagonist infusions also resulted in a significant improvement in QRS prolongation as compared with the control group. In addition, pretreatment with the adenosine antagonists prevented amitriptyline-induced hypotension and QRS widening and prolonged survival following amitriptyline infusion. Adenosine receptors appear to play a role in the development of amitriptyline-induced cardiotoxicity and administration of adenosine receptor antagonists may be helpful in reducing or preventing amitriptyline-induced cardiotoxicity; however, further studies are warranted (Kalkan et al, 2004).

Enhanced Elimination

    A) SUMMARY
    1) There is no role for hemodialysis or other enhanced elimination techniques. Evidence of ongoing drug absorption or prolonged effects should prompt consideration of repeat doses of activated charcoal or whole bowel irrigation.
    2) The Extracorporeal Treatments in Poisoning (EXTRIP) workgroup conducted an extensive literature review, identifying 77 studies that described extracorporeal treatment in the setting of tricyclic antidepressant toxicity. The 77 studies included 1 observational study, 5 animal studies, 4 in vitro studies, 4 pharmacokinetic studies, and 63 case reports. There were no randomized trials or large observational series identified for inclusion. Based on the results of this literature review, which yielded a very low quality of evidence, the EXTRIP workgroup determined that there is no clinical benefit in utilizing extracorporeal removal and therefore recommends that extracorporeal treatments are NOT performed for tricyclic antidepressant poisoning (Yates et al, 2014).
    B) DIURESIS
    1) Diuresis is not effective in increasing tricyclic antidepressant clearance.
    C) HEMODIALYSIS
    1) Hemodialysis is not usually effective in the removal of tricyclic antidepressants from the body. Frank & Kierdorf (2000) report that hemoperfusion/hemodialysis should be considered a potential treatment option in acute TCA intoxication with severe cardiotoxicity, since the toxicokinetics of these drugs may totally differ from their usual pharmacokinetic behavior (Frank & Kierdorf, 2000).
    2) ADOLESCENT: A 15-year-old girl required a temporary pacemaker, as well as hemodialysis and hemoperfusion, after intentionally ingesting 12 25-mg imipramine tablets (approximately 6 mg/kg) and subsequently developing hypotension and junctional escape rhythm, refractory to standard supportive measures (Sert et al, 2011).
    D) HEMOPERFUSION
    1) Hemoperfusion is not routinely recommended following antidepressant overdose. Frank & Kierdorf (2000) report that hemoperfusion/hemodialysis should be considered a potential treatment option in acute TCA intoxication with severe cardiotoxicity, as the toxicokinetics of drugs may totally differ from their usual pharmacokinetic behavior (Frank & Kierdorf, 2000).
    2) Although the quantitative amount of tricyclic removed from the body is small (less than 1 to 3 percent of total body burden), clinical improvement has been reported following hemoperfusion in patients with life threatening hemodynamic and neurological complications following massive tricyclic overdose (Comstock et al, 1983; Heath et al, 1982; Koppel et al, 1992; Pentel & Bullock, 1982).
    3) CASE REPORTS
    a) ADULT: A 37-year-old man with endogenous depression developed cardiac arrest, severe hypotension, and wide QRS complexes (230-260 ms) after ingestion of at least 5000 milligrams of doxepin. Although fluid load, alkalinization, hypertonic saline and high-dose vasoactive substances did not improve the patient's condition, he improved after treatment with hemoperfusion/hemodialysis (Frank & Kierdorf, 2000). The plasma doxepin level decreased from 5150 mcg/l to 1150 mcg/l immediately after hemoperfusion/hemodialysis; drug clearance by the procedure was not determined.
    b) CHILD: A 17-month-old child ingested approximately 75 mg/kg of amitriptyline and presented to the hospital, 2 hours later, with lethargy, progressing to coma, and seizures. An ECG showed ventricular tachycardia and wide QRS complexes. Despite supportive therapy, the patient's dysrhythmias and seizures persisted. Hemoperfusion was performed for 2 hours, during which cardioversion with a defibrillator was performed 6 times because of ventricular fibrillation. The patient's cardiac rhythm normalized, her seizures resolved, and she was discharged approximately 9 days postingestion (Donmez et al, 2005). The patient's serum amitriptyline levels before and after hemoperfusion were 1,299 mcg/L and 843 mcg/L, respectively.
    c) CHILD: A 2.5 year-old child presented to the ED comatose (Glasgow Coma Scale(GCS) of 4), with seizures, hypotension, and tachycardia 4 hours after ingesting 20 x 25-mg tablets of amitriptyline. An ECG showed diffuse ventricular extrasystole, QRS and QT prolongation, and diffuse ventricular dysrhythmias. Initial serum amitriptyline concentration was 150 mcg/mL. Charcoal hemoperfusion was performed 7 hours postingestion. Serum amitriptyline concentration at the start of hemoperfusion was 150 mcg/ml and dropped to 37.5 mcg/mL at the end of hemoperfusion and the patient became responsive with a GCS of 7. His ECG normalized 3 hours after the first hemoperfusion session. At the start of a second hemoperfusion session the following day, serum amitriptyline concentration was 18.75 mcg/mL, decreasing to 4.7 mcg/mL by the end of that session. He completed recovered, and was discharged approximately 5 days post-ingestion (Islek et al, 2004).
    d) CHILD: An 18-month-old infant presented to the emergency department comatose (Glasgow coma score of 6), hypotensive (85/47 mmHg), and tachycardic (188 bpm), with shallow and irregular breathing, approximately 2 hours after ingesting approximately 57 mg/kg amitriptyline. An ECG showed a narrow complex tachycardia, indicative of re-entrant supraventricular tachycardia, that persisted despite continued administration of antiarrhythmics. Approximately 45 minutes after admission, the patient experienced tonic-clonic seizures that continued to recur despite administration of benzodiazepines. Because of the patient's refractory tachycardia and seizures, hemoperfusion was initiated and continued for 4 hours. At the end of the 4 hours, the patient's tachycardia resolved and there was no recurrence of his seizures. Within 24 hours his mental status normalized, and he was discharged approximately 5 days post-ingestion (Mutlu et al, 2011)
    e) ADOLESCENT: A 15-year-old girl required a temporary pacemaker, as well as hemodialysis and hemoperfusion, after intentionally ingesting 12 25-mg imipramine tablets (approximately 6 mg/kg) and subsequently developing hypotension and junctional escape rhythm, refractory to standard supportive measures (Sert et al, 2011).
    E) PLASMA EXCHANGE/PLASMAPHERESIS
    1) Plasma exchange has been used to treat a small number of patients. There is no good data to support its use, and it is not routinely recommended.
    2) CASE REPORTS - Therapeutic plasma exchange has been used in 3 adolescent patients following overdose ingestions of amitriptyline. Two patients (a 13-year-old and a 14-year-old) became unresponsive with Glasgow Coma Scale scores of 3 and 4, respectively, following overdose ingestions of 25 mg/kg and 18 mg/kg amitriptyline, respectively. Both patients became responsive to verbal stimuli within 6 hours after receiving plasma exchange, and were discharged without sequelae 3 days post-ingestion. The third patient, a 16-year-old girl, developed hypotension, first degree AV block, pancytopenia, prolonged INR, and hypokalemia after intentionally ingesting 12 mg/kg amitriptyline and 0.15 mg/kg colchicine. Following plasma exchange approximately 39 hours post-ingestion, the patient showed hemodynamic improvement with normalization of her INR (Bayrakci et al, 2007).
    3) CASE REPORT: A 15-year-old girl presented to the emergency department comatose (Glasgow Coma Scale (GCS) score of 5) approximately 4 hours after intentionally ingesting 22 mg/kg of amitriptyline. Prior to presentation, she had been mechanically ventilated at a local hospital due to decreased respiration and level of consciousness. Due to respiratory depression and the amount of amitriptyline ingested, plasmapheresis was performed for 4 hours, removing 2015 mL of plasma volume and using fresh frozen plasma as a replacement fluid. Four hours after initiating the procedure, the patient's mental status improved with a GCS score of 13, and she was extubated. Plasma amitriptyline concentration prior to plasmapheresis was 112.78 ng/mL compared to 9.23 ng/mL post-procedure. The patient recovered uneventfully and was discharged on hospital day 5 without sequelae (Karaci et al, 2013).

Case Reports

    A) SPECIFIC AGENT
    1) AMITRIPTYLINE: A 2-month-old infant was found unresponsive at home and was declared dead on arrival. Particulate material noted in the endotracheal tube at postmortem was identified as amitriptyline. The mother admitted to giving the infant the grandmother's pills to make the infant stop crying (Perrot, 1988).
    2) CLOMIPRAMINE: A 27-year-old man ingested 5 to 5.75 grams of clomiPRAMINE. He was found unconscious approximately 2 hours postingestion. Paramedics found the patient asystolic and apneic. The patient died despite resuscitative efforts. The clomiPRAMINE serum level during resuscitation was 6560.6 nanograms/milliliter; postmortem level was 8510 nanograms/milliliter.
    a) Although clomiPRAMINE has been characterized as a serotonin reuptake inhibitor, its active metabolite (desmethylclomiPRAMINE) inhibits catecholamine reuptake. Thus, in contrast to other serotonin reuptake inhibitors such as trazodone and fluoxetine, clomiPRAMINE appears to have the same morbidity and mortality that is associated with tricyclic antidepressants (Swanson-Biearman et al, 1989).
    3) CLOMIPRAMINE: Roberge et al (1994) report a case of pancreatitis and ileus in a pure clomiPRAMINE overdose ingestion of 750 to 1500 mg in a 48-year-old woman. Six hours postingestion she presented to the emergency department comatose with temperature 36 degrees C, respirations 20, heart rate 94 bpm, and blood pressure 122/76 mmHg. ECG revealed a slightly prolonged QRS complex; chest X-ray was normal.
    a) Treatment was begun with nasogastric sorbitol/activated charcoal and IV sodium bicarbonate. Plasma clomiPRAMINE level was 752 ng/mL and desmethylclomiPRAMINE was 1,543 ng/mL. After 7 hours she regained consciousness and complained of abdominal tenderness. Fifteen hours after admission, bowel sounds were absent and radiographic studies showed the presence of an ileus.
    b) Serum lipase on day 3 was 350 IU, and rose to 2,546 on day 5 with a corresponding amylase level of 112 IU. No evidence of biliary tract, renal, or pancreatic disease was noted on abdominal sonogram or tomography. Total parenteral nutrition was required as the pancreatitis and ileus resolved over a 2 week period (Roberge et al, 1994).
    4) DOTHIEPIN: Overdose involving 0.75 to 5 g has been associated with ECG changes, including depressed ST segment, prolonged PR interval, widened QRS, atrial fibrillation, ventricular fibrillation, and heart block (Goldstein & Claghorn, 1980).
    5) DOTHIEPIN CASE SERIES: A prospective study of patients admitted for tricyclic antidepressant overdose, 67 of whom had taken only dothiepin and 220 of whom had taken other TCAs, patients ingesting dothiepin had a higher incidence of seizures (9 of 67 vs 5 of 220) and dysrhythmias (4 of 67 vs 3 of 220) (Buckley et al, 1994). The dothiepin group had ingested a larger dose on average (1190 mg vs 868 mg), probably related to the larger average tablet strength (75 mg). Other studies have not shown a clear association between dothiepin use and increased seizure incidence (McGrady & Rees, 1994).
    6) DOXEPIN: A 54-year-old woman took 1.5 grams of doxepin and within 1 hour developed generalized convulsions, was deeply unconscious, and cyanotic. The patient exhibited extremely poor respiratory effort and no reflexes could be elicited.
    a) Pupils were dilated and unreactive and blood pressure was 65/30 mm Hg. Pulse was feeble and rapid and the ECG revealed ventricular tachycardia (60 to 150 beats per minute).
    b) Assisted respirations returned the ECG to normal within an hour and the patient required assisted ventilation for 16 hours. The endotracheal tube was removed in 24 hours and the patient subsequently made a good recovery (Williams, 1972).
    7) IMIPRAMINE: A 12-month-old child developed apnea and tonic-clonic seizures after desipramine overdose. Serum imipramine level 2 days later was 0.27 mg/L and desipramine level was 0.32 mg/L. Clinical course was characterized by tachycardia, wide QRS complexes, agitation, and mild hypotension. Treatment included supportive care, antibiotics, sedation with fentanyl, digitalization, and repeated doses of edrophonium. The patient recovered without sequelae and was discharged 4 days later (Tribble et al, 1989).
    8) NORTRIPTYLINE: Ingestions of up to 1.875 g have been reported with recovery. Nortriptyline overdose is characteristic of all tricyclic antidepressant poisonings and may be accompanied by hypotension, coma, bradycardia, apnea, convulsions, and cardiac dysrhythmias (Rendoing et al, 1969).
    9) DESIPRAMINE INSUFFLATION: A 31-year-old man experienced seizures, tachycardia (120 bpm), and QRS widening after intranasal inhalation of a few 50 mg crushed desipramine tablets in addition to drinking alcohol and smoking cocaine. The patient recovered after intubation, defibrillation, lidocaine, and sodium bicarbonate administration. Desipramine blood level was 1978 nanograms/mL upon admission (Marshall et al, 1998).
    10) CASE REPORTS (FATALITIES): Deaths have been reported (Vohra, 1975; Moccetti, 1973; Sjoqvist et al, 1972; Duke & Horton, 1969; Rendoing et al, 1969; Brackenridge, 1968; Stinnett, 1968) .

