MOBILE VIEW  | 

VENLAFAXINE AND RELATED AGENTS

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Venlafaxine and desvenlafaxine (the major metabolite of venlafaxine) are potentiators of neurotransmitter activity in the CNS. They are potent inhibitors of neuronal serotonin and norepinephrine reuptake and weak inhibitors of dopamine reuptake, without inhibiting monoamine oxidase for the treatment of depression.

Specific Substances

    A) VENLAFAXINE
    1) Wy-45030
    2) Molecular Formula: C17-H27-N-O2.Cl-H
    3) CAS 93413-69-5 (venlafaxine)
    4) CAS 99300-78-4 (venlafaxine hydrochloride)
    DESVENLAFAXINE
    1) Desvenlafaxine succinate
    2) DVS
    3) Molecular Formula: C16-H25-NO2 (free base)

    1.2.1) MOLECULAR FORMULA
    1) DESVENLAFAXINE: C16H25NO2
    2) DESVENLAFAXINE SUCCINATE MONOHYDRATE: C16H25NO2.C4H6O4.H2O
    3) VENLAFAXINE HYDROCHLORIDE: C17H27NO2 HCl

Available Forms Sources

    A) FORMS
    1) Venlafaxine is commercially available as either an immediate-release tablet (25 mg, 37.5 mg, 50 mg, 75 mg, and 100 mg) and extended-release capsules of 37.5 mg, 75 mg, and 150 mg (Prod Info Effexor(R) oral tablets, 2010; Prod Info Effexor XR (R) extended-release oral capsules, 2010).
    2) Desvenlafaxine is available as 50 mg (light pink) and 100 mg (reddish-orange) square pyramid tablets. Each tablet contains 76 or 152 mg of desvenlafaxine succinate equivalent to 50 or 100 mg of desvenlafaxine, respectively (Prod Info PRISTIQ(TM) oral extended-release tablets, 2008).
    B) SOURCES
    1) POTENTIAL MISUSE: Venlafaxine has been found to be widely available for purchase online without a prescription. It was noted that almost half of the online pharmacies did not require a medical prescription (Francesconi et al, 2015).
    C) USES
    1) Venlafaxine is indicated to treat major depressive disorder (Prod Info Effexor(R) oral tablets, 2010).
    2) Venlafaxine extended-released is indicated to treat major depressive disorder, generalized anxiety disorder, social anxiety disorder, and panic disorder (Prod Info Effexor XR (R) extended-release oral capsules, 2010). Venlafaxine has also been used for fibromyalgia, tension-type headache, migraine prophylaxis, and cocaine dependence based on off-label use (Francesconi et al, 2015).
    3) Desvenlafaxine, a major metabolite of venlafaxine, is also indicated for use in the treatment of major depressive disorder, generalized anxiety disorder, social anxiety disorder and panic disorder (Prod Info PRISTIQ(TM) oral extended-release tablets, 2008).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Venlafaxine and desvenlafaxine are primarily used to treat anxiety and depressive disorders. Off-label uses for venlafaxine include treatment of ADHD, eating disorders, chronic pain, headache, and agoraphobia. Extended release formulations of both drugs are available.
    B) PHARMACOLOGY: Venlafaxine and desvenlafaxine inhibit neuronal reuptake of both serotonin and norepinephrine in the CNS.
    C) TOXICOLOGY: Increases in serotonin, norepinephrine, and dopamine can lead to cardiovascular toxicity (due to sodium and potassium channel blockade), and seizures. Venlafaxine and desvenlafaxine are highly serotonergic and have been implicated in causing serotonin syndrome in therapeutic doses or (rarely) in overdose, as single agents or in combination with other serotonergic agents.
    D) EPIDEMIOLOGY: Poisoning is common and several deaths occur each year.
    E) WITH THERAPEUTIC USE
    1) ADVERSE EVENTS: Mild effects include nausea/vomiting, dry mouth, constipation, loss of appetite, asthenia, dizziness, headache, insomnia, somnolence, blurred vision, nervousness and tremor following therapeutic venlafaxine therapy. The most commonly reported adverse events reported with desvenlafaxine were similar to venlafaxine. The following adverse effects have also rarely occurred with therapeutic doses of these agents: hyponatremia, hypochloremia, hepatitis, seizures, serotonin syndrome (generally if used with another serotonergic agent), thrombocytopenia, exacerbation of congestive heart failure, and elevated blood pressure.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Mild serotonergic effects include tremor, hyperreflexia, anxiety, and agitation. Palpitations, tachycardia and exacerbation of congestive heart failure have been reported with both therapeutic use and overdose. Somnolence is common in mild toxicity though delirium is more likely as toxicity increases.
    2) SEVERE TOXICITY: Serotonin syndrome (altered mental status, neuromuscular abnormalities/rigidity, autonomic instability) is a severe manifestation of overdose. Cardiovascular toxicity includes, PR, QRS, and QT prolongation occasionally culminating in ventricular tachycardia, fibrillation, and cardiac arrest. Interval prolongation is common though ventricular dysrhythmias are rare and are associated with large overdose (greater than 8 g). Seizures are common in patients taking over 1 g. Seizures have been reported in neonates exposed in utero from mothers taking therapeutic doses. Rhabdomyolysis is common in severe toxicity. Development of eosinophilic pneumonia was reported in a young adult following overdose. Coma is a rare effect of severe toxicity.
    0.2.5) CARDIOVASCULAR
    A) WITH POISONING/EXPOSURE
    1) Venlafaxine has caused hypotension, hypertension, palpitations, tachycardia, acute heart failure, cardiac arrest, and prolongation of the QRS and QT intervals. Doses of 8 g or greater are more likely to produce cardiac toxicity. Tachycardia and mild hypotension are relatively common adverse events. Fatalities have been reported after acute exposure.
    0.2.6) RESPIRATORY
    A) WITH THERAPEUTIC USE
    1) Interstitial pneumonitis has rarely been reported with therapeutic use of venlafaxine.
    0.2.7) NEUROLOGIC
    A) WITH THERAPEUTIC USE
    1) Hypomanic symptoms have been reported in patients taking venlafaxine for depression.
    B) WITH POISONING/EXPOSURE
    1) CNS depression, headache, fatigue, dizziness, nervousness, tremors, extrapyramidal effects, and serotonin syndrome have occurred as adverse effects following venlafaxine ingestion.
    2) Somnolence and generalized seizures have been reported following overdoses of venlafaxine.
    0.2.8) GASTROINTESTINAL
    A) WITH THERAPEUTIC USE
    1) Venlafaxine therapeutic use has been associated with nausea, vomiting, dry mouth, and constipation.
    B) WITH POISONING/EXPOSURE
    1) Bowel perforation occurred in one fatal case of intentional venlafaxine overdose. Gastric bezoar has been reported following overdose with a sustained-release product.
    0.2.10) GENITOURINARY
    A) WITH THERAPEUTIC USE
    1) Sexual dysfunction has been reported in a small number of patients receiving therapeutic doses of venlafaxine.
    0.2.12) FLUID-ELECTROLYTE
    A) WITH THERAPEUTIC USE
    1) Hyponatremia and hypochloremia were reported following venlafaxine therapy. Anasarca has also been reported.
    0.2.13) HEMATOLOGIC
    A) WITH THERAPEUTIC USE
    1) Abnormal platelet function has been reported with therapeutic use.
    0.2.14) DERMATOLOGIC
    A) WITH THERAPEUTIC USE
    1) Sweating has occurred with venlafaxine use.
    2) Rare cases of Stevens-Johnson syndrome and psoriasiform rashes have been reported with venlafaxine use.
    0.2.17) METABOLISM
    A) WITH THERAPEUTIC USE
    1) Venlafaxine has been associated with weight loss and an increase in serum cholesterol levels.
    0.2.20) REPRODUCTIVE
    A) Venlafaxine and desvenlafaxine are classified as FDA pregnancy category C. Current data does not demonstrate an increased risk of major malformations following venlafaxine use in pregnancy. However, neonates exposed to serotonin and norepinephrine reuptake inhibitors (SNRIs) or SSRIs late in the third trimester have developed serious complications that could be indicative of a toxic effect of the drug, a drug discontinuation syndrome, and in some cases, findings were consistent with serotonin syndrome. An increased risk for social-behavioral abnormalities at 2 to 6 years of age was reported in children exposed to SSRIs or SNRIs in utero who developed neonatal abstinence syndrome (NAS) at birth. Venlafaxine and its active metabolite, desvenlafaxine, are excreted in human breast milk; however, no adverse effects have been noted in nursing infants. Neither venlafaxine nor desvenlafaxine affected fertility in rats at doses that were close to the human dose.
    0.2.21) CARCINOGENICITY
    A) At the time of this review, the manufacturer does not report any carcinogenic potential in humans.
    0.2.22) OTHER
    A) DRUG INTERACTION: A drug interaction may occur between cimetidine and venlafaxine resulting in a reduction of venlafaxine clearance and an increase in the peak serum concentration of venlafaxine. Monoamine oxidase inhibitors may interact with venlafaxine resulting in a serotonin syndrome or seizures.
    B) WITH THERAPEUTIC USE
    1) WITH THERAPEUTIC USE
    a) A withdrawal syndrome was reported following the rapid reduction of venlafaxine therapy.

Laboratory Monitoring

    A) Monitor vital signs, institute continuous cardiac monitoring and obtain an ECG.
    B) In moderate to severe toxicity serum electrolytes (including glucose), CBC, and creatinine kinase should be obtained.
    C) EEG monitoring may be necessary for patients that require paralysis.
    D) Monitor glucose following a significant exposure. Infrequent reports of prolonged/recurrent hypoglycemia have occurred following overdose.
    E) Venlafaxine may cause false positive results for phencyclidine or tramadol on drug screening assays.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Supportive care is the mainstay of mild/moderate toxicity. Benzodiazepines are the treatment of choice for agitation, mild serotonergic effects, and hyperadrenergic vital signs. Treat mild hypotension with intravenous fluids.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Benzodiazepines should be used for seizures and agitation; often large doses are necessary. Patients with agitation, tachycardia, fever, or seizures that are unresponsive to benzodiazepines should be intubated and sedated with propofol or barbiturates. Neuromuscular paralysis with continuous EEG monitoring should be considered if hyperthermia persists despite adequate sedative administration. Isotonic fluids should be given to replace large insensible losses, facilitate myoglobin clearance, and promote renal clearance of the agent. Severe hypotension may require vasopressors, if hypotension persists consider insulin and dextrose infusion, or intravenous fat emulsion therapy. Consider extracorporeal support (cardiopulmonary bypass, extracorporeal membrane oxygenation, aortic balloon pump) in patients who remain unstable despite maximal medical therapy.
    C) DECONTAMINATION
    1) PREHOSPITAL: In general, there is no role for prehospital decontamination because of the risk of seizures and subsequent aspiration.
    2) HOSPITAL: Activated charcoal should be considered in asymptomatic or minimally symptomatic patients who present within a few hours of ingestion, or in symptomatic patients who have a secure airway. Gastric lavage may be considered for large overdoses who present within approximately 1 hour of ingestion, though airway protection should be considered prior to the procedure. Gastroscopy has been used rarely to remove extended release formulations from the stomach.
    3) COMBINED THERAPY: The combination of single-dose activated charcoal and whole bowel irrigation has been shown to decrease peak blood concentrations and may be beneficial in decreasing the risk of seizures and cardiovascular toxicity, following airway management, in a large overdose predominated by CNS depression.
    D) AIRWAY MANAGEMENT
    1) Though respiratory depression is not a manifestation of toxicity, airway protection may be necessary in patients with profound agitation, seizures, or cardiac dysrhythmias.
    E) ANTIDOTE
    1) There is no known antidote.
    F) SEIZURE
    1) Seizures and agitated delirium should be treated with benzodiazepines; large doses may be necessary. Diazepam: ADULT: 5 to 10 mg repeat every 10 to 15 minutes as needed. CHILD: 0.2 to 0.5 mg/kg, repeat every 5 minutes as needed; or Lorazepam: ADULT: 2 to 4 mg; CHILD: 0.05 to 0.1 mg/kg. If unresponsive to benzodiazepines, propofol or barbiturates can be used.
    G) HYPOTENSIVE EPISODE
    1) Treat with isotonic fluids. If unresponsive to 2 liters (or 40 mL/kg in children) direct acting vasopressor agents should be used (norepinephrine or phenylephrine). If hypotension persists consider insulin and dextrose infusion, or intravenous fat emulsion therapy.
    2) INSULIN
    a) Administer a bolus of 1 unit/kg of insulin followed by an infusion of 0.1 to 1 unit/kg/hr, titrated to a systolic blood pressure of greater than 90 to 100 mmHg (bradycardia may or may not respond). Reassess every 30 minutes to titrate insulin infusion. Administer dextrose bolus to patients with an initial blood glucose of less than 250 mg/dL (adults 25 to 50 mL dextrose 50%, children 0.25 g/kg dextrose 25%). Begin a dextrose infusion of 0.5 g/kg/hr in all patients. Monitor blood glucose every 15 to 30 minutes until consistently 100 to 200 mg/dL for 4 hours, then monitor every hour. Titrate dextrose infusion to maintain blood glucose in the range of 100 to 200 mg/dL. As the patient improves, insulin resistance abates and dextrose requirements will increase. Supplemental dextrose will be needed for at least several hours after the insulin infusion is discontinued. Administer supplemental potassium initially if patient is hypokalemic (serum potassium less than 2.5 mEq/L). Monitor serum potassium every 4 hours and supplement as needed to maintain potassium of 2.5 to 2.8 mEq/L.
    3) LIPID EMULSION
    a) Lipid emulsion has been successful in animal studies and case reports of patients with refractory hypotension and dysrhythmias after overdose from other lipid soluble xenobiotics. It should be considered in patients with severe cardiac toxicity from venlafaxine overdose. 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.
    H) CONDUCTION DISTURBANCE OF THE HEART
    1) QRS widening (progressive widening on serial ECGs or a single ECG with a duration longer than 120 msec) should be treated with bicarbonate boluses of 50 mEq until the QRS narrows. The serum pH should be maintained between 7.45 and 7.55 (CHILD: 1 to 2 mEq/kg of sodium bicarbonate).
    I) VENTRICULAR DYSRHYTHMIAS
    1) Unstable ventricular dysrhythmias should be treated according to ACLS protocols. Lidocaine has been suggested as the antiarrhythmic of choice for ventricular tachycardia or ventricular fibrillation.
    J) NAUSEA AND VOMITING
    1) Severe vomiting can be treated with antiemetics. Metoclopramide: ADULT: 10 to 20 mg every hour as needed; CHILD: 0.1 to 0.2 mg/kg repeated every 6 hours as needed.
    K) HYPERTHERMIA
    1) Benzodiazepines should be used liberally to control agitation. Enhance heat loss using evaporation (keep skin damp and use fans to encourage air circulation), ice water immersion or packing the patient in ice for severe hyperthermia. In severe cases, the patient should be intubated and higher doses of sedatives, such as propofol, should be used. Occasionally, paralysis is necessary to eliminate the neuromuscular tone that is the etiology of rhabdomyolysis and fever.
    L) SEROTONIN SYNDROME
    1) Benzodiazepines should be administered to control neuromuscular hyperactivity. Cyproheptadine has been used as a serotonin receptor antagonist minimizing the serotonergic effects in overdose. Adult dosing of cyproheptadine is 8 mg every 6 hours. It is only available in an oral formulation; it can be crushed and administered via nasogastric tube if the patient is unable to take orally. CHILD: 0.25 mg/kg, maximum 8 mg/dose.
    M) ENHANCED ELIMINATION
    1) Venlafaxine and desvenlafaxine have large volumes of distribution, so hemodialysis is unlikely to be effective in overdose.
    N) PATIENT DISPOSITION
    1) HOME CRITERIA: An inadvertent ingestion of a single agent of less than 5.5 mg/kg venlafaxine in an asymptomatic child may be managed at home.
    2) OBSERVATION CRITERIA: Patients who are symptomatic, those who have taken deliberate ingestions, and children with inadvertent ingestions of more than 5.5 mg/kg venlafaxine should be sent to a healthcare facility for observation. IMMEDIATE RELEASE: Observe patients for at least 6 hours (Tmax of venlafaxine is 2 hours for venlafaxine and 3 hours for the active metabolite, O-desmthylvenlafaxine (ODV)) as symptoms will likely develop within this time period. EXTENDED RELEASE: Patients that have ingested an extended release product (Tmax of venlafaxine (150 mg) extended release capsule is 5.5 hours for venlafaxine and 9 hours for the active metabolite, ODV) have the potential to manifest delayed symptoms and should be observed for 11 to 18 hours. Patients should be observed until vital signs are normal and symptoms have resolved.
    3) ADMISSION CRITERIA: Overdose patients with delirium, seizures, rhabdomyolysis, or ventricular dysrhythmias should be admitted until toxicity resolves. All patients with cardiac dysrhythmias or serotonin syndrome should be admitted to an ICU.
    4) CONSULT CRITERIA: A medical toxicologist or poison control center should be consulted for any patient with moderate or severe toxicity.
    O) PITFALLS
    1) Failure to adequately sedate severely symptomatic patients may lead to progressive rhabdomyolysis, seizures, acidosis, hyperthermia, and multiorgan failure. Failure to adequately fluid resuscitate can lead to renal insufficiency and potentiate tachycardia.
    P) PHARMACOKINETICS
    1) VENLAFAXINE: IMMEDIATE RELEASE: Rapidly absorbed; 92% of a single dose is absorbed. It has a parent half-life of 3 to 4 hours though it has active metabolites with a half life of 10 hours. It is primarily metabolized via CYP 2D6 and 85% renally cleared. Hepatic impairment may prolong elimination and poor 2D6 metabolizers may reach toxicity at lower doses. It is about 27% protein bound, with a volume of distribution of 6 to 7 L/kg. Tmax for venlafaxine immediate release formulation is 2 hours for venlafaxine and 3 hours for the active metabolite, O-desmethylvenlafaxine (ODV). EXTENDED RELEASE: Minimally bound to plasma protein (27%). Slightly slower rate of absorption but same extent of absorption compared with an immediate release formulation. Tmax for venlafaxine extended release capsules (150 mg every 24 hours) is 5.5 hours for venlafaxine and 9 hours for the active metabolite, ODV. Following the administration of an extended release capsule it usually results in lower Cmax (150 ng/mL for venlafaxine and 260 ng/mL for ODV).
    2) DESVENLAFAXINE: It is 30% protein bound with a volume of distribution of 3.4 L/kg and a half-life of 11 hours. Absolute bioavailability is approximately 80%. Mean time to peak plasma concentration is approximately 7.5 hours after oral administration.
    Q) TOXICOKINETICS
    1) Seizures generally occur 2 to 4 hours after overdose. Elimination half-life is prolonged in overdose ranging from 10 to 15 hours.
    R) DIFFERENTIAL DIAGNOSIS
    1) CNS depression and tachycardia should prompt consideration of anticholinergic syndrome. Anticholinergic syndrome should lack the hyperreflexia that is prominent in serotonin syndrome and may include dry mucous membranes, mydriasis, and decreased bowel sounds. Sympathomimetic agents should be considered, many of the clinical features are similar, though hyperreflexia and clonus is more prominent in serotonin syndrome.