Summary

    A) TOXICITY: Ingestion of 10 to 20 mg/kg of most antidepressants constitutes a moderate to serious exposure where coma and cardiovascular symptoms are expected. Ingestion of either more than the maximal single dose (for age) or the following mg/kg dose (whichever is less) is considered potentially toxic: AMITRIPTYLINE: greater than 5 mg/kg. CLOMIPRAMINE: greater than 5 mg/kg. DESIPRAMINE: greater than 2.5 mg/kg. DOXEPIN: greater than 5 mg/kg. DOXEPIN CREAM: greater than 5 mg/kg. IMIPRAMINE: greater than 5 mg/kg. NORTRIPTYLINE: greater than 2.5 mg/kg. PROTRIPTYLINE: greater than 1 mg/kg. TRIMIPRAMINE: greater than 2.5 mg/kg . Fatalities have occurred in children following the ingestion of as little as 250 mg of amitriptyline or imipramine.
    B) THERAPEUTIC DOSE: ADULT DOSES include: Amitriptyline 75 to 300 mg/day in divided doses; desipramine 100 to 300 mg/day; doxepin 75 to 300 mg/day; imipramine 100 to 300 mg/day in divided doses; nortriptyline 75 to 150 mg/day in divided doses. PEDIATRIC DOSES include: Amitriptyline 0.1 to 2 mg/kg/dose, 10 to 20 mg/dose in adolescents; desipramine 1 to 5 mg/kg/day, 25 to 150 mg/day in adolescents; imipramine 1 to 4 mg/kg/day, 30 to 100 mg/day in adolescents; nortriptyline 1 to 3 mg/kg/day, 30 to 50 mg/day in adolescents.

Therapeutic Dose

    7.2.1) ADULT
    A) SPECIFIC SUBSTANCE
    1) AMITRIPTYLINE
    a) ORAL: Usual initial oral dose is 75 milligrams/day in divided doses. Dose can be increased to 150 milligrams/day in outpatients and 200 to 300 milligrams/day in hospitalized patients (Prod Info ELAVIL(R) oral tablets injection, 2000)..
    b) INTRAMUSCULAR: Initially, the recommended dose is 20 to 30 milligrams (2 to 3 milliliters) four times daily (Prod Info ELAVIL(R) oral tablets injection, 2000).
    2) DESIPRAMINE
    a) ORAL: Usual oral dose is 100 to 200 milligrams/day to a maximum of 300 milligrams/day (Prod Info desipramine hcl oral tablets, 2006).
    3) DOXEPIN
    a) ORAL: For mild to moderate depression/anxiety, the usual dosage regimen is 75 to 150 milligrams/day, up to 300 milligrams/day in patients with severe depression/anxiety (Prod Info SINEQUAN(R) oral capsule, 2005).
    4) IMIPRAMINE
    a) HOSPITALIZED PATIENTS: The usual initial dose is 100 milligrams/day in divided doses. The dose may be increased up to 300 milligrams/day if needed (Prod Info TOFRANIL(R) tablets, 2005).
    b) OUTPATIENTS: The usual initial dose is 75 milligrams/day. The dose may be increased up to 150 milligrams/day if needed. The maximum dose is 200 milligrams/day (Prod Info TOFRANIL(R) tablets, 2005).
    5) NORTRIPTYLINE
    a) ORAL: Usual initial dose is 75 to 100 milligrams/day in three or four divided doses. Dose can be increased to a maximum of 150 milligrams/day (Prod Info PAMELOR(R) oral capsules, oral solution, 2006).
    6) PROTRIPTYLINE
    a) ORAL: The recommended dose is 15 to 40 milligrams/day in divided doses, up to 60 milligrams/day (Prod Info VIVACTIL(R) tablets, 2004).
    7) TRIMIPRAMINE MALEATE
    a) HOSPITALIZED PATIENTS: Initially, the recommended dose is 100 milligrams/day in divided doses, up to a MAXIMUM dose of 300 milligrams/day (Prod Info trimipramine maleate oral capsules, 2006).
    b) OUTPATIENTS: Initially, the recommended dose is 75 milligrams/day in divided doses, up to a MAXIMUM dose of 200 milligrams/day if needed (Prod Info trimipramine maleate oral capsules, 2006).
    7.2.2) PEDIATRIC
    A) GENERAL
    1) CAUTION
    a) Doses above 3.5 milligrams/kilogram of tricyclic antidepressants or serum concentrations higher than 150 nanograms/milliliter may increase the risk of asymptomatic electrocardiographic changes (slight prolongation of PR interval, moderate increase in QRS duration) indicative of delayed cardiac conduction in children and adolescents (Biederman, 1991).
    B) SPECIFIC SUBSTANCE
    1) AMITRIPTYLINE -
    a) DEPRESSION
    1) 13 to 17 years: The usual recommended dose is 10 milligrams orally 3 times daily with 20 milligrams at bedtime (Prod Info amitriptyline HCl oral tablet, 2010).
    2) 12 years and younger: Safety and efficacy have not been established (Prod Info amitriptyline HCl oral tablet, 2010).
    2) DESIPRAMINE
    a) 13 to 17 years: The usual recommended oral dose is 25 to 100 milligrams/day, up to a maximum dose of 150 milligrams/day (Prod Info NORPRAMIN(R) oral tablets, 2012).
    b) 12 years and younger: Safety and efficacy have not been established (Prod Info NORPRAMIN(R) oral tablets, 2012).
    3) IMIPRAMINE
    a) DEPRESSION
    1) 13 to 17 years: Initially, the recommended dose is 30 to 40 milligrams/day. The dose may be increased up to a MAXIMUM of 100 milligrams/day (Prod Info imipramine HCl oral film coated tablets, 2012).
    2) 12 years and younger: Safety and efficacy have not been established (Prod Info imipramine HCl oral film coated tablets, 2012).
    b) ENURESIS
    1) 6 to 12 years: Initial, 25 mg orally 1 hour before bedtime, may increase in 25 mg increments to MAX dose of 50 mg/d or 2.5 mg/kg/d (Prod Info imipramine HCl oral film coated tablets, 2012).
    2) 13 years and older: Initial, 25 mg orally before bedtime, may increase in 25 mg increments to MAX dose of 75 mg/d or 2.5 mg/kg/d (Prod Info imipramine HCl oral film coated tablets, 2012).
    4) NORTRIPTYLINE
    a) 13 to 17 years: The recommended dose is 30 to 50 milligrams/day given either as a single dose once daily or in divided doses (Prod Info PAMELOR(TM) oral capsules, oral solution, 2012).
    b) 12 years and younger: Safety and efficacy have not been established (Prod Info PAMELOR(TM) oral capsules, oral solution, 2012).
    5) PROTRIPTYLINE
    a) ADOLESCENTS: The recommended dose is 5 milligrams 3 times daily, up to 20 milligrams/day (Prod Info VIVACTIL(R) tablets, 2004).
    6) TRIMIPRAMINE MALEATE
    a) ADOLESCENTS: 50 milligrams/day, up to 100 milligrams/day (Prod Info trimipramine maleate oral capsules, 2006).

Minimum Lethal Exposure

    A) PEDIATRIC
    1) Fatal poisonings have occurred in children following the ingestion of as little as 250 milligrams of amitriptyline or imipramine(Rosenbaum & Kou, 2005; Linakis, 1988; Manoguerra, 1982).
    a) Assuming 15 to 20 milligrams/kilogram may be a lethal dose, if no medical attention is available, for a child weighing 10 kilograms, four 50-milligram tablets may be fatal (Frommer et al, 1987).
    2) AMITRIPTYLINE: Two children died following ingestion of amitriptyline. The first child was a 1-year-old who ingested 97.5 mg/kg of amitriptyline and died following development of status epilepticus. The second child was a 1.5-year-old who ingested 36 mg/kg of amitriptyline and died due to cardiopulmonary arrest (Caksen et al, 2006).
    3) CASE REPORT/IMIPRAMINE: A 6-year-old boy, who was taking imipramine 200 mg/day for 3 months, collapsed at school, requiring CPR. Although the patient's heartbeat was re-established, he presented to the emergency department with fixed and dilated pupils and no spontaneous respiration. An ECG showed ventricular tachycardia and fibrillation with one episode of QT prolongation (580 msecs). A plasma tricyclic antidepressant level was 1000 ng/mL (imipramine and desipramine). Despite continuous supportive measures, the patient died following recurrent ventricular fibrillation subsequently leading to cardiac arrest (Preskorn, 2011).
    B) ADULT
    1) A fatal intoxication due to an unknown quantity of Harmomed(R) (dothiepin and diazepam) capsules has been reported. The peripheral blood concentrations of dothiepin and desmethyldothiepin were 7.28 milligrams/liter and 2.03 milligrams/liter, respectively (Keller et al, 2000).
    2) DOTHIEPIN: Fatalities have been reported with dothiepin blood concentrations between 1 and 19 milligrams/liter (Keller et al, 2000).
    3) CLOMIPRAMINE/CASE REPORT: A 42-year-old woman developed a pharmacobezoar after intentionally ingesting 60 75-mg sustained release clomiPRAMINE tablets and 30 25-mg doxepin capsules. Following surgical removal, crushing of the pharmacobezoar showed that it was comprised entirely from tablets, which had the appearance of clomiPRAMINE tablets. Despite removal of the bezoar, the patient clinically deteriorated and, 32 hours later, died from multi-organ failure (Magdalan et al, 2013).