Range Of Toxicity

    A) VENLAFAXINE
    1) ADULTS: Doses larger than 1500 mg have been associated with seizures. Life-threatening cardiovascular toxicity manifested by ventricular dysrhythmias are associated with doses larger than 8000 mg.
    2) PEDIATRIC: Doses less than 5.5 mg/kg in children are unlikely to result in toxicity.
    B) DESVENLAFAXINE
    1) ADULT: Limited data. In a pre-marketing study, 4 adults ingested desvenlafaxine succinate (4000 mg [desvenlafaxine alone], 900, 1800 and 5200 mg [in combination with other drugs] and each patient recovered. Adverse events reported and likely associated with a 5-day overdose of greater than 600 mg daily, included: headache, nausea/vomiting, diarrhea, agitation, dizziness, paresthesia, and tachycardia.
    2) PEDIATRIC: An 11-month-old ingested 600 mg of desvenlafaxine succinate and recovered completely with treatment.
    C) THERAPEUTIC DOSE: VENLAFAXINE: ADULT: 37.5 to 75 mg/day orally; MAX dose of 375 mg/day of immediate-release and 225 mg of extended-release. DESVENLAFAXINE: ADULT: 50 mg daily; PEDIATRIC: Not approved for use.

Summary Of Exposure

    A) USES: Venlafaxine and desvenlafaxine are primarily used to treat anxiety and depressive disorders. Off-label uses for venlafaxine include treatment of ADHD, eating disorders, chronic pain, headache, and agoraphobia. Extended release formulations of both drugs are available.
    B) PHARMACOLOGY: Venlafaxine and desvenlafaxine inhibit neuronal reuptake of both serotonin and norepinephrine in the CNS.
    C) TOXICOLOGY: Increases in serotonin, norepinephrine, and dopamine can lead to cardiovascular toxicity (due to sodium and potassium channel blockade), and seizures. Venlafaxine and desvenlafaxine are highly serotonergic and have been implicated in causing serotonin syndrome in therapeutic doses or (rarely) in overdose, as single agents or in combination with other serotonergic agents.
    D) EPIDEMIOLOGY: Poisoning is common and several deaths occur each year.
    E) WITH THERAPEUTIC USE
    1) ADVERSE EVENTS: Mild effects include nausea/vomiting, dry mouth, constipation, loss of appetite, asthenia, dizziness, headache, insomnia, somnolence, blurred vision, nervousness and tremor following therapeutic venlafaxine therapy. The most commonly reported adverse events reported with desvenlafaxine were similar to venlafaxine. The following adverse effects have also rarely occurred with therapeutic doses of these agents: hyponatremia, hypochloremia, hepatitis, seizures, serotonin syndrome (generally if used with another serotonergic agent), thrombocytopenia, exacerbation of congestive heart failure, and elevated blood pressure.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Mild serotonergic effects include tremor, hyperreflexia, anxiety, and agitation. Palpitations, tachycardia and exacerbation of congestive heart failure have been reported with both therapeutic use and overdose. Somnolence is common in mild toxicity though delirium is more likely as toxicity increases.
    2) SEVERE TOXICITY: Serotonin syndrome (altered mental status, neuromuscular abnormalities/rigidity, autonomic instability) is a severe manifestation of overdose. Cardiovascular toxicity includes, PR, QRS, and QT prolongation occasionally culminating in ventricular tachycardia, fibrillation, and cardiac arrest. Interval prolongation is common though ventricular dysrhythmias are rare and are associated with large overdose (greater than 8 g). Seizures are common in patients taking over 1 g. Seizures have been reported in neonates exposed in utero from mothers taking therapeutic doses. Rhabdomyolysis is common in severe toxicity. Development of eosinophilic pneumonia was reported in a young adult following overdose. Coma is a rare effect of severe toxicity.

Heent

    3.4.3) EYES
    A) WITH THERAPEUTIC USE
    1) Blurred vision has occurred in approximately 6% of patients receiving therapeutic doses of venlafaxine in clinical trials (Anon, 1993).
    2) GLAUCOMA: Bilateral acute angle closure glaucoma was reported in a 45-year-old woman after the third day of venlafaxine therapy (75 mg/day) (Ng et al, 2002). A 35-year-old man experienced bilateral acute angle closure glaucoma 10 days after starting venlafaxine 75 mg daily. Intraocular pressures were 69 mmHg (right) and 62 mmHg (left). In addition to medical therapy, the patient required bilateral laser iridotomies (deGuzman et al, 2005).

Cardiovascular

    3.5.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    1) Venlafaxine has caused hypotension, hypertension, palpitations, tachycardia, acute heart failure, cardiac arrest, and prolongation of the QRS and QT intervals. Doses of 8 g or greater are more likely to produce cardiac toxicity. Tachycardia and mild hypotension are relatively common adverse events. Fatalities have been reported after acute exposure.
    3.5.2) CLINICAL EFFECTS
    A) CARDIOVASCULAR FINDING
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a prospective review of consecutive patients presenting with a venlafaxine alone overdose between January 1997 and December 2007, 369 occasions of venlafaxine overdoses (median dose: 1500 mg; Interquartile Range (IQR): 600 to 3000 mg; range: 75 to 13500 mg) in 273 patients were observed with no reports of dysrhythmias and only minor ECG changes. Tachycardia was observed in 54% of patients and mild hypertension was reported in 40% of patients; severe hypertension (systolic BP greater than 180 mm Hg) developed in 3% of patients, and mild hypotension systolic BP less than 90 mm Hg) developed in 5%. Conduction disturbances were found in 7 patients with 5 patients having a preexisting history of conduction disturbances. Alterations in QRS and QT interval were considered mild (the median maximum QRS width was 85 ms (IQR: 80 to 90 ms; range: 70 to 145 ms); 24 patients (7%) had a QRS of greater than or equal to 120 ms. QTc was prolonged in 22 patients (6%) and patients ingesting larger doses were more likely to develop QTc prolongation. The findings suggest that a mild exposure does not result in cardiotoxicity; however, doses greater than 8 g may produce significant cardiotoxicity (Isbister, 2009).
    B) TACHYARRHYTHMIA
    1) WITH POISONING/EXPOSURE
    a) A low incidence of cardiovascular effects has been reported for venlafaxine as compared to tricyclic antidepressants. Out of 14 reported cases of venlafaxine acute overdoses two patients were reported to have developed a mild sinus tachycardia and a third patient was reported to have a prolongation of QTc to 500 msec, compared with 405 msec at baseline(Prod Info Effexor(R) oral tablets, 2010).
    b) CASE SERIES: In a prospective review of consecutive patients presenting with a venlafaxine alone overdose between January 1997 and December 2007, 369 occasions of venlafaxine overdoses (median dose: 1500 mg; Interquartile Range (IQR): 600 to 3000 mg; range: 75 to 13500 mg) in 273 patients, tachycardia was observed in 54% of patients (Isbister, 2009).
    c) CASE SERIES: In a retrospective review of 235 consecutive cases of venlafaxine (median dose 1500 mg; range: 919 to 2800 mg) overdose, tachycardia (heart rate greater than 100 beats per minute) occurred in 94 patients (40%) and was found to be dose-dependent (Howell et al, 2007).
    d) CASE REPORT: A 41-year-old woman who ingested 4.5 g venlafaxine, 500 mg diphenhydramine and 50 mg thiothixene presented to the ED with sinus tachycardia and diffuse nonspecific T wave changes on ECG. Treatment was symptomatic with resultant full recovery (Fantaskey & Burkhart, 1995).
    e) Tachycardia has also been reported in other patients with venlafaxine overdose (Paparrigopoulos et al, 2011; Pascale et al, 2005; Shaw & Sheard, 2005; Hanekamp et al, 2005; Mazur et al, 2003; Woo et al, 1995; Dahl et al, 1996; Peano et al, 1996; Kokan & Dart, 1996; Adesanya & Varma, 1997; Leaf, 1998; Coorey & Wenck, 1998; Rosen et al, 1997; Blythe & Hackett, 1999; Thorsson et al, 2000).
    C) VENTRICULAR TACHYCARDIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 38-year-old man developed ventricular tachycardia after ingesting venlafaxine and lamotrigine (Peano et al, 1996).
    b) CASE REPORT/FATALITY: A 45-year-old woman ingested 8.4 g of venlafaxine and was found pulseless. She was in ventricular fibrillation on paramedics arrival; spontaneous rhythm was restored with intubation/oxygenation, epinephrine, atropine, and direct current shock but she remained severely hypotensive and was determined to be brain dead (Banham, 1998).
    c) CASE REPORT: Ventricular tachycardia, ventricular fibrillation, and supraventricular tachycardia have been reported in a patient who took 7.5 g of venlafaxine and an unknown amount of oxazepam (Thorsson et al, 2000).
    d) CASE REPORT: A 24-year-old woman developed sinus tachycardia (130 to 155 beats per minute) and seizures followed by intraventricular conduction abnormalities and an episode of torsades de pointes after an intentional ingestion of 8.85 g venlafaxine. ECG abnormalities resolved over 24 hours following administration of intravenous magnesium. The patient was discharged home without further complications (Oliver et al, 2002).
    D) PROLONGED QT INTERVAL
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a prospective review of consecutive patients presenting with a venlafaxine alone overdose between January 1997 and December 2007, 369 occasions of venlafaxine overdoses (median dose: 1500 mg; Interquartile Range (IQR): 600 to 3000 mg; range: 75 to 13500 mg) in 273 patients were observed with no reports of dysrhythmias and only minor ECG changes. QTc was prolonged in 22 patients (6%) and patients ingesting larger doses were more likely to develop QTc prolongation(Isbister, 2009).
    b) CASE SERIES: In a retrospective review of 235 consecutive cases of venlafaxine (median dose 1500 mg; range: 919 to 2800 mg) overdose, QTc prolongation (greater than 450 ms) occurred in 24 patients (10.7%) and was found to be dose-dependent. No related dysrhythmias or fatalities were reported (Howell et al, 2007).
    c) CASE REPORT: A 41-year-old man developed hypotension, QRS widening and QTc prolongation after ingesting 5.6 g of venlafaxine (Kokan & Dart, 1996).
    d) Out of 14 reported cases of venlafaxine acute overdoses, 2 patients were reported to have developed a mild sinus tachycardia and a third patient was reported to have a prolongation of QTc to 500 msec, compared with 405 msec at baseline (Prod Info Effexor(R) oral tablets, 2010).
    e) CASE REPORT: A 29-year-old man ingested 2.8 g of venlafaxine and developed prolonged QRS complexes (limb leads, 0.12 sec) and rSR' pattern in the anterior chest leads on ECG (Coorey & Wenck, 1998).
    f) CASE REPORT: A 45-year-old woman ingested 10.9 g of venlafaxine and 35 mg of clonazepam, presented to the Emergency Department with a normal ECG, but subsequently developed, over the next hour, flattening of ST segments and intraventricular conduction delay (Rosen et al, 1997).
    g) CASE REPORT: A 33-year-old woman ingested 8.4 g of venlafaxine and developed right axis deviation, a prolonged QT interval (469 msec) and a prolonged QRS complex (129 msec). It is suggested that cardiotoxicity is more likely in patients with the poor CYP2D6 metabolizer phenotype (Blythe & Hackett, 1999).
    h) CASE REPORT: A 44-year-old woman overdosed with venlafaxine (3 g), clonazepam (20 mg), lormetazepam (24 mg) and thioridazine (10 mg). She presented with incomplete right bundle branch block, and subsequently developed hypotension followed by atrial fibrillation with wide QRS complexes. Sinus rhythm with normal QRS complexes resumed within 15 minutes of infusion of 100 mL 1M sodium bicarbonate (Combes et al, 2001).
    i) CASE REPORT: After ingesting 4.5 g of venlafaxine, a 25-year-old woman developed a generalized seizure, sinus tachycardia (150 bpm), incomplete right bundle branch block, QRS duration of 100 msec, prolonged QTc duration (0.46 seconds), and rhabdomyolysis (CK 52,600 Units/L). ECG abnormalities resolved by the day after admission (Pascale et al, 2005).
    E) VENTRICULAR FIBRILLATION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT/FATALITY: A 40-year-old man intentionally ingested 19 g of extended release venlafaxine (90 150-mg venlafaxine and 75 75-mg venlafaxine extended-release tablets) and was admitted within 45 minutes of exposure, and was alert with mild tachycardia. An initial ECG showed tachycardia with no evidence of ischemia. Decontamination included activated charcoal and whole bowel irrigation. Toxicology screening was negative. Over the course of several hours the patient developed 2 seizures and was admitted to the intensive care. Approximately 9 hours after exposure, the patient was increasingly lethargic and developed refractory ventricular fibrillation (VF). The authors suggest that sodium channel toxicity was responsible for the fatal dysrhythmia. Although the patient developed no evidence of ventricular ectopy, the ECG showed a new onset of QRS widening (158 msec) and QTc prolongation (564 msec) shortly before the patient's death (Bosse et al, 2008).
    F) CONDUCTION DISORDER OF THE HEART
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a prospective review of consecutive patients presenting with a venlafaxine alone overdose between January 1997 and December 2007, 369 occasions of venlafaxine overdoses (median dose: 1500 mg; Interquartile Range (IQR): 600 to 3000 mg; range: 75 to 13500 mg) in 273 patients were observed with no reports of dysrhythmias and only minor ECG changes. Alterations in QRS interval was considered mild (the median maximum QRS width was 85 ms (IQR: 80 to 90 ms; range: 70 to 145 ms; 24 patients (7%) had a QRS of greater than or equal to 120 ms (Isbister, 2009).
    b) CASE REPORT/FATALITY: A 30-year-old woman developed tachycardia followed by asystole after a mixed ingestion including venlafaxine and carbamazepine (Dahl et al, 1996). Postmortem blood venlafaxine level was 89,000 ng/mL (225 times therapeutic).