Maximum Tolerated Exposure

    A) GENERAL/SUMMARY
    1) Ingestion of either more than the maximal single dose (for age) or the following mg/kg dose (whichever is less) is considered potentially toxic(Woolf et al, 2007):
    a) AMITRIPTYLINE: greater than 5 mg/kg
    b) CLOMIPRAMINE: greater than 5 mg/kg
    c) DESIPRAMINE: greater than 2.5 mg/kg
    d) DOXEPIN: greater than 5 mg/kg
    e) DOXEPIN CREAM: greater than 5 mg/kg
    f) IMIPRAMINE: greater than 5 mg/kg
    g) NORTRIPTYLINE: greater than 2.5 mg/kg
    h) PROTRIPTYLINE: greater than 1 mg/kg
    i) TRIMIPRAMINE: greater than 2.5 mg/kg
    2) Ingestion of 10 to 20 milligrams/kilogram of most antidepressants constitutes a moderate to serious exposure where coma and cardiovascular symptoms are expected. As little as 15mg/kg may be fatal in a child (Rosenbaum & Kou, 2005). One child developed severe toxicity after a dose of 100 milligrams of desipramine, estimated ingestion 6.67 milligrams/kilogram.
    3) According to a retrospective study that evaluated 246 cases of unintentional cyclic antidepressant ingestions in pediatric patients, ingestions of less than 5 milligrams/kilogram resulted in little or no toxic effects (Spiller et al, 2003).
    B) SPECIFIC SUBSTANCE
    1) AMITRIPTYLINE
    a) ADULT
    1) A 39-year-old woman experienced prolonged coma (for 5 days) and loss of brainstem reflexes following amitriptyline overdose. Two and 8 hours after the patient was found comatose, the GC/MS serum analysis revealed total TCA levels of 1,348 ng/mL (amitriptyline 1,310 ng/mL, nortriptyline 39 ng/mL) and 1,020 ng/mL (amitriptyline 868 ng/mL, nortriptyline 152 ng/mL), respectively (Roberge & Krenzelok, 2001).
    2) A 44-year-old woman developed coma and abnormal atrial and ventricular repolarization resembling acute infarction following ingestion of an unknown amount of amitriptyline (serum amitriptyline 4880 ng/mL at presentation). In addition, migrating ST segment elevation to another territory with abnormally inflected T wave (mimicking subepicardial injury) was noted. After 10 days in the intensive care unit, she recovered with no apparent neurological sequelae (Zakynthinos et al, 2000).
    3) A 27-year-old man, who intentionally ingested 85 50-mg amitriptyline tablets (total dose 4.25 g) was comatose (Glasgow coma score [GCS] of 3) and developed hypotension, seizures, and persistent pulseless ventricular tachycardia requiring continued resuscitative efforts with CPR and IV boluses of epinephrine and sodium bicarbonate. With administration of intralipids with IV infusions of epinephrine and norepinephrine, the patient gradually recovered (Engels & Davidow, 2010).
    b) PEDIATRIC
    1) A 2-year-old child presented to the emergency department comatose (Glasgow Coma Scale of 8), tachycardic (170 bpm) and hypotensive (60/40 mmHg) approximately 1 hour after ingestion of 500 mg (35.7 mg/kg) of amitriptyline. Within 10 min post-presentation, the patient developed generalized seizures responsive to IV benzodiazepines. With supportive therapy, he gradually recovered, without sequelae, and was discharged 2 weeks post-ingestion (Deegan & O'Brien, 2006).
    2) Seizure-like movements and altered mental status were reported in an 8-year-old child who ingested one 25-mg (0.8 mg/kg) amitriptyline tablet (Grover et al, 2012).
    3) ADOLESCENTS: Intentional overdose ingestions of amitriptyline were reported in 3 adolescent girls. The first patient, a 13-year-old, ingested 25 mg/kg amitriptyline and subsequently became comatose (Glasgow Coma Scale (GCS) score of 3) and acidotic. The second patient, a 14-year-old, was unresponsive (GCS of 4), and developed hypotension and first degree AV block after ingesting 18 mg/kg amitriptyline. The third patient, a 16-year-old, developed hypotension, first degree AV block, pancytopenia, and hypokalemia after ingesting 12 mg/kg amitriptyline and 0.15 mg/kg colchicine. All 3 patients recovered without sequelae after receiving therapeutic plasma exchange and supportive care (Bayrakci et al, 2007).
    4) A retrospective review of amitriptyline poisoning cases in pediatric patients (n=52; age range 1 to 14 years, mean age 4.6 years) reported the development of lethargy, coma, hypotension, tachycardia, and seizures. The mean ingested amitriptyline dose, known in 23 patients, was 9.4 +/- 5.8 mg/kg (range 2.3 to 27 mg/kg, median 8.6 mg/kg) (Olgun et al, 2009).
    5) CASE REPORT/INFANT: An 18-month-old infant ingested approximately 57 mg/kg amitriptyline and, within 3 hours, became comatose (Glasgow coma score of 6), and developed supraventricular tachycardia, hypotension, and seizures. Following hemoperfusion, the patient gradually recovered and was discharged approximately 5 days post-ingestion (Mutlu et al, 2011).
    6) CHRONIC OVERDOSE: Generalized tonic-clonic seizures and ECG abnormalities were reported in a 6-year-old child who had been receiving 300 mg (15 mg/kg) amitriptyline every night for 1 month instead of the prescribed 30 mg nightly for treatment of insomnia secondary to ADHD. With supportive treatment, the patient recovered and was discharged on hospital day 7 (Clement et al, 2012).
    2) DESIPRAMINE
    a) ADULT
    1) A 30-year-old woman ingested 2950 mg desipramine, developed seizures, respiratory arrest, cardiac dysrhythmia and circulatory collapse, and survived (Hughes & Rome, 1984).
    b) PEDIATRIC
    1) A 3-year-old girl (weight unknown, estimated 15 kilograms) ingested 100 mg desipramine and developed severe toxicity including cardiac dysrhythmias and seizures. The child survived without permanent sequelae (Jue, 1976).
    3) DOXEPIN
    a) ADULT
    1) A successful treatment with hemoperfusion over hemoresin and hemodialysis in a 37-year-old man (after ingestion of at least 5000 mg doxepin) lowered the doxepin plasma level from 5150 (toxic>500, therapeutic 100-250 mcg/L) to 1150 mcg/L (Frank & Kierdorf, 2000).
    2) Torsade de pointes tachycardia due to the long QT syndrome was reported in a 50-year-old woman after taking 60 doxepin tablets (6 grams); maximum serum level of doxepin was 2845 nmol/L 12 hours after admission (Alter et al, 2001).
    b) PEDIATRIC
    1) A 5-year-old girl developed CNS depression, hypotension, and tachycardia after receiving a total of 1329 mg doxepin-containing cream topically used for exanthematous rash. The patient completely recovered 18 hours after presentation (Toerne et al, 1997).
    2) Topical administration of 30 grams of doxepin 5% cream in 24 hours for a generalized eczematous rash resulted in altered mental status and ECG changes in a 5-year-old girl. Serum concentrations of doxepin and desmethyldoxepin (major active metabolite) were 11.95 nanograms per milliliter (ng/mL) and 17.71 ng/mL, respectively. Eighteen hours following skin decontamination with soap and water, a full recovery was made and the patient was discharged (Zell-Kanter et al, 2000).
    4) IMIPRAMINE
    a) ADULT
    1) A 27-year-old woman survived ventricular fibrillation, cardiovascular collapse, and seizures after a massive ingestion of imipramine in a suicide attempt. Plasma imipramine levels reached a peak of 6000 nanograms/mL (Sandeman et al, 1997).
    b) PEDIATRIC
    1) A 13 year-old boy ingested 1000 mg imipramine, developed seizures, respiratory arrest, and coma, and survived (Pulst & Lombroso, 1983).
    2) A 15-year-old girl required a temporary pacemaker, as well as hemodialysis and hemoperfusion, after intentionally ingesting 12 25-mg imipramine tablets (approximately 6 mg/kg) and subsequently developing hypotension and junctional escape rhythm, refractory to standard supportive measures (Sert et al, 2011).
    3) An 18-month-old child lost consciousness for 5 minutes, and developed facial twitching, up-rolling eyes, metabolic acidosis, hypotension, and ECG abnormalities (widened QRS complex, sinus tachycardia) after a suspected ingestion of approximately 7 25-mg imipramine tablets. The patient gradually recovered following supportive therapy, including IV sodium bicarbonate and dopamine infusions (Hon et al, 2015).
    5) NORTRIPTYLINE
    a) ADULT
    1) Ingestions of up to 1.875 grams have been reported with recovery in adults (Rendoing et al, 1969).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) GENERAL
    a) The therapeutic range for most tricyclic antidepressants is 100 to 260 nanograms/milliliter.
    b) Sirota et al (1990) studied 20 patients taking up to 150 milligrams/day amitriptyline. Levels found 3 weeks after initiation of treatment were: amitriptyline, approximately 114 nanograms/milliliter; and nortriptyline, 178 nanograms/milliliter (Sirota et al, 1990).
    c) Therapeutic doses of nortriptyline in elderly patients resulted in an increased PR interval associated with increasing nortriptyline concentrations, and increased QRS duration and QTc intervals associated with increasing Z-10 hydroxynortriptyline metabolite concentration (Schneider et al, 1988).
    2) SPECIFIC SUBSTANCE
    a) TRIMIPRAMINE: Therapeutic blood or plasma concentrations: 0.01-0.24 milligram/liter (Musshoff et al, 1999).
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) GENERAL
    a) The therapeutic range for most tricyclic antidepressants is 100 to 260 nanograms/milliliter with toxic symptoms sometimes seen below 500 nanograms/milliliter. Serious symptoms are usually associated with levels greater than 1000 nanograms/milliliter.
    b) The development of delirium and seizures was associated with TCA plasma levels above 300 nanograms/milliliter and 1000 nanograms/milliliter respectively.
    c) Lethal tricyclic antidepressant levels reported from forensic studies have ranged from 4000 to 80000 nanomoles/liter (1100 to 21800 nanograms/milliliter).
    d) Patients with severe burns may have unusually high therapeutic levels of tricyclics without experiencing toxicity.
    e) Seizures were only reported in patients with serum levels of 1000 nanograms/milliliter or greater in a study of 187 cases (Lavoie et al, 1990).
    2) CASE REPORTS
    a) A 6-year-old girl developed seizures and ECG abnormalities after taking 300 mg (15 mg/kg) of amitriptyline every night for 1 month instead of the prescribed 30 mg nightly for treatment of insomnia secondary to ADHD. Her total amitriptyline/nortriptyline concentration, obtained at hospital admission, was 1676 ng/mL (normal therapeutic, 50 to 300 ng/mL). With supportive treatment, the patient gradually recovered and was discharged, without apparent sequelae, approximately 7 days post-admission (Clement et al, 2012)
    b) FATALITIES/POSTMORTEM -
    1) Lethal tricyclic antidepressant levels reported from forensic studies have ranged from 4000 to 80000 nanomoles/liter (1100 to 21800 nanograms/milliliter) (Frommer et al, 1987).
    2) PARENT/METABOLITE RATIO: Apple (1989) studied 13 postmortem cases of tricyclic overdose and compared them to therapeutic cases. In overdose, he found the ratio between parent drug and major metabolite in the liver to be 6.3:1 and 0.5:1 in therapeutic cases (Apple, 1989).
    3) CASE SERIES: In a review of 24 tricyclic antidepressant- related deaths, a heart blood level of 0.1 milligram/deciliter was suggested as an indicator of lethality.
    4) POSTMORTEM TCA LEVELS: There are isolated cases that suggest that postmortem TCA concentrations can increase. Additional research is needed to clarify the significance and frequency of this occurrence (Hanzlick, 1989).
    5) AMITRIPTYLINE: A case of fatal amitriptyline poisoning yielded the following amitriptyline levels: blood, 0.82 milligram/liter; urine, 2.58 milligrams/liter; and vitreous humor, 6.05 milligrams/liter (Tracqui et al, 1990).
    a) The ratio of amitriptyline to nortriptyline is a useful parameter in the determination of time since ingestion in forensic cases (Spiehler et al, 1988).
    6) IMIPRAMINE: A case of fatal imipramine poisoning yielded the following imipramine levels: CSF, 1.41 milligrams/liter; vitreous humor, 1.86 milligrams/liter; urine, 20 milligrams/liter (Pounder & Jones, 1990).
    7) IMIPRAMINE: A 6-year-old boy, who was taking imipramine 200 mg/day for 3 months, developed ventricular fibrillation leading to cardiac arrest and died. His antemortem plasma tricyclic antidepressant (TCA) level was 1000 ng/mL (imipramine plus desipramine) and his postmortem TCA level, obtained 20 hours after the patient died, was 6,500 ng/mL. The difference between the antemortem and postmortem TCA levels are attributed to postmortem re-equilibration between the tissue compartments and the central compartment (Preskorn, 2011).
    8) NORTRIPTYLINE: An unusually high heart blood concentration of nortriptyline, 86.4 milligrams/liter, was found in an adult who died from an overdose of an unknown amount (possibly as much as 3.75 grams) (Rohrig & Prouty, 1989).
    9) TRIMIPRAMINE (Postmortem): A case of fatal trimipramine-citalopram-zolpidem intoxication yielded the following levels in femoral blood: trimipramine (2.33 milligrams/liter), citalopram (4.81 milligrams/liter) and zolpidem (0.07 milligram/liter) (Musshoff et al, 1999). Trimipramine levels greater than 1.0 milligram/liter are considered toxic and greater than 6 milligrams/liter is usually fatal (therapeutic blood or plasma concentrations: 0.01-0.24 milligram/liter).
    10) DOTHIEPIN (Postmortem): A fatal intoxication due to an unknown quantity of Harmomed(R) (dothiepin and diazepam) capsules has been reported. The peripheral blood concentrations of dothiepin and desmethyldothiepin were 7.28 milligrams/liter and 2.03 milligrams/liter, respectively (Keller et al, 2000).
    3) ROUTE OF EXPOSURE
    a) BURNS: Patients with severe burns may have unusually high therapeutic levels of tricyclics without experiencing toxicity. One patient had a plasma amitriptyline level of 480 micrograms/liter without clinical evidence of toxicity (Crompton et al, 1991).

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) Amitriptyline
    1) LD50- (ORAL)MOUSE:
    a) 305 mg/kg
    2) LD50- (ORAL)RAT:
    a) 464 mg/kg
    B) Dothiepin
    1) LD50- (ORAL)MOUSE:
    a) 320 mg/kg
    2) LD50- (ORAL)RAT:
    a) 450 mg/kg
    C) Imipramine
    1) LD50- (ORAL)MOUSE:
    a) 660 mg/kg
    2) LD50- (ORAL)RAT:
    a) 518 mg/kg
    D) References: Dorman, 1985
    1) LD50- (ORAL)MOUSE:
    a) >2500 mg/kg
    2) LD50- (ORAL)RAT:
    a) 5580 mg/kg

Toxicologic Mechanism

    A) Cyclic antidepressants, notably the tricyclics, are structurally similar to the phenothiazines with similar anticholinergic, adrenergic, and alpha-blocking properties of the phenothiazines (Frommer et al, 1987).
    B) Following absorption, these agents are extensively bound to plasma proteins and also bind to tissue and cellular sites, including the mitochondria (Frommer et al, 1987).
    C) Cyclic antidepressants block the neuronal reuptake of norepinephrine, serotonin, and dopamine.
    1) Golden et al (1988) found that 6-hydroxymelatonin excretion increased following antidepressant treatment, and whole-body norepinephrine turnover was reduced (Golden et al, 1988).
    D) Therapeutic doses initially may cause drowsiness and difficulty concentrating and thinking; dulling of depressive ideation may explain the efficacy of these agents in depressive disorders. Hallucinations, excitement, and confusion have occurred in a small percentage of patients during antidepressant therapy.
    E) These agents also appear to have a slight alpha-adrenergic blocking effect.
    F) Cyclic antidepressants are very lipophilic and significantly bound to proteins; (the blood tissue ratio varies from 1:10 to 1:30) which explains the ineffectiveness of forced diuresis and dialysis techniques in removal of the drug (Frommer et al, 1987).
    G) It has been postulated that tricyclics exert their major toxicity via a non-specific membrane-stabilizing effect, similar to other drugs such dextropropoxyphene, chlorpromazine, and the beta-blockers (Henry & Cassidy, 1986).
    H) ADENOSINE RECEPTORS: A study, involving rats, was conducted to determine if adenosine receptors contribute to the cardiotoxicity induced by amitriptyline. The mean arterial pressure of rats who were given an amitriptyline infusion followed by a continuous infusion of either a selective adenosine (A1) antagonist (DPCPX) or a selective adenosine (A2a) antagonist (CSC), increased significantly more than in the control group (rats who were given a continuous dextrose infusion). The adenosine antagonist infusions also resulted in a significant improvement in QRS prolongation as compared with the control group. Pretreatment with adenosine antagonists prevented amitriptyline-induced hypotension and QRS widening, and increased survival times. Adenosine receptors appear to play a role in the development of amitriptyline-induced cardiotoxicity and administration of adenosine receptor antagonists may be helpful in reducing or preventing amitriptyline-induced cardiotoxicity; however, further studies are warranted (Kalkan et al, 2004).
    I) CARDIOTOXICITY: An in vivo study, involving rats, determined that administration of a single dose of clomiPRAMINE resulted in oxidative stress, as evidenced by an increase in myocardial lipid peroxides levels and a significant decrease in myocardial reduced glutathione levels, which may contribute to clomiPRAMINE-induced cardiotoxicity. An in vitro study also showed that clomiPRAMINE administration induced a significant increase in hydroxyl radical generation leading to oxidative damage of deoxyribose. Addition of a hydroxyl radical scavenger or iron chelator appeared to significantly counteract the effect of clomiPRAMINE on hydroxyl radical generation (El-Demerdash & Mohamadin, 2004).