    c) CASE REPORT: A 44-year-old woman overdosed with venlafaxine (3 g), clonazepam (20 mg), lormetazepam (24 mg) and thioridazine (10 mg). She presented with incomplete right bundle branch block, and subsequently developed hypotension followed by atrial fibrillation with wide QRS complexes. Sinus rhythm with normal QRS complexes resumed within 15 minutes of infusion of 100 mL 1M sodium bicarbonate (Combes et al, 2001).
    d) CASE REPORT: A 35 year-old woman taking 225 mg daily of venlafaxine and 300 mg daily of bupropion received an ICD for severe, nonischemic cardiomyopathy. Initial testing of the ICD failed to convert induced ventricular fibrillation at 15 Joules and 20 Joules. Venlafaxine and bupropion were discontinued and the ICD performed normally on testing 4 days later. Venlafaxine was thought to be responsible for the elevated defibrillation threshold due to its inhibition of cardiac sodium channels (Carnes et al, 2004).
    G) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Hypotension has been reported following venlafaxine overdose (Durback & Scharman, 1996; Kokan & Dart, 1996; Rosen et al, 1997).
    b) CASE REPORT/FATALITY: A 39-year-old woman presented to the ED after ingesting 30 g of extended-release venlafaxine capsules approximately 12 to 24 hours prior to arrival. Initial blood pressure was 85/45 mmHg, but gradually worsened requiring intensive care and ventilatory support. A dopamine infusion was titrated up to 20 mcg/kg/min for worsening hypotension; norepinephrine and phenylephrine infusions were subsequently added. A lidocaine infusion was started following two episodes of ventricular tachycardia. Dobutamine was also started, but discontinued due to tachycardia. Weaning from all vasopressors and inotropic support occurred by hospital day 4; however, the patient remained unresponsive, developed complications of bowel perforation and died on hospital day 43 (Mazur et al, 2003).
    c) CASE REPORT: A 41-year-old man developed hypotension after ingesting 5.6 grams of venlafaxine (Kokan & Dart, 1996).
    d) CASE REPORT: A 45-year-old woman developed somnolence and hypotension after an overdose ingestion of venlafaxine (10.9 grams) and clonazepam (35 mg). The patient's blood pressure increased following supportive care (Rosen et al, 1997).
    e) CASE REPORT: A 44-year-old woman overdosed with venlafaxine (3 g), clonazepam (20 mg), lormetazepam (24 mg) and thioridazine (10 mg). She presented with incomplete right bundle branch block, and subsequently developed hypotension followed by atrial fibrillation with wide QRS complexes. Sinus rhythm with normal QRS complexes resumed within 15 minutes of infusion of 100 mL 1M sodium bicarbonate (Combes et al, 2001).
    f) CASE REPORT/FATALITY: A 45-year-old woman ingested 8.4 g of venlafaxine and was found pulseless. She was in ventricular fibrillation on paramedics arrival; spontaneous rhythm was restored with intubation/oxygenation, epinephrine, atropine, and direct current shock but she remained severely hypotensive and was determined to be brain dead (Banham, 1998).
    H) HYPERTENSIVE EPISODE
    1) WITH THERAPEUTIC USE
    a) Sustained increases in blood pressure have been reported in patients receiving therapeutic doses of venlafaxine. Premarketing studies have shown an incidence of sustained increased supine diastolic blood pressure of 3% for venlafaxine doses less than 100 mg/day, 5% for doses between 101 and 200 mg/day, 7% for doses between 201 and 300 mg/day, and 13% for doses greater than 300 mg/day. Most of the blood pressure increases were between 10 and 15 mmHg (Prod Info Effexor(R) oral tablets, 2010).
    2) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a retrospective review of 235 consecutive cases of venlafaxine (median dose 1500 mg; range: 919 to 2800 mg) overdose, hypertension (systolic greater than 140 mmHg (28.4%; n=64)) occurred in 64 patients (28.4%) (Howell et al, 2007).
    b) CASE REPORT: Hypertension (BP 168/101 mmHg) was evident in a 41-year-old woman following ingestion of 4.5 g venlafaxine, 500 mg diphenhydramine and 50 mg thiothixene (Fantaskey & Burkhart, 1995).
    I) PALPITATIONS
    1) WITH THERAPEUTIC USE
    a) Palpitations were reported in 3 of 66 patients receiving venlafaxine 75 to 375 mg/day in one study; however, a causal relationship was not established (Khan et al, 1991).
    J) ACUTE HEART FAILURE
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 21-year-old woman experienced progressive dyspnea, cough, vomiting, and weight loss over two months. She had been taking venlafaxine since two months prior to symptom onset. Echocardiography demonstrated an ejection fraction of 38% with normal coronary angiography. CT of the chest showed multiple, small interstitial nodules. Lung biopsy noted a lymphocytic interstitial infiltrate with occasional granulomas. Clinical condition improved over two weeks following corticosteroid therapy and discontinuation of venlafaxine. She remained asymptomatic at 3 years follow-up (Drent et al, 2003).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 30-year-old woman took an overdose of 5.5 g venlafaxine, as well as 250 mg hydroxyzine and 500 mg alprazolam. She presented with symptoms of seizures and severe serotonin syndrome. Within 10 hours she developed cardiogenic pulmonary edema. Echocardiography demonstrated global left ventricular hypokinesia (sparing the apex) and an ejection fraction of 35%. She improved with supportive care. Follow-up echocardiography obtained 10 days later was normal (Fangio et al, 2007).
    b) CASE REPORTS: Four healthy women with no history of cardiovascular disease were admitted with left ventricular (LV) failure following overdose of venlafaxine. Three patients died and one completely recovered. The amount of venlafaxine ranged from 3150 to 13500 mg (extended-release preparations in 2 cases). LV ejection fraction on echocardiography was between 15% and 18% in 3 patients. A 38-year-old woman ingested 4200 mg and developed serotonin syndrome and LV failure and was improving, but died on day 11 of refractory hypoxemia. The second patient, a 35-year-old woman ingested 90 (150 mg extended release) tablets and developed generalized seizures and LV failure (15% ejection fraction). At 17 hours, despite aggressive care the patient died of a sudden cardiac arrest. The final patient, a 65-year-old woman ingested 7000 mg of immediate-release venlafaxine and was comatose within 7 hours and died 7 hours later. The patient developed low cardiac output (1 L/min) and died of refractory cardiogenic shock; an echocardiogram was not performed (Batista et al, 2013).
    K) CARDIAC ARREST
    1) WITH POISONING/EXPOSURE
    a) CASE REPORTS/FATALITY: Two adult women were admitted with CNS depression, tachycardia and hypotension following large ingestions of venlafaxine (approximately 14.7 g of extended-release capsules in one patient). Shortly after admission, episodes of ventricular tachycardia and ventricular fibrillation (in one patient) occurred and were initially responsive to therapy. However, both patients developed cardiac arrest which failed to respond to resuscitation efforts. One patient died within 3.5 hours of admission, and the second patient died within the first 24 hours of admission. Both had high venlafaxine concentrations at autopsy performed 48 hours after death (Hojer et al, 2008).
    b) CASE REPORT: Prolonged cardiac arrest has been reported in a patient who took 7.5 g of venlafaxine and an unknown amount of oxazepam (Thorsson et al, 2000).
    c) CASE REPORT/FATALITY: After ingesting an unknown amount of venlafaxine, a 34-year-old man developed signs and symptoms of serotonin syndrome, and developed rhabdomyolysis, renal and cardiac failure and died a day later (Shaw & Sheard, 2005).
    L) ACUTE MYOCARDIAL INFARCTION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 51-year-old woman with no prior history of heart disease developed a non-ST elevation myocardial infarction and severe CNS depression following a mixed ingestion of an unknown amount of venlafaxine, metoprolol and acetaminophen. One day after ingestion, serum concentrations of venlafaxine and O-desmethylvenlafaxine were 21.82 mg/L (therapeutic range: 0.1 to 0.5 mg/L) and 3.33 mg/L (0.2 to 0.4 mg/L), respectively. Elevated levels of alcohol and acetaminophen were also reported. The patient was found to have normal coronary arteries following cardiac catheterization and gradually improved with supportive care. It was suggested that venlafaxine may have been associated with an acute ischemic event, although the role of coingestants in this patient cannot be determined (Godkar et al, 2009).
    M) SUBENDOCARDIAL ISCHEMIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 27-year-old man developed serotonin syndrome (diaphoresis, tremor, tachycardia and hypertension) and chest pain after overdose with venlafaxine and paroxetine. ECG showed LVH with strain, initial CK was 233 IU/L with an MB fraction of 34 IU/L (14.6%) with a peak CK of 1748 IU/L. Echocardiogram showed an area of hypokinesia of the anterior septum and reduced LV function (fractional shortening 20%). Chest pain resolved, and blood pressure and ECG normalized after several days but a repeat echocardiogram showed a persistent area of hypokinesia and improved LV function (fractional shortening 29%) (Patridge et al, 2000).

Respiratory

    3.6.1) SUMMARY
    A) WITH THERAPEUTIC USE
    1) Interstitial pneumonitis has rarely been reported with therapeutic use of venlafaxine.
    3.6.2) CLINICAL EFFECTS
    A) PNEUMONITIS
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 55-year-old woman developed worsening fatigue and exertional dyspnea over 6 months. Her medication regimen included: venlafaxine (for 18 months), furosemide, valdecoxcib, atorvastatin, levothyroxine, ezetimibe, and an estradiol patch. CT of the chest noted bilateral ground-glass opacities and prominent hilar lymph nodes. Cardiac catheterization noted an ejection fraction of 60% to 65%. Lung biopsy showed chronic interstitial pneumonia and chronic bronchiolitis consistent with extrinsic allergic alveolitis. Evaluations for fungal infections or hypersensitivity pneumonitis were negative. The patient was treated with methylprednisolone and venlafaxine was discontinued. She recovered 10 days later with improved appearance of her chest radiograph. The patient remained well 4 months later (Turner et al, 2005).
    b) CASE REPORT: A 41-year-old man developed fever, cough, dyspnea and myalgias approximately two weeks after initiation of venlafaxine therapy. CT of the chest noted patchy ground-glass opacities, thickened interlobular septae and bilateral pleural effusions. Transbronchial biopsy showed intravascular and intra-alveolar accumulation of eosinophils, neutrophils and macrophages. Evaluations for infectious or autoimmune etiologies were negative. Venlafaxine was discontinued and the patient was treated with methylprednisolone and clarithromycin. He recovered 10 days later and remained well 2 months later (Fleisch et al, 2000).
    B) SIMPLE PULMONARY EOSINOPHILIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 27-year-old man with schizoaffective disorder developed dyspnea and tachycardia of 125 beats/minute on day 2 of admission after intentionally ingesting 6.3 g of extended-release venlafaxine. Diffuse crackles were noted on exam with bilateral interstitial infiltrates and pulmonary consolidations confirmed on x-ray. Arterial blood gases showed PaCO2 30 mmHg, PaO2 49 mmHg, SaO2 84%, and pH 7.44. He was started on empiric antibiotics with a WBC of 18.5 x 10(3)mcL. On day 3 he developed respiratory failure and shock; bronchoalveolar lavage showed 80% eosinophils, supporting a potential diagnosis of eosinophilic pneumonia. He was intubated, treated with vasopressors and corticosteroids, and discharged uneventfully from the ICU 7 days later. Ten days from admission a chest x-ray was clear. Eosinophilia resolved within 2 months (Paparrigopoulos et al, 2011).