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) Abramowicz M: Sudden death in children treated with a tricyclic antidepressant. Med Lett Drugs Ther 1990; 32:53.
    3) Akhtar M & Goldschlager Nora F: Brugada electrocardiographic pattern due to tricyclic antidepressant overdose. J Electrocardiol 2006; 39(3):336-339.
    4) 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.
    5) Alderman CP & Lee PC: Comment: serotonin syndrome associated with combined sertraline-amitriptyline treatment. Ann Pharmacother 1996; 30:1499-1500.
    6) Alter P, Tontsch D, & Grimm W: Doxepin-induced torsade de pointes tachycardia (letter). Ann Intern Med 2001; 135(5):384-385.
    7) American College of Medical Toxicology : ACMT Position Statement: Interim Guidance for the Use of Lipid Resuscitation Therapy. J Med Toxicol 2011; 7(1):81-82.
    8) American College of Medical Toxicology: ACMT position statement: guidance for the use of intravenous lipid emulsion. J Med Toxicol 2016; Epub:Epub-.
    9) Anon: American academy of pediatrics committee on drugs: transfer of drugs and other chemicals into human milk. Pediatrics 2001; 108(3):776-789.
    10) Ansel GM, Coyne K, & Arnold S: Mechanisms of ventricular arrhythmia during amitriptyline toxicity. J Cardiovasc Pharmacol 1993; 22:798-803.
    11) Ansel GM, Meimer JP, & Nelson SD: Prevention of tricyclic antidepressant-induced ventricular tachyarrhythmia by a specific bradycardic agent in a canine model. J Cardiovasc Pharmacol 1994; 24:256-260.
    12) Ansseau M, Reynolds CF, & Kupfer DJ: Central dopaminergic and noradrenergic receptor blockade in a patient with neuroleptic malignant syndrome. J Clin Psychiatry 1986; 47:320-321.
    13) Apple FS: Postmortem tricyclic antidepressant concentrations: assessing cause of death using parent drug to metabolite ratio. J Anal Toxicol 1989; 13:197-198.
    14) Appleton R: Lorazepam vs diazepam in the acute treatment of epileptic seizures and status epilepticus.. Dev Med Child Neuro 1995; 37:682-688.
    15) Aquilonius SM & Hedstrand U: The use of physostigmine as an antidote in tricyclic anti-depressant intoxication. Acta Anaesth, Scand 1978; 22:40-45.
    16) Arya B, Hirudayaraj P, & Willmer K: Myocardial infarction: a rare complication of dothiepin overdose. Int J Cardiol 2004; 96(3):493-494.
    17) Ayd FJ: Postmortem pharmacokinetics/tricyclic antidepressants. Intern Drug Ther Newsletter 1995; 30:1-4.
    18) Bailey DN & Shaw RF: Interpretation of blood and tissue concentrations in fatal self-ingested overdose involving amitriptyline: An update (1978-1979). J Anal Toxicol 1980; 4:232-236.
    19) Banahan BF Jr & Schelkun PH: Tricyclic antidepressant overdose: Conservative management in a community hospital with cost-saving implications. J Emerg Med 1990; 8:451-461.
    20) Bania TD & Chu J: Hemodynamic effect of intralipid in amitriptyline toxicity (abstract). Acad Emerg Med 2006; 13(5 Suppl 1):S177-.
    21) Barefoot JC & Williams RB: Antidepressant use and the risk of myocardial infarction. Am J Med 2000; 108:87-88.
    22) Barnes RJ, Kong SM, & Wu RWY: Electrocardiographic changes in amitriptyline poisoning. Brit Med J 1968; 3:222-223.
    23) Barry JD, Durkovich DW, & Williams SR: Vasopressin treatment for cyclic antidepressant overdose. J Emerg Med 2006; 31(1):65-68.
    24) Bayrakci B, Unal S, Erkocoglu M, et al: Case reports of successful therapeutic plasma exchange in severe amitriptyline poisoning. Ther Apher Dial 2007; 11(6):452-454.
    25) Benazzi F & Ciucci G: Neuropathy associated with tricyclic antidepressants. Int J Ger Psychiatr 1997; 12:868-869.
    26) Benitez J, Dahlqvist R, & Gustafsson LL: Clinical pharmacological evaluation of an assay kit from intoxications with tricyclic antidepressants. There Drug Monitor 1986; 8:102-105.
    27) Berkelhammer C, Kher N, & Berry C: Nortriptyline-induced fulminant hepatic failure. J Clin Gastroenterol 1995; 20:54-56.
    28) Berkovitch M, Matsui D, & Fogelman R: Assessment of the terminal 40-millisecond QRS vector in children with a history of tricyclic antidepressant ingestion. Pediatr Emerg Care 1995; 11:75-77.
    29) Bertschy G, Vandel S, & Vandel B: Fluvoxamine-tricyclic antidepressant interactions: an accidental finding. Eur J Clin Pharmacol 1991; 40:119-120.
    30) Bessen HA & Niemann JT: Improvement of cardiac conduction after hyperventilation in tricyclic antidepressant overdose. J Toxicol 1985-86; 23:537-546.
    31) Bessen HA, Niemann JT, & Haskell RJ: Effect of respiratory alkalosis in tricyclic antidepressant overdose. West J Med 1983; 139:373-376.
    32) Biederman J, Thisted RA, & Greenhill LL: Estimation of the association between desipramine and the risk for sudden death in 5- to 14-year-old children. J Clin Psychiatry 1995; 56:87-93.
    33) Biederman J: Sudden death in children treated with a tricyclic antidepressant. J Am Acad Child Adolesc Psychiatry 1991; 30:495-498.
    34) Biggs JT, Spiker DG, & Petit JM: Tricyclic antidepressant overdose. JAMA 1977; 238:135-138.
    35) Bigwood B, Galler D, Amir N, et al: Brugada syndrome following tricyclic antidepressant overdose. Anaesth Intensive Care 2005; 33(2):266-270.
    36) Birns J, Henderson K, & Bhalla A: Recreational amitriptyline toxicity mimicking basilar artery stroke. Eur J Emerg Med 2013; 20(2):139-140.
    37) Blaber MS, Khan JN, Brebner JA, et al: "Lipid rescue" for tricyclic antidepressant cardiotoxicity. J Emerg Med 2012; 43(3):465-467.
    38) Boehnert MT & Lovejoy FH: Value of the QRS duration versus the serum drug level in predicting seizures and ventricular s after an acute overdose of tricyclic antidepressants. N Engl J Med 1985; 313:474-479.
    39) Bosse GM, Barefoot JA, & Pfeifer MP: Comparison of three methods of gut decontamination in tricyclic antidepressant overdose. J Emerg Med 1995; 13:203-209.
    40) Boyer EW & Shannon M: The serotonin syndrome. N Eng J Med 2005; 352(11):1112-1120.
    41) Brackenridge RG: Myocardial damage in amitriptyline and nortriptyline poisoning. Scot Med J 1968; 13:208.
    42) Braithwaite RA, Crome P, & Dawling S: The in vitro and in vivo evaluation of activated charcoal as an absorbent of tricyclic antidepressants. Br J Clin Pharmacol 1978; 5:369.
    43) Briggs GG, Freeman RK, & Yaffe SJBriggs GG, Freeman RK, & Yaffe SJ (Eds): Drugs in Pregnancy and Lactation, 5th. Williams & Wilkins, Baltimore, MD, 1998.
    44) Brodribb TR, Downey M, & Gilbar PJ: Efficacy and adverse effects of moclobemide (letter). Lancet 1994; 343:475-476.
    45) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    46) Brown D, Winsberg BG, & Bialer I: Imipramine therapy and seizures: three children treated for hyperactive behavior disorders. Am J Psychiatry 1973; 130:210-212.
    47) Brown K, Stork C, & Cantor R: Phenochromocytoma unmasked by imipramine. J Toxicol Clin Toxicol 2001; 49(5):490.
    48) Brown TCK: Tricyclic antidepressant overdosage: experimental studies on the management of circulatory complications. Clin Toxicol 1976; 9:255-272.
    49) Brunn GJ, Keyler DE, & Pond SM: Reversal of desipramine toxicity in rats using drug-specific antibody fab' fragment: effects on hypotension and interaction with sodium bicarbonate. J Pharmacol Exp There 1992; 250:1392-1399.
    50) Buckley NA, Chevalier S, Leditschke IA, et al: The limited utility of electrocardiography variables used to predict arrhythmia in psychotropic drug overdose. Crit Care 2003; 7(5):R101-R107.
    51) Buckley NA, Dawson AH, & Whyte IM: Greater toxicity in overdose of dothiepin than of other tricyclic antidepressants. Lancet 1994; 343:159-162.
    52) Buckley NA, O'Connell DL, & Whyte IM: Interrater agreement in the measurement of QRS interval in tricyclic antidepressant overdose: implications for monitoring and research. Ann Emerg Med 1996; 28:515-519.
    53) Burckhardt D, Raeder E, & Muller V: Cardiovascular effects of tricyclic and tetracyclic antidepressants. JAMA 1978; 239:213-216.
    54) Burr W, Sandham P, & Judd A: Death after flumazenil. Br Med J 1989; 298:1713.
    55) Caksen H, Akbayram S, Odabas D, et al: Acute amitriptyline intoxication: an analysis of 44 children. Hum Exp Toxicol 2006; 25(3):107-110.
    56) Callaham M & Kassel D: Epidemiology of fatal tricyclic antidepressant ingestion: implications for management. Ann Emerg Med 1985; 14:1-9.
    57) Callaham M, Schumaker H, & Pentel P: Phenytoin prophylaxis of cardiotoxicity in experimental amitriptyline poisoning. Pharmacol Exp There 1988; 245:216-220.
    58) Callaham M: Admission criteria for tricyclic antidepressant ingestion. West J Med 1982; 137:425-429.
    59) Cano-Munoz JL, Montejo-Iglesias ML, & Yanez-Saez RM: Possible serotonin syndrome following the combined administration of clomipramine and alprazolam (letter). J Clin Psychiatry 1995; 56:122.
    60) Caravati EM & Bossart PJ: Demographic and electrocardiographic factors associated with severe tricyclic antidepressant toxicity. J Toxicol Clin Toxicol 1991; 29(1):31-43.
    61) Caravati EM, Knight HH, & Linscott MS: Esophageal laceration and charcoal mediastinum complicating gastric lavage. J Emerg Med 2001; 20:273-276.
    62) Caravati EM: The electrocardiogram as a diagnostic discriminator for acute tricyclic antidepressant poisoning. Clin Toxicol 1999; 37:113-115.
    63) Carpenter P, Gobel FL, & Hulsing DJ: Desipramine cardiac toxicity. Minnesota Med 1982; 231-234.
    64) Casarino JP: Neuropathy associated with amitriptyline. State J Med 1977; 77:2124-2126.
    65) Chakrabarti S , Fraser J , & Tsai-Goodman B : Accidental ingestion of dosulepin presenting as atrial flutter in a child. Eur J Pediatr 2010; 169(1):113-115.
    66) 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.
    67) Chamsi-Pasha H & Barnes PC: Myocardial infarction: a complication of amitriptyline overdose. Postgrad Med J 1988; 64:968-970.
    68) 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.
    69) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    70) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    71) Claassen JAHR & Gelissen HPMM: The serotonin syndrome (letter). N Eng J Med 2005; 352(23):2455.
    72) Clark S, Catt JW, & Caffery T: Rapid diagnosis and treatment of severe tricyclic antidepressant toxicity. BMJ Case Rep 2015; 2015:Epub.
    73) Clement A, Raney JJ, Wasserman GS, et al: Chronic amitriptyline overdose in a child. Clin Toxicol (Phila) 2012; 50(5):431-434.
    74) Cohen HW, Gibson G, & Alderman MH: Excess risk of myocardial infarction in patients treated with antidepressant medications: association with use of tricyclic agents. Am J Med 2000; 108:2-8.
    75) Comstock TJ, Watson WA, & Jennison TA: Severe amitriptyline intoxication and the use of charcoal hemoperfusion. Clin Pharm 1983; 2:85-88.
    76) Corrigan FM & Coulter F: Neuroleptic malignant syndrome: amitriptyline, and thioridazine (letter). Biol Psychiatry 1988; 23:317-323.
    77) Couper FJ, McIntyre IM, & Drummer OH: Detection of antidepressant and antipsychotic drugs in postmortem human scalp hair. J Forensic Sci 1995; 40:87-90.
    78) Crome P & Newman B: Fatal tricyclic antidepressant poisoning. J Roy Soc Med 1979; 72:649-653.
    79) Crome P: Antidepressant poisoning. Acta Psychiatr Scand 1983; (suppl)302:95-101.
    80) Crompton DR, Pegg SP, & Jellett LB: Altered pharmacokinetics of tricyclic antidepressants in burns. Aust N Z J Psychiatry 1991; 25:419-421.
    81) 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.
    82) Dale O & Hole A: Biphasic time-course of serum concentrations of clomipramine and desmethylclomipramine after a near-fatal overdose: report. There Drug Monit 1993; 2:148.
    83) Dale O & Hole A: Biphasic time-course of serum concentrations of clomipramine and desmethylclomipramine after a near-fatal overdose: report. Vet Hum Toxicol 1994; 36:309-310.
    84) Dart RC, Sidki A, & Sullivan JB: Ovine desipramine antibody fragments reverse desipramine cardiovascular toxicity in the rat. Ann Emerg Med 1996; 27:309-315.
    85) Dart RC, Sullivan JB Jr, & Egen N: Effect of anti-desipramine Fab on desipramine toxicity in the rat (Abstract 31). Vet Hum Toxicol 1991; 33:359.
    86) Dasgupta A, Wells A, & Datta P: False-positive serum tricyclic antidepressant concentrations using fluorescence polarization immunoassay due to the presence of hydroxyzine and cetirizine. Ther Drug Monit 2006; 28(5):662-667.
    87) De Negri M & Baglietto MG: Treatment of status epilepticus in children. Paediatr Drugs 2001; 3(6):411-420.
    88) De Roock S, Beauloye C, De Bauwer I, et al: Tako-tsubo syndrome following nortriptyline overdose. Clin Toxicol (Phila) 2008; 46(5):475-478.
    89) DeToledo JC, Haddad H, & Ramsay RE: Status epilepticus associated with the combination of valproic acid and clomipramine. Ther Drug Monit 1997; 19:71-73.
    90) DeVane CL: Cyclic antidepressants. In: Evans WE, Schentag JJ, Jusko WJ et al (eds): Applied Pharmacokinetics. Principles of Therapeutic Drug Monitoring, 2nd ed, Applied Therapeutics, Inc, Spokane, WA, 1986, pp 852-907.
    91) Deegan C & O'Brien K: Amitriptyline poisoning in a 2-year old. Paediatr Anaesth 2006; 16(2):174-177.
    92) Dick WF & Hack JB: TCA overdose (letter). Ann Emerg Med 1999; 33:6.
    93) Donmez O, Cetinkaya M, & Canbek R: Hemoperfusion in a child with amitriptyline intoxication. Pediatr Nephrol 2005; 20:105-107.
    94) Dorsey ST & Biblo LA: Prolonged QT interval and torsades de pointes caused by the combination of fluconazole and amitriptyline (letter). Am J Emerg Med 2000; 18:227-229.
    95) Duke WW & Horton JP: Nortriptyline (Aventyl(R)) overdosage. South Med J 1969; 62:1348.
    96) Dziukas LJ & Vohra J: Tricyclic antidepressant poisoning. Med J Aust 1991; 154:344-350.
    97) Eiser AR, Neff MS, & Slifkin RF: Acute myoglobinuric renal failure: a consequence of the neuroleptic malignant syndrome. Arch Int Med 1982; 142:601-603.
    98) El-Demerdash E & Mohamadin AM: Does oxidative stress contribute in tricyclic antidepressants-induced cardiotoxicity?. Toxicol Lett 2004; 152(2):159-166.
    99) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    100) Ellison DW & Pentel PR: Clinical features and consequences of seizures due to cyclic antidepressant overdose. Am J Emerg Med 1989; 7:5-10.
    101) Emamhadi M, Mostafazadeh B, & Hassanijirdehi M: Tricyclic antidepressant poisoning treated by magnesium sulfate: a randomized, clinical trial. Drug Chem Toxicol 2012; 35(3):300-303.
    102) Emerman CL, Connors AF, & Burma GM: Level of consciousness as a predictor of complications following tricyclic overdose. Ann Emerg Med 1987; 16:326-330.
    103) Engels PT & Davidow JS: Intravenous fat emulsion to reverse haemodynamic instability from intentional amitriptyline overdose. Resuscitation 2010; 81(8):1037-1039.
    104) Eyer F, Stenzel J, Schuster T, et al: Risk assessment of severe tricyclic antidepressant overdose. Hum Exp Toxicol 2009; 28(8):511-519.
    105) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    106) Fasoli RA & Glauser FL: Cardiac and ECG abnormalities in tricyclic antidepressant overdose. Clin Toxocol 1981; 18:155-163.
    107) Fehr C, Grunder G, & Hiemke C: Increase in serum clomipramine concentrations caused by valproate. J Clin Psychopharmacol 2000; 20(4):493-494.
    108) Ferrer-Dufol A, Perez-Aradros C, & Murillo EC: Fatal serotonin syndrome caused by moclobemide-clomipramine overdose (letter). Clin Toxicol 1998; 36:31-32.
    109) Flaherty JJ, Cerva D, & Graff J: ARDS associated with massive imipramine overdose (letter). Am J Emerg Med 1986; 4:195-197.
    110) Flechter S, Rabey JM, & Regev I: Convulsive attacks due to antidepressant drug overdoses: case reports and discussion. Gen Hosp Psychiatry 1983; 5:217-221.
    111) Fogarty BJ & Khan K: Drug induced blistering and the plastic surgeon: a case of amitriptyline induced skin necrosis. Burns 1999; 25:768-770.
    112) Follmer CH & Lum BKB: Protective action of diazepam and sympathomimetic amines against amitriptyline-induced toxicity. J Pharmacol Exp Ther 1982; 222:424.
    113) Foulke GE & Albertson TE: QRS interval in tricyclic antidepressant overdosage: inaccuracy as a toxicity indicator in emergency settings. Ann Emerg Med 1986; 16:160-163.
    114) Fouron JC & Chicoine R: ECG changes in fatal imipramine (tofranil) intoxication. Pediatrics 1971; 48:777-781.
    115) Frank RD & Kierdorf HP: Is there a role for hemoperfusion/hemodialysis as a treatment option in severe tricyclic antidepressant intoxication?. Int J Artif Organs 2000; 23:618-623.
    116) Freeman JW, Mundy GR, & Beattie RR: CArdiac abnormalities in poisoning with tricyclic antidepressants. Br Med J 1969; 2:610-611.
    117) Frey OR, Scheidt P, & von Brenndorff AI: Adverse effects in a newborn infant breast-fed by a mother treated with doxepin. Ann Pharmacother 1999; 33:690-693.
    118) Frommer DA, Kulig KW, Marx JA, et al: Tricyclic antidepressant overdose. JAMA 1987; 257(4):521-526.
    119) Galynker II, Rosenthal RN, & Perkel C: Doxepin withdrawal mania (letter). J Clin Psychiatry 1995; 56:122-123.
    120) Gangat AE, Luiz HA, & Ibrahim NI: Tricyclic-induced acute tardive dyskinesia. A case report. S Afr Med J 1986; 71:729.
    121) Garcia-Doval I, Peteiro C, & Toribio J: Amitriptyline-induced erythema annulare centrifugum. Cutis 1999; 63:35-36.
    122) Garner EM, Kelly MW, & Thompson DF: Tricyclic antidepressant withdrawal syndrome. Ann Pharmacother 1993; 27:1068-1072.
    123) Geller E, Crome P, & Schaller MD: Risks and benefits of therapy with flumazenil (Anexate) in mixed drug intoxications. Eur Neurol 1991; 31:241-250.
    124) Ghaemi SN & Kirkwood CK: Elevation of nortriptyline plasma levels after cotreatment with paroxetine and thioridazine. J Clin Psychopharmacol 1998; 18:342-343.
    125) Giang DW & McBride MC : Lorazepam versus diazepam for the treatment of status epilepticus. Pediatr Neurol 1988; 4(6):358-361.
    126) Giles HM: Imipramine poisoning in childhood. Br Med J 1963; 844-946.
    127) Gillman PK: Ecstasy, serotonin syndrome and the treatment of hyperpyrexia (letter). MJA 1997; 167:109-111.
    128) Gillman PK: Successful treatment of serotonin syndrome with chlorpromazine (letter). MJA 1996; 165:345.
    129) Goldberg RJ & Huk M: Serotonin syndrome from trazodone and buspirone (letter). Psychosomatics 1992; 33:235-236.
    130) Goldberg RJ, Capone RJ, & Hunt JD: Cardiac complications following tricyclic antidepressant overdose. JAMA 1985; 254:1772-1775.
    131) Golden RN, Markey SP, & Risby ED: Antidepressants reduce whole-body norepinephrine turnover while enhancing 6-hydroxymelatonin output. Arch Gen Psychiatry 1988; 45:150-154.
    132) Goldman LS, Alexander RC, & Luchins DJ: Monoamine oxidase inhibitors and tricyclic antidepressants: comparison of their cardiovascular effects. J Clin Psychiatr 1986; 47:225-229.
    133) Goldstein BJ & Claghorn JL: An overview of 17 years of experience with dothiepin in the treatment of depression in Europe. J Clin Psychiatr 1980; 41:64-70.
    134) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    135) Gomez HF, Brent JA, & Munoz DC: Charcoal stercolith with intestinal perforation in a patient treated for amitriptyline ingestion. J Emerg Med 1994; 12:57-60.
    136) Goodwin DA, Lally KP, & Null DM Jr: Extracorporeal membrane oxygenation support for cardiac dysfunction from tricyclic antidepressant overdose. Crit Care Med 1993; 21:625-627.
    137) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    138) Graham PM: Successful treatment of the toxic serotonin syndrome with chlorpromazine (letter). Med J Australia 1997; 166:166-167.
    139) Granacher RP & Baldessarini RJ: Physostigmine. Arch Gen Psychiatry 1975; 32:375-380.
    140) Gravenor DS, Leclerc JR, & Blake G: Tricyclic antidepressant agranulocytosis. Can J Psychiatr 1986; 31:661.
    141) Greenblatt DJ, Koch-weser J, & Shader RI: Multiple complications and death following protriptyline overdose. JAMA 1974; 229:556-557.
    142) Grover CA, Flaherty B, Lung D, et al: Significant toxicity in a young female after low-dose tricyclic antidepressant ingestion. Pediatr Emerg Care 2012; 28(10):1066-1069.
    143) 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.
    144) Guharoy SR: Adult respiratory distress syndrome associated with amitriptyline overdose. Vet Human Toxicol 1994; 36:316-317.
    145) Guthrie RM & Lott JA: Abnormal serum creatine kinase and MB fraction following an amitriptyline overdose. J Fam Pract 1986; 22:550-555.
    146) Hagerman GA & Hanashiro PK: Reversal of tricyclic-antidepressant-induced cardiac conduction abnormalities by phenytoin. Ann Emerg Med 1981; 10:82-86.
    147) Hantson PH, Benaissa ML, & Clemessy JL: Hyperthermia complicating tricyclic antidepressant (TCA) overdose, EAPCCT XVI Int Congress, Vienna, Austria, 1994.
    148) Hanzlick R: Postmortem tricyclic antidepressant concentrations: lethal vs nonlethal levels. Am J Forensic Med Pathol 1989; 10:326-329.
    149) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    150) Harvey M & Cave G: Case report: successful lipid resuscitation in multi-drug overdose with predominant tricyclic antidepressant toxidrome. Int J Emerg Med 2012; 5(1):8-8.
    151) Harvey M & Cave G: Intralipid outperforms sodium bicarbonate in a rabbit model of clomipramine toxicity. Ann Emerg Med 2007; 49(2):178-185.
    152) Heard K, O'Malley G, & Bogdan GM: Serum and urine concentrations of tricyclic antidepressants (TCA) following administration of TCA immune Fab (ovine) (abstract). J Toxicol - Clin Toxicol 1999; 37:670.
    153) Heath A, Marin P, & Sjostrand I: Inotropic effect of prenalterol in amitriptyline poisoning. Intensive Care Med 1984; 10:209.
    154) Heath A, Wickstrom I, & Martenson E: Treatment of antidepressant poisoning with resin hemoperfusion. Human Toxicol 1982; 1:361-371.
    155) Hedges JR, Baker PB, & Tasset JJ: Bicarbonate therapy fro the cardiovascular toxicity in an animal model. J Emer Med 1985; 3:253-260.
    156) Hedges JR, Otten EJ, & Schroeder TJ: Correlation of initial amitriptyline concentration reduction with activated charcoal therapy in overdose patients. Am J Emerg Med 1987; 5:48-51.
    157) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    158) Heiser JF & Wilbert DE: Reversal of delirium induced by tricyclic antidepressant, drugs with physostigmine. Am J Psychiatry 1974; 11:1275-1277.
    159) Henry JA & Cassidy SL: Membrane stabilizing activity: a major cause of fatal poisoning. Lancet 1986; 1:1414-1417.
    160) Heyland D & Sauve M: Neuroleptic malignant syndrome without the use of neuroleptics. Can Med Assoc J 1991; 145:817-819.
    161) Hoffman JR & McElroy CR: Bicarbonate therapy for and hypotension in tricyclic antidepressant overdose. West J Med 1981; 60-64.
    162) Hoffman JR, Votey SR, & Bayer M: Effect of hypertonic sodium bicarbonate in the treatment of moderate-to-severe cyclic antidepressant overdose. Am J Emerg Med 1993; 11:336-341.
    163) Hojer J & Personne M: Endoscopic removal of slow release clomipramine bezoars in two cases of acute poisoning. Clin Toxicol (Phila) 2008; 46(4):317-319.
    164) Holger JS, Engebretsen KM, & Marini JJ: High dose insulin in toxic cardiogenic shock. Clin Toxicol (Phila) 2009; 47(4):303-307.
    165) Hon KL, Fung CK, Lee VW, et al: Neurologic and cardiovascular complications in pediatric life threatening imipramine poisoning. Curr Drug Saf 2015; 10(3):261-265.
    166) Horn E, Lach B, & Lapierre Y: Hypothalamic pathology in the neuroleptic malignant syndrome. Am J Psychiatry 1988; 145:617-620.
    167) Hughes PL & Rome JD: Cardiopulmonary collapse associated with an overdose of desipramine. Mayo Clin Proc 1984; 59:574.
    168) Hulten BA & Heath A: Clinical aspects of tricyclic antidepressant poisoning. Acta Med Scand 1983; 213:275-278.
    169) Hulten BA, Adams R, & Askenasi R: Predicting the severity of tricyclic antidepressant overdose. Clin Toxicol 1992; 30:161-170.
    170) Hulten BA, Adams R, & Askenasi R: Activated charcoal in tricyclic antidepressant poisoning. Human Toxicol 1988; 7:307-310.
    171) Hurst HE & Jarboe CH: Clinical findings, elimination pharmacokinetics, and tissue drug concentrations following a fatal amitriptyline intoxication. Clin Toxicol 1981; 18:119-125.
    172) Hursting MJ, Opheim KE, & Raisys VA: Tricyclic antidepressant-specific fab fragments alter the distribution and elimination of desipramine in the rabbit: a model for overdose treatment. J Toxicol Clin Toxicol 1989; 27:53-56.