Neurologic

    3.7.1) SUMMARY
    A) WITH THERAPEUTIC USE
    1) Hypomanic symptoms have been reported in patients taking venlafaxine for depression.
    B) WITH POISONING/EXPOSURE
    1) CNS depression, headache, fatigue, dizziness, nervousness, tremors, extrapyramidal effects, and serotonin syndrome have occurred as adverse effects following venlafaxine ingestion.
    2) Somnolence and generalized seizures have been reported following overdoses of venlafaxine.
    3.7.2) CLINICAL EFFECTS
    A) SEIZURE
    1) WITH THERAPEUTIC USE
    a) Seizures were reported to occur in 0.26% of patients receiving therapeutic doses of venlafaxine (Prod Info Effexor(R) oral tablets, 2010).
    2) WITH POISONING/EXPOSURE
    a) INCIDENCE: In a prospective series of 146 cases of venlafaxine overdose, 7 (5%) had seizures (Setz et al, 1995).
    1) A retrospective review of 235 venlafaxine ingestion cases reported seizures in 21 (9%) of patients. Patients experiencing seizures ingested a higher median dose than patients without seizures (2800 mg compared to 1500 mg) (Wilson et al, 2007).
    2) In a prospective clinical study of 51 sequential venlafaxine poisonings and a retrospective study of 632 poisonings, seizures were reported in 14% and 4.8% of poisonings, respectively. Seizures were most frequently seen in those that ingested over 1.5 g (Buckley & Faunce, 2003).
    b) COMBINATION OVERDOSE: A 44-year-old woman ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g, and was found unconscious at home. After admission, a tonic-clonic seizure, rigidity, hyperreflexia, and reflex myocloni were observed. Laboratory results revealed sodium level of 129 mmol/L, creatinine kinase of 1915 Units/L, and creatine kinase-MB of 31 Units/L. Despite supportive therapy, including 8 hours of hemodialysis, her rigidity and hyperreflexia did not improve. Following treatment with an intravenous bolus (150 mL, 2.5 mL/kg) of 20% lipid emulsion, all symptoms resolved completely. On day 6, she was transferred to a psychiatric ward. Before hemodialysis, plasma concentrations of lamotrigine, diazepam, and venlafaxine were 42.4 mg/L (therapeutic range, 1 to 4 mg/L), 560 mcg/L (therapeutic range, 200 to 500 mcg/L), and 1254 mcg/L (therapeutic range, 30 to 150 mcg/L), respectively (Dagtekin et al, 2011).
    c) In preclinical trials there were 14 reports of venlafaxine overdoses. One patient, who ingested 2.75 grams, along with 0.5 milligrams thyroxine, was reported to have two generalized seizures and recovered without sequelae (Anon, 1993a).
    d) Seizures have been reported in several other overdose cases (Paparrigopoulos et al, 2011; Pascale et al, 2005; Kokan & Dart, 1996; Durback & Scharman, 1996; Dahl et al, 1996; Peano et al, 1996; Woo et al, 1995; White et al, 1997; Coorey & Wenck, 1998).
    e) CASE REPORT: A case of venlafaxine overdose (1.3 g total), superimposed on a venlafaxine-phenelzine (MAO inhibitor) drug interaction, resulted in a generalized seizure which did not require treatment and no further sequelae occurred (White et al, 1997).
    f) CASE REPORT: A 34-year-old woman ingested 1400 to 1500 mg of venlafaxine, and approximately 1 hour later, experienced a generalized seizure (Leaf, 1998).
    g) CASE REPORT: A 45-year-old woman ingested 10.9 g of venlafaxine and 35 mg of clonazepam and subsequently developed myoclonic jerking of the face and extremities (Rosen et al, 1997).
    h) CASE REPORT: A 22-year-old man ingested approximately 3 g of venlafaxine and subsequently developed tonic-clonic activity with facial grimacing and teeth clenching (Zhalkovsky et al, 1997).
    i) CASE REPORT: A 33-year-old woman ingested 8.4 g of venlafaxine and experienced a grand mal seizure (Blythe & Hackett, 1999).
    B) MYOCLONUS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 39-year-old woman presented to the ED after ingesting 30 g of extended-release venlafaxine capsules approximately 12 to 24 hours prior to arrival. She gradually became obtunded and required intensive care and ventilatory support. She was noted to have several episodes of myoclonic jerking; however, seizure activity was ruled out following EEG. The patient remained minimally responsive, developed complications of hypotension and bowel perforation and died on hospital day 43 (Mazur et al, 2003).
    C) DROWSY
    1) WITH THERAPEUTIC USE
    a) Somnolence was reported in 24% of patients receiving therapeutic doses of venlafaxine in one clinical trial (Anon, 1993).
    2) WITH POISONING/EXPOSURE
    a) Out of 14 cases of acute venlafaxine overdoses, the most commonly reported symptom was somnolence (Prod Info Effexor(R) oral tablets, 2010).
    b) CASE REPORT: A man presented to the ED drowsy, but arousable following an ingestion of 2.25 g venlafaxine. The patient recovered after receiving supportive treatment (Adesanya & Varma, 1997).
    D) CENTRAL NERVOUS SYSTEM DEFICIT
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Profound CNS depression requiring intubation was reported in a 41-year-old woman following ingestion of 4.5 g venlafaxine, 500 mg diphenhydramine, and 50 mg thiothixene (Fantaskey & Burkhart, 1995).
    b) CASE SERIES: In a prospective series of 146 cases of venlafaxine overdose, 57 (39%) were drowsy, 6 (4%) were responsive to pain only, and 4 (3%) were unresponsive (Setz et al, 1995).
    c) CASE REPORT: A 45-year-old woman became somnolent with nonreactive dilated pupils and roving eye movements following an ingestion of 10.9 g venlafaxine and 35 mg clonazepam (Rosen et al, 1997).
    E) SEROTONIN SYNDROME
    1) WITH THERAPEUTIC USE
    a) Serotonin syndrome, which may include cognitive changes, rigidity, hyperreflexia, tachycardia, diaphoresis, tremulousness, and fever, has been reported after venlafaxine ingestion in conjunction with tranylcypromine or phenelzine (Buckley & Faunce, 2003; Hodgman et al, 1995; Brubacher et al, 1995; Weiner et al, 1998).
    b) CASE REPORT: A 60-year-old woman presented to the ED obtunded, tachycardic, hyperthermic, hyperreflexic, diaphoretic, weak, and confused following unintentional ingestion of a single dose of venlafaxine while on maintenance tranylcypromine therapy. The patient recovered following supportive treatment (Hodgman et al, 1997).
    c) CASE REPORT: A 44-year-old woman ingested phenelzine and venlafaxine concurrently and experienced nausea, anxiety, shortness of breath, inability to swallow, and fever 30 minutes later. The patient presented to the ED 45 minutes after ingestion with lower extremity shaking and increasingly rapid respirations. A diagnosis of serotonin syndrome was subsequently made. The patient recovered following supportive care (Weiner et al, 1998).
    2) WITH POISONING/EXPOSURE
    a) Serotonin syndrome, which may include cognitive changes, rigidity, hyperreflexia, tachycardia, diaphoresis, tremulousness, and fever, has been reported after venlafaxine overdose, alone or in conjunction with other serotonergic agents (Kolecki & Miller, 1996; Daniels, 1998; Patridge et al, 2000).
    b) INCIDENCE: A retrospective review of 96 cases of venlafaxine overdose found features suggestive of serotonin syndrome in 8 (8.3%) patients (Kelly et al, 2004).
    c) CASE REPORT: After ingesting an unknown amount of venlafaxine, a 34-year-old man developed signs and symptoms of serotonin syndrome, and developed rhabdomyolysis, renal and cardiac failure and died a day later (Shaw & Sheard, 2005).
    d) CASE REPORT: Serotonin syndrome and rhabdomyolysis occurred in a 21-year-old woman after ingesting 7.8 g of the extended-release venlafaxine. Her plasma CK levels, 5 hours and 41 hours after ingestion, were 43 Units/L and 42,340 Units/L, respectively. She developed somnolence and disorientation with a Glasgow Coma Scale of 4-6-3, nausea, vomiting, diaphoresis, and sinus tachycardia 160 to 170 bpm, and tonic-clonic seizures. Following supportive care, she recovered (Hanekamp et al, 2005).
    e) CASE REPORT: A 29-year-old woman developed anxiety, tremor, shivering, diarrhea, nausea, vomiting, myoclonus, ataxia, and tachycardia three days after starting venlafaxine at 18.75 to 37.5 mg daily. She was taking imipramine 50 mg daily immediately prior to starting venlafaxine (Pan & Shen, 2003).
    f) CASE REPORT: Features consistent with serotonin syndrome developed in a 44-year-old woman who ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g, and was found unconscious at home. After admission, a tonic-clonic seizure, rigidity, hyperreflexia, and reflex myocloni were observed. Laboratory results revealed sodium level of 129 mmol/L, creatinine kinase of 1915 Units/L, and creatine kinase-MB of 31 Units/L. Despite supportive therapy, including 8 hours of hemodialysis, her rigidity and hyperreflexia did not improve. Following treatment with an intravenous bolus (150 mL, 2.5 mL/kg) of 20% lipid emulsion, all symptoms resolved completely. On day 6, she was transferred to a psychiatric ward. Before hemodialysis, plasma concentrations of lamotrigine, diazepam, and venlafaxine were 42.4 mg/L (therapeutic range, 1 to 4 mg/L), 560 mcg/L (therapeutic range, 200 to 500 mcg/L), and 1254 mcg/L (therapeutic range, 30 to 150 mcg/L), respectively (Dagtekin et al, 2011).
    g) CASE REPORT: A 30-year-old woman was thought to have intentionally ingested a one month supply of 150 mg venlafaxine, 300 mg bupropion, and 3 mg lorazepam that were prescribed 3 days earlier. Soon after admission the patient developed tachycardia, fever (40.6 degrees C), a prolonged QRS complex with a deep slurred S wave on lead I and a R wave on lead aVR. Within an hour she required endotracheal intubation due to respiratory distress and decreased mental alertness; sodium bicarbonate was given for ECG changes. In addition, the patient had an elevated creatine kinase (520 Units/L) and creatinine (1.5 mg/dL) levels. The combination of findings suggested serotonin syndrome, and cyproheptadine and lorazepam were given. Clinically, she improved over the next 2 days. Her creatine kinase and alanine transaminase levels peaked at 107,895 Units/L and 2453 Units/L, respectively, approximately 43 and 102 hours after exposure. She was extubated on day 7 and the patient was found to have persistent choreoathetosis with frontal release signs, impaired recent memory, language and executive function. A MRI performed 7 weeks after exposure showed bilateral pallidal necrosis (Lin et al, 2012).
    F) ANTICHOLINERGIC ADVERSE REACTION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 10-year-old boy was inadvertently given venlafaxine (extended-release formulation) over a 2-day period (total ingested 600 mg; 6.86 mg/kg/day) instead of trimethoprim/sulfamethoxazole and developed anticholinergic-type symptoms. The child was on no other medications. Mydriasis, urinary retention, constipation and a brief episode of auditory and visual hallucinations were reported. By day 4, most symptoms had completely resolved, and the patient was discharged to home (Miller et al, 2008).
    G) DIZZINESS
    1) WITH THERAPEUTIC USE
    a) Dizziness is a relatively common side effect with venlafaxine and usually occurs at higher doses and disappears spontaneously without treatment (Klamerus et al, 1992).
    2) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a prospective series of 146 cases of venlafaxine overdose, 4 (3%) were dizzy (Setz et al, 1995).
    H) HEADACHE
    1) WITH THERAPEUTIC USE
    a) Headache and fatigue are frequently reported side effects and have occurred with higher single doses of venlafaxine; these effects may be due to its serotonergic activity (Saletu et al, 1992). Migraine headache is reported as a frequent adverse effect in premarketing trials of venlafaxine (Prod Info Effexor(R) oral tablets, 2010).
    2) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a prospective series of 146 cases of venlafaxine overdose, 2 complained of headache (Setz et al, 1995).
    I) FEELING NERVOUS
    1) WITH THERAPEUTIC USE
    a) Nervousness was reported in 25% of patients taking therapeutic doses of venlafaxine in one study compared to placebo (Schweizer et al, 1991).
    2) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a prospective series of 146 cases of venlafaxine overdose, 2 were nervous (Setz et al, 1995).
    J) DRUG-INDUCED DYSTONIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A healthy 4-year-old girl inadvertently ingested an unknown amount of venlafaxine (urine mass spectrometry was positive) and developed an acute dystonic reaction (including: mutism, akinesis, and hypomimia). She had significant limb rigidity and was unable to walk due to rigid ankle equinus and marked truncal flexion when held upright. A head CT and serum creatinine kinase level was normal. She was transferred to the ICU and given IV benztropine and showed some improvement 4 hours later. Sixteen hours after admission, hypomimia, bradykinesia and rigidity were still present. Symptoms completely resolved within 2 days with no permanent sequelae (Ware et al, 2012).
    K) EXTRAPYRAMIDAL DISEASE
    1) WITH THERAPEUTIC USE
    a) Extrapyramidal effects have been reported after therapeutic use of venlafaxine (Tzallas & Rynn, 1995).
    L) TREMOR
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a prospective series of 146 cases of venlafaxine overdose, 7 (5%) were tremulous (Setz et al, 1995).
    M) NEUROLEPTIC MALIGNANT SYNDROME
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 44-year-old man developed neuroleptic malignant syndrome 12 hours after adding venlafaxine 75 milligrams daily to trifluoperazine 1 milligram three times daily. The patient presented with profound anxiety, malaise, rigidity, tremor, and severe diaphoresis. On examination, the blood pressure was between 130/80 mm Hg and 165/100 mm Hg. His pulse was 163 bpm, temperature 38.3 degrees C, and respiratory rate 25 breaths per minute (Nimmagadda et al, 2000). This case may actually represent serotonin syndrome (Cassidy & O'Kearne, 2000).