    173) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    174) Ilett KF, Hackett LP, & Dusci LJ: Disposition of dothiepin after overdose: effects of repeated-dose activated charcoal. Ther Drug Monit 1991; 13:485-489.
    175) Isaacs AD & Carlish S: Peripheral neuropathy after amitriptyline. Br Med J 1963; 1:1739.
    176) Islek I, Degim T, Akay C, et al: Charcoal haemoperfusion in a child with amitriptyline poisoning. Nephrol Dial Transplant 2004; 19(12):3190-3191.
    177) Jackson JE & Banner W: Tricyclic antidepressant overdose: cardiovascular responses to catecholamines (abstract). Vet Hum Toxicol 1981; 23(Suppl 1):59.
    178) James LP & Kearns GL: Cyclic antidepressant toxicity in children and adolescents. J Clin Pharmacol 1995; 35:343-350.
    179) Jorens PG, Joosens EJ, & Nagler JM: Changes in arterial oxygen tension after gastric lavage for drug overdose. Hum Exp Toxicol 1991; 10:221-224.
    180) Jue SG: Desipramine - accidental poisoning (letter). Drug Intelligence & Clin Pharm 1976; 10:52-53.
    181) June R, Yunus M, & Gossman W: Neuroleptic malignant syndrome associated with nortriptyline (letter). Am J Emerg Med 1999; 17(7):736-737.
    182) Kalkan S, Aygoren O, Akgun A, et al: Do adenosine receptors play a role in amitriptyline-induced cardiovascular toxicity in rats?. J Toxicol Clin Toxicol 2004; 42(7):945-954.
    183) Karaci M, Ozcetin M, Dilsiz G, et al: Severe childhood amitriptyline intoxication and plasmapheresis: a case report. Turk J Pediatr 2013; 55(6):645-647.
    184) Karkkainen S & Neuvonen PJ: Pharmacokinetics of amitriptyline influenced by oral charcoal and urine pH. Int J Clin Pharmacol There Toxicol 1986; 24:326-332.
    185) Keller T, Schneider A, & Tutsch-Bauer E: Case report: fatal intoxication due to dothiepin. Forensic Sci Int 2000; 109:159-166.
    186) Keyler DE, Shelver WL, & Landon J: Toxicity of high doses of polyclonal drug-specific antibody fab fragments. Int J Immunopharmac 1994; 16:1027-1034.
    187) Kiberd MB & Minor SF: Lipid therapy for the treatment of a refractory amitriptyline overdose. CJEM 2012; 14(3):193-197.
    188) Kingston ME: Hyperventilation in tricyclic antidepressant poisoning. Crit Care Med 1979; 7:550-551.
    189) Kiyan S, Aksay E, Yanturali S, et al: Acute myocardial infarction associated with amitriptyline overdose. Basic Clin Pharmacol Toxicol 2006; 98(5):462-466.
    190) Kleinman ME, Chameides L, Schexnayder SM, et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Part 14: pediatric advanced life support. Circulation 2010; 122(18 Suppl.3):S876-S908.
    191) Kline JA, DeStefano AA, & Schroeder JD: Magnesium potentiates imipramine toxicity in the isolated rat heart. Ann Emerg Med 1994; 24:224-232.
    192) Kline SS, Mauro LS, & Scala-Barnett DM: Serotonin syndrome versus neuroleptic malignant syndrome as a cause of death. Clin Pharmac 1989; 8:510-514.
    193) Knudsen K & Abrahamsson J: Effects of epinephrine and norepinephrine on hemodynamic parameters and arrhythmias during a continuous infusion of amitriptyline in rats. Clin Toxicol 1993; 31:461-471.
    194) Knudsen K & Abrahamsson J: Effects of epinephrine, norepinephrine, magnesium sulfate, and milrinone on survival and the occurrence of arrhythmias in amitriptyline poisoning in the rat. Crit Care Med 1994; 22:1851-1855.
    195) Knudsen K & Abrahamsson J: Epinephrine and sodium bicarbonate independently and additively increase survival in experimental amitriptyline poisoning. Crit Care Med 1997a; 25:669-674.
    196) Knudsen K & Abrahamsson J: Magnesium sulphate in the treatment of ventricular fibrillation in amitriptyline poisoning (letter). Eur Heart J 1997; 18:881-882.
    197) Koppel C, Wiegreffe A, & Tenczer J: Clinical course, therapy, outcome and analytical data in amitriptyline and combined amitriptyline/chlordiazepoxide overdose. Hum & Experiment Toxicol 1992; 11:458-465.
    198) Korzets A, Floro S, & Ori Y: Clomipramine-induced pheochromocytoma crisis: a near fatal complication of a tricyclic antidepressant. J Clin Psychopharmacol 1997; 17:428-430.
    199) Kresse-Hermsdorf M, Muller-Oerlinghausen B, & Ibe K: Poisoning with antidepressive drugs: a five-year retrospective study. Intern J Clin Pharmacol, Ther & Toxicol 1985; 23:540-547.
    200) Kuisma MJ: Fatal serotonin syndrome with trismus (letter). Ann Emerg Med 1995; 26:108.
    201) Kulig K: Cyclic antidepressant overdose. JCP Monograph 1985; 3:19-23.
    202) Kutcher SP & Shulman KI: Desipramine-induced delirium at "subtherapeutic" concentrations. Can J Psychiatr 1985; 30:368-369.
    203) Labrosse KR & McCoy HG: Reliability of antidepressant assays: a reference laboratory perspective on antidepressant monitoring. Clin Chem 1988; 34:859-862.
    204) Langlow JR & Alarcon RD: Trimipramine-induced neuroleptic malignant syndrome after transient psychogenic polydipsia in one patient. J Clin Psychiatry 1989; 50:144-145.
    205) Langou RA, Dyke CV, & Rahan SR: Cardiovascular manifestations of tricyclic antidepressant overdose. Am Heart J 1980; 100:458-464.
    206) Lappa A, Castagna A, & Imperiale C: Near fatal case of atrio-ventricular block induced by amitriptyline at therapeutic dose (letter). Intensive Care Med 2000; 26:1399.
    207) Larkin GL, Graeber GM, & Hollingsed MJ: Experimental amitriptyline poisoning: treatment of severe cardiovascular toxicity with cardiopulmonary bypass. Ann Emerg Med 1994; 23:480-486.
    208) Larrey D, Rueff B, & Pessayre D: Cross hepatotoxicity between tricyclic antidepressants. Gut 1986; 27:726-727.
    209) Lavoie RW, Gansert GG, & Weiss RE: Value of initial ECG findings and plasma drug levels in cyclic antidepressant overdose. Ann Emerg Med 1990; 19(6):696-700.
    210) Lavonas EJ, Drennan IR, Gabrielli A, et al: Part 10: Special Circumstances of Resuscitation: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S501-S518.
    211) LeWitt PA & Forno LS: Peripheral neuropathy following amitriptyline overdose. Muscle Nerve 1985; 8:723-724.
    212) Levine M, Brooks DE, Franken A, et al: Delayed-onset seizure and cardiac arrest after amitriptyline overdose, treated with intravenous lipid emulsion therapy. Pediatrics 2012; 130(2):e432-e438.
    213) Levitt MA, Sullivan JB, & Owens SM: Amitriptyline plasma protein binding: effect of plasma pH and relevance to clinical overdose. Am J Emerg Med 1986; 4:121-125.
    214) Leys D, Pasquier F, & Lamblin MD: Acute polyradiculoneuropathy after amitriptyline overdose. Br Med J 1987; 294:608.
    215) Lheureux P, Vranckx M, & Leduc D: Flumazenil in mixed benzodiazepine/tricyclic antidepressant overdose: a placebo-0controlled study in the dog. Am J Emer Med 1992; 10:184-188.
    216) Liebelt EL, Francis PD, & Woolf AD: ECG lead a VR versus QRS interval in predicting seizures and arrhythmias in acute tricyclic antidepressant toxicity. Ann Emerg Med 1995; 26:195-201.
    217) Liebelt EL, Ulrich A, & Francis PD: Serial electrocardiogram changes in acute tricyclic antidepressant overdoses. Crit Care Med 1997; 25:1721-1726.
    218) Linakis JG: Amoxapine. Clin Tox Rev 1988; 10.
    219) Lindstrom FD, Flodmark O, & Gustafsson B: Respiratory distress syndrome and thrombotic, non-bacterial endocarditis after amitriptyline overdose. Acta Med Scand 1977; 202:203-212.
    220) Lipper B, Bell A, & Gaynor B: Recurrent hypotension immediately after seizures in nortriptyline overdose. Am J Emerg Med 1994; 12:452-453.
    221) Livingston RL, Zucker DK, & Isenberg K: Tricyclic antidepressants and delirium. J Clin Psychology 1983; 44:173-176.
    222) Ljunggren B & Bojs G: A case of photosensitivity and contact allergy to systemic tricyclic drugs, with unusual features. Contact Dermatitis 1991; 24:259-265.
    223) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    224) Mackway-Jones K: Alkalinisation in the management of tricyclic antidepressant overdose. J Accid Emerg Med 1999; 16:139-140.
    225) Magdalan J, Zawadzki M, Sloka T, et al: Suicidal overdose with relapsing clomipramine concentrations due to a large gastric pharmacobezoar. Forensic Sci Int 2013; 229(1-3):e19-e22.
    226) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    227) Manoguerra AS: Tricyclic antidepressants. Crit Care Quarterly 1982; 43-51.
    228) Marchant B, Wray R, & Leach A: Flumazenil causing convulsions and ventricular tachycardia. Br Med J 1989; 299:860.
    229) Marshall A & Moore K: Pulmonary disease after amitriptyline overdosage: a case report. Br Med J 1973; 1:716-717.
    230) Marshall L, Martin P, & Cobaugh D: Intra-nasal inhalation of desipramine with resultant toxicity (abstract). J Toxicol - Clin Toxicol 1998; 36:520.
    231) Mayron R & Ruiz E: Phenytoin: does it reverse tricyclic-antidepressant-induced cardiac conduction abnormalities?. Ann Emerg Med 1986; 15:876-880.
    232) McAlpine SB, Calabro JJ, & Robinson MD: Late death in tricyclic antidepressant overdose revisited. Ann Emerg Med 1986; 15:1349-1352.
    233) McCabe JL, Cobaugh DJ, & Menegazzi JJ: A comparison of hypertonic saline, sodium bicarbonate, and hyperventilation in severe tricyclic antidepressant overdose in swine. Vet Hum Toxicol 1993; 35:367.
    234) McCabe JL, Cobaugh DJ, & Menegazzi JJ: Experimental tricyclic antidepressant toxicity: a randomized, controlled comparison of hypertonic saline solution, sodium bicarbonate, and hyperventilation. Ann Emerg Med 1998; 32:329-333.
    235) McCabe JL, Menegazzi JJ, & Cobaugh DJ: Recovery from severe cyclic antidepressant overdose with hypertonic saline/dextran in a swine model. Acad Emerg Med 1994; 1:111-115.
    236) McDuffee AT & Tobias JD: Seizure after flumazenil administration in a pediatric patient. Pediatr Emerg Care 1995; 11:186-187.
    237) McGrady H & Rees JA: Toxicity of dothiepin in overdose (letter). Lancet 1994; 343:292-293.
    238) McIntyre IM, King CV, & Cordner SM: Postmortem clomipramine: therapeutic or toxic concentrations?. J Forensic Sci 1994; 39:486-493.
    239) McKinney PE & Rasmussen R: Reversal of severe tricyclic antidepressant-induced cardiotoxicity with intravenous hypertonic saline solution. Ann Emerg Med 2003; 42:20-24.
    240) McMahon AJ: Amitriptyline overdose complicated by intestinal pseudo-obstruction and caecal perforation. Postgrad Med J 1989; 65:948-949.
    241) McMahon TC: A clinical overview of syndromes following withdrawal of antidepressants. Hosp Comm Psychiatr 1986; 37:883-884.
    242) Meenan GM, Barlotta S, & Lehrer M: Urinary tricyclic antidepressant screening: comparison of results obtained with Abbott FPIA reagents and Syva EIA reagents. J Anal Toxicol 1990; 14:273-276.
    243) Mehta NJ & Alexandrou NA: Tricyclic antidepressant overdose and electrocardiographic changes. J Emerg Med 2000; 18(4):463-464.
    244) Meredith TJ & Vale JA: Poisoning due to psychotropic agents. Adverse Drug Reaction Acute Poisoning Rev 1985; 4:83-122.
    245) Merriam AE: Neuroleptic malignant syndrome after imipramine withdrawal (letter). J Clin Psychopharmacol 1987; 7:53-54.
    246) Merriman T & Stokes K: Small bowel obstruction secondary to administration of activated charcoal. Aust NZ J Surg 1995; 65:288-289.
    247) Milionis HJ, Skopelitou A, & Elisaf MS: Hypersensitivity syndrome caused by amitriptyline administration. Postgrad Med J 2000; 76:361-363.
    248) Mills KC: Serotonin syndrome: a clinical update. Med Toxicol 1997; 13:763-783.
    249) Ming ME, Bhawan J, & Stefanato CM: Imipramine-induced hyperpigmentation: four cases and review of the literature. J Am Acad Dermatol 1999; 40:159-166.
    250) Misri S & Sivertz K: Tricyclic drugs in pregnancy and lactation: a preliminary report. Int J Psychiatry Med 1991; 21:157-171.
    251) Mitchell WG: Status epilepticus and acute repetitive seizures in children, adolescents, and young adults: etiology, outcome, and treatment. Epilepsia 1996; 37(S1):S74-S80.
    252) Moccetti T: Cardiotoxic drugs. Schweiz Med Wschr 1973; 103:621.
    253) Molloy DW, Penner SB, & Rabson J: Use of sodium bicarbonate to treat tricyclic antidepressant-induced s in a patient with alkalosis. Can Med Assoc J 1984; 130:1457-1459.
    254) Monteban-Kooistra WE, vandenBerg MP, Tulleken JE, et al: Brugada electrocardiographic pattern elicited by cyclic antidepressants overdose. Intensive Care Med 2006; 32(2):281-285.
    255) Mordel A, Winkler E, & Almog S: Seizures after flumazenil administration in a case of combined benzodiazepine and tricyclic antidepressant overdose. Critical Care Medicine 1992; 20:1733-4.
    256) Mortensen SA: Cyclic antidepressants and cardiotoxicity. Practioner 1984; 228:1180-1183.
    257) Mullins ME, Cristofani CB, & Warden CR: Amitriptyline-associated seizures in a toddler with Munchausen-by-proxy. Pediatr Emer Care 1999; 15:202-205.
    258) Musshoff F, Schmidt P, & Madea B: Fatality caused by a combined trimipramine-citalopram intoxication. Forensic Sci Int 1999; 106:125-131.
    259) Mutlu M, Karaguzel G, Bahat E, et al: Charcoal hemoperfusion in an infant with supraventricular tachycardia and seizures secondary to amitriptyline intoxication. Hum Exp Toxicol 2011; 30(3):254-256.
    260) Myrenfors PG, Eriksson T, & Sandstedt CS: Moclobemide overdose. J Intern Med 1993; 233:113-115.
    261) Nattel S & Mittleman M: Treatment of ventricular tachyarrhythmias resulting from amitriptyline toxicity in dogs. J Pharmacoal Exp Ther 1984a; 231(2):430-435.
    262) Nattel S & Mittleman M: Treatment of ventricular tachyarrhythmias resulting from amitriptyline toxicity in dogs. J Pharmacol Exp There 1984; 231:430-435.
    263) Nattel S, Keable H, & Sasyniuk BI: Experimental amitriptyline intoxication: electrophysiologic manifestations and management. J Cardiovas Pharmacol 1984; 5:83-89.
    264) Nebinger P & Koel M: Specificity data of the tricyclic antidepressants assay by fluorescent polarization immunoassay. J Anal Toxicol 1990; 14:219-221.
    265) 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.
    266) Neuvonen PJ, Pohjola-Sintonen S, & Tacke U: Five fatal cases of serotonin syndrome after meclobemide-clomipramine overdoses (letter). Lancet 1993; 342:1419.
    267) Newton RW: Physostigmine salicylate in the treatment of tricyclic antidepressant overdosage. JAMA 1975; 231:941-943.
    268) Nimmo Smith RC & Grieve RC: Peripheral neuropathy after amitriptyline. Br Med J 1963; 2:254.
    269) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    270) Nulman I, Rovet J, Steward DE, et al: Child development following exposure to tricyclic antidepressants or fluoxetine throughout fetal life: A prospective, controlled study. Am J Psychiatry 2002; 159(11):1889-1895.
    271) Nyanda AM, Nunes MG, & Ramesh A: A simple high-performance liquid chromatography method for the quantitation of tricyclic antidepressant drugs in human plasma or serum. Clin Toxicol 2000; 38(6):631-636.
    272) O'Connor N, Greene S, Dargan P, et al: Prolonged clinical effects in modified-release amitriptyline poisoning. Clin Toxicol (Phila) 2006; 44(1):77-80.
    273) Ogawa Y, Tenderich G, Minami K, et al: Successful use of a cardiac allograft from tricyclic antidepressant intoxication. Transplantation 2003; 76(8):1239-1240.
    274) Olgun H, Yildirim ZK, Karacan M, et al: Clinical, electrocardiographic, and laboratory findings in children with amitriptyline intoxication. Pediatr Emerg Care 2009; 25(3):170-173.
    275) Patterson JF: Myoclonus caused by a tricyclic antidepressant. South Med J 1990; 83:463-465.
    276) Peberdy MA , Callaway CW , Neumar RW , et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care science. Part 9: post–cardiac arrest care. Circulation 2010; 122(18 Suppl 3):S768-S786.
    277) Pentel P & Benowitz N: Efficacy and mechanism of action of sodium bicarbonate in the treatment of desipramine toxicity in rats. J Pharmacol Exp There 1984; 230:12-19.
    278) Pentel P & Peterson CD: Asystole complicating physostigmine treatment of tricyclic antidepressant overdose. Ann Emer Med 1980; 9:588-590.
    279) Pentel P & Sioris L: Incidence of late arrhythmias following tricyclic antidepressant overdose. Clin Toxicol 1981; 18:543-548.
    280) Pentel PR & Bullock ML: Hemoperfusion for imipramine overdose: elimination of active metabolites. J Toxicol Clin Toxicol 1982; 19:239-248.
    281) Pentel PR, Kyler DE, & Brunn GJ: Redistribution of tricyclic antidepressants in rats using a drug- specific monoclonal antibody: dose-response relationship. Drug Metabolism & Disposition 1991; 19:24-28.
    282) Pentel PR, Landon J, & Sidki A: Effects of drug-specific polyclonal fab on desipramine cardiotoxicity and lethality in rats (abstract). Vet Human Toxicol 1994; 36.
    283) Pentel PR, Scarlett W, & Ross CA: Reduction of desipramine cardiotoxicity and prolongation of survival in rats with the use of polyclonal drug-specific antibody fab fragments. Ann Emerg Med 1995; 26:334-341.
    284) Peters RW, Buser GA, & Kim HJ: Tricyclic overdose causing sustained monomorphic ventricular tachycardia. Am J Cardiol 1992; 70:1226-1228.
    285) Pezzilli R, Melandri R, & Barakat B: Pancreatic involvement associated with tricyclic overdose. Ital J Gastroenterol Hepatol 1998; 30:418-420.
    286) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    287) Popli AP, Baldessarini RJ, & Cole JO: Interactions of serotonin reuptake inhibitors with tricyclic antidepressants. Arch Psychiatry 1994; 51:666-667.
    288) Postlethwaite RJ & Price DA: Amitriptyline and imipramine poisoning in children (letter). Br Med J 1974; 2:504.
    289) Pounder DJ & Jones GR: Post-mortem drug redistribution - a toxicological nightmare. Forensic Sci Int 1990; 45:253-263.
    290) Pounder DJ, Owen V, & Quigley C: Postmortem changes in blood amitriptyline concentration. Am J Forensic Med Pathol 1994; 15:224-230.
    291) Preskorn SH: Inadvertent fatal imipramine poisoning of a child: what happened to Tommy?. J Psychiatr Pract 2011; 17(2):118-123.
    292) Product Information: ATIVAN(R) injection, lorazepam injection. Baxter Healthcare Corporation, Deerfield, IL, 2003.
    293) Product Information: ELAVIL(R) oral tablets injection, amitriptyline oral tablets injection. Zeneca Pharmaceuticals, Wilmington, DE, 2000.
    294) Product Information: Lidocaine HCl intravenous injection solution, lidocaine HCl intravenous injection solution. Hospira (per manufacturer), Lake Forest, IL, 2006.
    295) Product Information: NORPRAMIN(R) oral tablets, desipramine HCl oral tablets. Sanofi-Aventis U.S. LLC (per FDA), Bridgewater, NJ, 2012.
    296) Product Information: PAMELOR(R) oral capsules, oral solution, nortriptyline hcl oral capsules, oral solution. Mallinckrodt Inc., St. Louis, MO, 2006.
    297) Product Information: PAMELOR(TM) oral capsules, oral solution, nortriptyline HCl oral capsules, oral solution. Mallinckrodt Inc. (per FDA), Hazelwood, MO, 2012.
    298) Product Information: ROMAZICON(R) IV injection, flumazenil IV injection. Roche Pharmaceuticals, Nutley, NJ, 2004.
    299) Product Information: SILENOR(R) oral tablets, doxepin oral tablets. Somaxon Pharmaceuticals, Inc., San Diego, CA, 2010.
    300) Product Information: SINEQUAN(R) oral capsule, doxepin hydrochloride oral capsule. Roerig Division, New York, NY, 2005.
    301) Product Information: TOFRANIL(R) tablets, imipramine hydrochloride tablets. Mallinckrodt Inc., St. Louis, MO, 2005.
    302) Product Information: VIVACTIL(R) tablets, protriptyline hcl tablets. Odyssey Pharmameuticals,Inc., East Hanover, NJ, 2004.
    303) Product Information: amitriptyline HCl oral tablet, amitriptyline HCl oral tablet. RemedyRepack Inc. (per DailyMed), Indiana, PA, 2010.
    304) Product Information: desipramine hcl oral tablets, desipramine hcl oral tablets. Sandoz,Inc, Princeton, NJ, 2006.
    305) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    306) Product Information: imipramine HCl oral film coated tablets, imipramine HCl oral film coated tablets. Mutual Pharmaceutical Company, Inc. (per DailyMed), Philadelphia, PA, 2012.
    307) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    308) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    309) Product Information: perphenazine and amitriptyline HCl oral tablet, perphenazine and amitriptyline HCl oral tablet. Mylan Pharmaceuticals Inc, Morgantown, WV, 2010.
    310) Product Information: trimipramine maleate oral capsules, trimipramine maleate oral capsules. Actavis Totowa,LLC, Totowa, NJ, 2006.
    311) Pulst SM & Lombroso CT: External ophthalmoplegia, alpha and spindle coma in imipramine overdose: case report and review of the literature. Ann Neurology 1983; 14(5):587-590.
    312) 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.
    313) Rasmussen R & McKinney PE: Hypertonic saline in severe tricylic antidepressant cardiotoxicity (abstract). J Toxicol Clin Toxicol 1999; 37:634.
    314) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    315) Rendoing J, Choisey H, & Pascalis G: Acute poisoning with nortriptyline. Presse Med 1969; 77:439-440.
    316) Riddle MA, Geller B, & Ryan N: Another sudden death in a child treated with desipramine. J Am Acad Child Adolesc Psychiatry 1993; 32:792-797.
    317) Riddle MA, Nelson JC, & Kleinman CS: Sudden death in children receiving Norpramin: a review of three reported cases and commentary. J Am Acad Child Adolesc Psychiatr 1991; 30:104-108.
    318) Rinder HM, Murphy JW, & Higgins GL: Impact of unusual gastrointestinal problems on the treatment of tricyclic antidepressant overdose. Ann Emerg Med 1988; 17:1079-1081.
    319) Roberge RJ & Krenzelok EP: Prolonged coma and loss of brainstem reflexes following amitriptyline overdose. Vet Human Toxicol 2001; 43(1):42-44.
    320) Roberge RJ, Martin TG, & Hodgman M: Acute chemical pancreatitis associated with a tricyclic antidepressant (clomipramine) overdose. Clin Tox 1994; 32:425-429.
    321) Robinson RF, Nahata MC, & Olshefski RS: Syncope associated with concurrent amitriptyline and fluconazole therapy. Ann Pharmacother 2000; 34:1406-1409.
    322) Rohrig TP & Prouty RW: A nortriptyline death with unusually high tissue concentrations. J Anal Toxicol 1989; 13:303-304.
    323) Romano G & Di Bono G: A fatality involving clothiapine and clomipramine. J Forensic Sci 1994; 39:877-882.
    324) Rosenbaum TG & Kou M: Are one or two dangerous? Tricyclic antidepressant exposure in toddlers. J Emerg Med 2005; 28(2):169-174.
    325) Ross JP, Small TR, & Lepage PA: Imipramine overdose complicated by toxic megacolon. Amer Surgeon 1998; 64:242-244.
    326) Rothenberg PA & Hall C: Agranulocytosis following use of imipramine hydrochloride (Tofranil). Am J Psychiatry 1960; 116:847.
    327) Roy TM, Ossorio MA, & Cipolla LM: Pulmonary complications after tricyclic antidepressant overdose. Chest 1989; 96:852-856.
    328) S Sweetman : Martindale: The Complete Drug Reference. Pharmaceutical Press. London, UK (Internet Version). Edition expires 2002; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    329) Sabouraud A, Denis H, & Urtizberea M: The effect of nortriptyline-specific active immunization on amitriptyline toxicity and disposition in the rabbit. Toxicology 1990; 62:349-360.
    330) Sanaei-Zadeh H , Shahmohammadi F , Zamani N , et al: Can death unrelated to secondary causes be predicted in intubated comatose tricyclic antidepressant-poisoned patients?. Clin Toxicol (Phila) 2011; 49(5):379-384.
    331) Sandeman DJ, Alahakoon TI, & Bentley SC: Tricyclic poisoning - successful management of ventricular fibrillation following massive overdose of imipramine. Anaesth Intens Care 1997; 25:542-545.
    332) Sasyniuk BI, Jhamandas V, & Valois M: Experimental amitriptyline intoxication: treatment of cardiac toxicity with sodium bicarbonate. Ann Emerg Med 1986; 1052-1059.
    333) Sawyer WT, Caudill JL, & Ellison MJ: A case of severe acute desipramine overdose. Am J Psychiatry 1984; 141:122-123.
    334) Scheinin M, Virtanen R, & Iisalo E: Effect of single and repeated doses of activated charcoal on the pharmacokinetics of doxepin. Intern J Clin Pharmacol Ther Toxicol 1985; 23:38-42.
    335) Schimmell MS, Katz E, & Shasg Y: Toxic neonatal effects following maternal clomipramine therapy. J Toxicol Clin Toxicol 1991; 29:479-484.
    336) Schlesinger JJ & Janz TG: The efficacy of continuous bicarbonate infusion in maintaining an alkaline pH (Abstract). Ann Emerg Med 1989; 18:916.
    337) Schlienger RG, Klink MH, & Eggenberger C: Seizures associated with therapeutic doses of venlafaxine and trimipramine. Ann Pharmacother 2000; 34:1402-1405.
    338) Schneider LS, Cooper TB, & Severson JA: Electrocardiographic changes with nortriptyline and 10-hydroxynortriptyline in elderly depressed outpatients. J Clin Psychopharmacol 1988; 6:402-408.
    339) Schwartz JG, Hurd IL, & Carnahan JJ: Determination of tricyclic antidepressants for ED analysis. Am J Emerg Med 1994; 12:513-516.
    340) 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.
    341) Sedal L, Korman MG, & Williams PO: Overdosage of tricyclic antidepressants - a report of 2 deaths and a prospective study of 24 patients. Med J Australia 1972; 2:74.
    342) Seger DL, Hantsch C, Zavoral T, et al: Variability of recommendations for serum alkalinization in tricyclic antidepressant overdose: a survey of U.S. Poison Center medical directors. J Toxicol Clin Toxicol 2003; 41:331-338.
    343) Sener EK, Gabe S, & Henry JA: Response to glucagon in imipramine overdose. Clin Toxicol 1995; 33:51-53.
    344) Sensky PR & Olczak SA: High-dose intravenous glucagon in severe tricyclic poisoning. Postgrad Med J 1999; 75:611-612.
    345) Sert A, Aypar E, Odabas D, et al: Temporary cardiac pacemaker in the treatment of junctional rhythm and hypotension due to imipramine intoxication. Pediatr Cardiol 2011; 32(4):521-524.
    346) Shannon M & Lovejoy FH: Pulmonary consequences of severe tricyclic antidepressant ingestion. Clin Toxicol 1987; 25:443-461.
    347) Shannon M: Serum enzyme disturbances after tricyclic antidepressant overdose. Vet Hum Toxicol 1989; 31:171-172.
    348) Shannon MW, Merola J, & Lovejoy FH Jr: Hypotension in severe tricyclic antidepressant overdose. Am J Emerg Med 1988; 6:439-442.
    349) Shannon MW: Duration of QRS disturbances after severe tricyclic antidepressant intoxication. Clin Toxicol 1992; 30:377-386.
    350) Shanon M & Liebelt EL: Toxicology reviews: targeted management strategies for cardiovascular toxicity from tricyclic antidepressant overdose: the pivotal role for alkalinization and sodium loading. Ped Emerg Care 1998; 14:293-298.
    351) Shearer WT, Schreiner RL, & Marshall RE: Urinary retention in a neonate secondary to maternal ingestion of nortriptyline. J Pediatrics 1972; 31:270571.
    352) Shelver WL, Keyler DE, & Lin G: Recombinant drug-specific single chain antibody Fv fragment redistributes (3)h-desipramine in rats (abstract). J Toxicol Clin Toxicol 1995; 33:559-560.
    353) Shenouda R & Desan PH: Abuse of tricyclic antidepressant drugs: a case series. J Clin Psychopharmacol 2013; 33(3):440-442.
    354) Shrand H: Agoraphobia and imipramine withdrawal? (Letter). Pediatrics 1982; 70:825.
    355) Shrivastava RK & Itil TM: Flu-like illness after discontinuance of imipramine. Biol Psychiatr 1985; 20:792-794.
    356) Sirota P, Ori J, & Schild K: Electrocardiographic effects of amitriptyline in therapeutic doses on 24-hr monitoring (Holter) with correlation to plasma levels. Biol Psychiatry 1990; 27:1053-1056.
    357) Sjoqvist F, Bergfors PG, & Borga O: Plasma disappearance of nortriptyline in a newborn infant following placental transfer from an intoxicated mother: evidence for drug metabolism. J Pediatrics 1972; 80:496-500.
    358) Slovis TL, Ott JE, & Teitelbaum DT: Physostigmine therapy in acute tricyclic antidepressant poisoning. Clin Toxicol 1971; 4:451-459.
    359) Smith DB & Tyznik JW: Desipramine-induced conduction disorder mimicking myocardial infarction. Postgrad Med 1987; 82:86-88.
    360) Sorisky A & Watson DC: Positive diphenhydramine interference in the EMIT-st assay for tricyclic antidepressants in serum (letter). Clin Chem 1986; 32:715.
    361) Spiehler V, Spiehler E, & Osselton MD: Application of expert systems analysis to interpretation of fatal cases involving amitriptyline. J Anal Toxicol 1988; 12:216-224.
    362) Spigset O, Mjorndal T, & Lovheim O: Serotonin syndrome caused by a moclobemide-clomipramine interaction (letter). Br Med J 1993; 306.
    363) Spiller HA & Rogers GC: Evaluation of administration of activated charcoal in the home. Pediatrics 2002; 108:E100.
    364) Spiller HA, Baker SD, Krenzelok EP, et al: Use of dosage as a triage guideline for unintentional cyclic antidepressant (UCA) ingestions in children. Am J Emerg Med 2003; 21(5):422-424.
    365) Squires LA: . Hyperpyrexia in an adolescent on desipramine treatment. Clin Pediat 1992; 31:635-636.
    366) 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.
    367) Stancer HC & Reed KL: Desipramine and 2-hydroxydesipramine in human breast milk and the nursing infant's serum. Am J Psychiatr 1986; 143:1597-1600.
    368) Steeds RP & Muthusamy R: Abnormal ventricular conduction following dothiepin overdose simulating acute myocardial infarction. Heart 2000; 83(3):289.
    369) Steele TE & Ashby J: Desipramine-related slate-gray skin pigmentation. J Clin Psychopharm 1993; 13:76-77.
    370) Stern TA, O'Gara PT, & Mulley AG: complications after overdose with tricyclic antidepressants. Crit Care Med 1985; 13:672-674.
    371) Sternbach H: The serotonin syndrome. Am J Psychiatr 1991; 148:705-713.
    372) Stewart GO: Convulsions after physostigmine (letter). Anaesth Intens Care 1979; 7:283.
    373) Stinnett JL: Nortriptyline hydrochloride overdosage. JAMA 1968; 204:69.
    374) Stone CK, Kraemer CM, & Carroll R: Does a sodium-free buffer affect QRS width in experimental amitriptyline overdose?. Ann Emerg Med 1995; 26:58-64.
    375) Suhara T, Sudo Y, & Yoshida K: Lung as reservoir for antidepressants in pharmacokinetic drug interactions. Lancet 1998; 351:332-335.
    376) Sunderragan S, Brooks CS, & Sunderrajan EV: Nortriptyline-induced severe hyperventilation. Arch Intern Med 1985; 145:746-747.
    377) Swanson-Biearman B, Goetz CM, & Dean BS: Anafronil overdose: a fatal outcome. Vet Hum Toxicol 1989; 31:378.
    378) Swartz CM & Sherman A: The treatment of tricyclic antidepressant overdose with repeated charcoal. J Clin Psychopharmacol 1984; 4:336-340.
    379) Taboulet P, Michard F & Muszynski J et al: Cardiovascular repercussions of seizures during cyclic antidepressant poisoning. EAPCCT XVI Int Congress, Vienna, 1994.
    380) Taniguchi S & Hamada T: Photosensitivity and thrombocytopenia due to amitriptyline (letter). Am J Hematol 1996; 53:49-56.
    381) Tatli O, Karaca Y, Gunaydin M, et al: Cerebellitis developing after tricyclic antidepressant poisoning. Am J Emerg Med 2013; 31(9):1419.e3-1419.e5.
    382) Teba L, Schiebel F, & Dedhia HV: Beneficial effect of norepinephrine in the treatment of circulatory shock caused by tricyclic antidepressant overdose. Am J Emerg Med 1988; 6:566-568.
    383) Thakore S & Murphy N: The potential role of prehospital administration of activated charcoal. Emerg Med J 2002; 19:63-65.
    384) Thomson JS, Donald C, & Lewin K: Use of Flumazenil in benzodiazepine overdose. Emerg Med J 2006; 23(2):162-.
    385) Tingelstad JB: The cardiotoxicity of the tricyclics. J Am Acad Child Adolesc Psychiatry 1991; 30:845-846.
    386) Toerne T, Dardis K, & Kanter M: Doxepin toxicity in a child from Zonalon(R) topical cream. J Tox-Clin Tox 1997; 35:496.
    387) Tokarski GF & Young MJ: Criteria for admitting patients with tricyclic antidepressant overdose. J Emerg Med 1988; 6:121-124.
    388) Tracqui A, Kintz P, & Ritter-Lohner S: Toxicological findings after fatal amitriptyline self-poisoning. Hum Exp Toxicol 1990; 9:257-261.
    389) Tran TP, Panacek EA, & Rhee KJ: Response to dopamine vs norepinephrine in tricyclic antidepressant-induced hypotension. Acad Emerg Med 1997; 4:864-868.
    390) Tribble J, Weinhouse E, & Garland J: Treatment of severe imipramine poisoning complicated by a negative history of drug ingestion. Pediatr Emerg Care 1989; 5:234-237.
    391) Trouve R, Sitbon M, & Latour C: Protective action of flunarizine in imipramine poisoning (Fre). J Toxicol Clin Exper 1986; 6:197-208.
    392) Vale JA, Kulig K, American Academy of Clinical Toxicology, et al: Position paper: Gastric lavage. J Toxicol Clin Toxicol 2004; 42:933-943.
    393) 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.
    394) Van der Kuy PH, Hooymans PM, & Verkaaik AJ: Nortriptyline intoxication induced by terbinafine (letter). Brit Med J 1998; 316:441.
    395) Varley CK & McClellan J: Case study: two additional sudden deaths with tricyclic antidepressants. J Am Acad Child Adolesc Psychiatr 1997; 36:390-394.
    396) Varnell RM, Godwin JD, & Richardson ML: Adult respiratory distress syndrome from overdose of tricyclic antidepressants. Radiology 1989; 170:667-670.
    397) Vernon DD, Banner W Jr, & Garrett JS: Efficacy of dopamine and norepinephrine for treatment of hemodynamic compromise in amitriptyline intoxication. Crit Care Med 1991; 19:544-549.
    398) Vexiau P, Gourmel B, & Julien R: Severe acne-like lesions caused by amineptine overdose. Lancet 1988; 1:585.
    399) Vohra J: Intracardiac conduction defects following overdose of tricyclic antidepressant drugs. Eur J Cardiol 1975; 2:453.
    400) Wallace DE: Bowel ischemia in two patients following tricyclic antidepressant overdose (Abstract 192). Vet Hum Toxicol 1989; 31:377.
    401) Walter DC & Kauffman RE: Doxepin poisoning in a child. Am J Dis Child 1980; 134:202-203.
    402) Watson WA, Leighton J, & Guy J: Recovery of cyclic antidepressants with gastric lavage. J Emerg Med 1989; 7:373-377.
    403) Weintraub D: Nortriptyline toxicity secondary to interaction with bupropion sustained-release. Depress Anxiety 2001; 13(1):50-52.
    404) Wertelecki W, Purvis-Smith SG, & Blackburn WR: Amitriptyline/perphenazine maternal overdose and birth defects (abstract). Teratology 1980; 21:74A.
    405) Wheless JW : Treatment of status epilepticus in children. Pediatr Ann 2004; 33(6):376-383.
    406) White A: Overdose of tricyclic antidepressants associated with absent brainstem reflexes. Can Med Assoc J 1988; 139:133-134.
    407) Wians FH & Norton JT: False-positive serum tricyclic antidepressant screen with cyproheptadine (letter). Clin Chemistry 1993; 39:1355-1356.
    408) Wilens TE, Stern TA, & O'Gara PT: Adverse cardiac effects of combined neuroleptic ingestion and tricyclic antidepressant overdose. J Clin Psychopharmacol 1990; 10:51-54.
    409) Williams JM, Hollingshed MJ, & Vasilakis A: Extracorporeal circulation in the management of severe tricyclic antidepressant overdose. Am J Emerg Med 1994; 12:456-458.
    410) Williams JO: Respiratory depression in tricyclic overdose. Br Med J 1972; 1:631.
    411) Winrow AP: Amitriptyline-associated seizures in a toddler with Munchausen-by-proxy (letter). Pediatr Emerg Care 1999; 14(6):462-463.
    412) Wirtheim E & Bloch Y: Dibenzepin overdose causing pulmonary edema. Ann Pharmacother 1996; 30:789-790.
    413) Wohlreich MM & Welch W: Amitriptyline abuse presenting as acute toxicity. Psychosomatics 1993; 34:191-192.
    414) Wolfe TR, Caravati EM, & Rollins DE: Terminal 40-ms frontal plane QRS axis as a marker for tricyclic antidepressant overdose. Ann Emerg Med 1989; 18:348-351.
    415) Woodhead R: Cardiac rhythm in tricyclic antidepressant poisoning. Clin Toxicol 1979; 14:499-505.
    416) Woolf AD, Erdman AR, Nelson LS, et al: Tricyclic antidepressant poisoning: an evidence-based consensus guideline for out-of-hospital management. Clin Toxicol (Phila) 2007; 45(3):203-233.
    417) Wrenn K, Smith BA, & Slovis CM: Profound alkalemia during treatment of tricyclic antidepressant overdose: a potential hazard of combined hyperventilation and intravenous bicarbonate. Am J Emerg Med 1992; 10:553-555.
    418) Yates C, Galvao T, Sowinski KM, et al: Extracorporeal treatment for tricyclic antidepressant poisoning: recommendations from the EXTRIP Workgroup. Semin Dial 2014; 27(4):381-389.
    419) Yoav G, Odelia G, & Shaltiel C: A lipid emulsion reduces mortality from clomipramine overdose in rats. Vet Hum Toxicol 2002; 44(1):30-.
    420) Yuan CM, Spandorfer PR, Miller SL, et al: Evaluation of tricyclic antidepressant false positivity in a pediatric case of cyproheptadine (periactin) overdose. Ther Drug Monit 2003; 25(3):299-304.
    421) Zakynthinos E, Vassilakopoulos T, & Roussos C: Abnormal atrial and ventricular repolarisation resembling myocardial injury after tricyclic antidepressant drug intoxication. Heart 2000; 83:353-354.
    422) Zell-Kanter M, Toerne TS, & Spiegel K: Doxepin toxicity in a child following topical administration. Ann Pharmacother 2000; 34:328-329.
    423) de Caen AR, Berg MD, Chameides L, et al: Part 12: Pediatric Advanced Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S526-S542.