Gastrointestinal

    3.8.1) SUMMARY
    A) WITH THERAPEUTIC USE
    1) Venlafaxine therapeutic use has been associated with nausea, vomiting, dry mouth, and constipation.
    B) WITH POISONING/EXPOSURE
    1) Bowel perforation occurred in one fatal case of intentional venlafaxine overdose. Gastric bezoar has been reported following overdose with a sustained-release product.
    3.8.2) CLINICAL EFFECTS
    A) NAUSEA AND VOMITING
    1) WITH THERAPEUTIC USE
    a) Schweizer et al reported a 55% incidence of nausea in 60 patients receiving therapeutic doses of venlafaxine as compared to 25% incidence in patients receiving placebo. Nausea and vomiting appear to be relatively common adverse reactions observed with venlafaxine; however, these effects usually occur with higher doses and subside with continued treatment (Schweizer et al, 1991; Klamerus et al, 1992; Saletu et al, 1992; Schweizer et al, 1988).
    2) WITH POISONING/EXPOSURE
    a) Nausea and vomiting have been reported following venlafaxine overdose (Hanekamp et al, 2005).
    B) CONSTIPATION
    1) WITH THERAPEUTIC USE
    a) Constipation was reported in a small number of patients receiving therapeutic venlafaxine doses but in no patients receiving placebo in one clinical trial (Khan et al, 1991). Dry mouth was reported in 22% of patients receiving venlafaxine in another clinical trial (Anon, 1993).
    C) PERFORATION OF INTESTINE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 39-year-old woman developed refractory hypotension after ingesting 30 g of extended-release venlafaxine. Abdominal distention with pain was noted 14 days after presentation and abdominal radiographs showed pneumatosis coli. Laparoscopy revealed perforations of the cecum and distal small bowel with numerous tiny, white drug pellets in the colon and peritoneum. Following antibiotic treatment, surgical reexploration found numerous abscesses and computed tomography (CT) scan revealed a new bowel perforation. The patient died on hospital day 43. Bowel perforation was thought to be a consequence of refractory hypotension resulting in bowel ischemia (Mazur et al, 2003).
    D) BEZOAR
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 47-year-old woman ingested an estimated 15 g of sustained-release venlafaxine and developed, seizures, severe respiratory distress, hemodynamic instability, and acute renal failure. A gastroscopy was performed 5 days after exposure and a gelatinous mass of partially dissolved tablets was found coating the entire greater curvature of the stomach. Gastric lavage was performed with no improvement noted; therefore, multiple gastroscopies using basket extraction were required to remove the bezoar. Despite intensive medical care, the patient had complications from cerebrovascular infarcts which resulted in a persistent vegetative state. Care was eventually withdrawn by the family (Djogovic et al, 2007).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) LIVER ENZYMES ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 22-year-old man ingested approximately 3 g of venlafaxine and subsequently developed repeated seizure activity and elevated serum creatine phosphokinase, lactate dehydrogenase, alanine aminotransferase and aspartate aminotransferase levels. Enzyme levels peaked on the second day following the overdose and gradually decreased to normal levels by day 20. The authors postulated that the elevated enzymes most likely represented muscle injury from seizures rather than hepatotoxicity (Zhalkovsky et al, 1997).
    b) CASE REPORT: After ingesting 4.5 g of venlafaxine, a 25-year-old woman developed a generalized seizure, sinus tachycardia, incomplete right bundle branch block, QRS duration of 100 msec, prolonged QTc duration (0.46 seconds), and rhabdomyolysis (CK 52,600 Units/L). Laboratory data revealed elevated liver enzymes, SGOT/AST 588 Units/Liter (reference less than 60 Units/L); SGPT/ALT 130 Units/L (less than 55 Units/L). Following supportive care, she was discharged to the psychiatry department on day 5 (Pascale et al, 2005).
    B) TOXIC HEPATITIS
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 78-year-old man with a past history of Parkinson's disease and a major depression episode developed icteric acute hepatitis after taking venlafaxine for almost 2 months. Liver function returned to normal after venlafaxine therapy was progressively discontinued (Cardona et al, 2000).
    b) CASE REPORT: A 60-year-old woman experienced right upper quadrant abdominal pain, fever, headache and myalgias one month after starting venlafaxine XR 75 mg daily for postmenopausal symptoms. Her other medications included acetaminophen, vitamin D, cetirizine, estradiol vaginal tablets, lansoprazole and assorted herbal supplements. Lab values included ALT 372 U/L, AST 99 U/L, alkaline phosphatase 483 U/L and GGT 962 U/L. Symptoms and lab abnormalities improved with discontinuation of acetaminophen and venlafaxine. Two months later, the patient noted recurrence of symptoms one week after restarting venlafaxine; hepatic enzymes were again abnormal. Symptoms and lab abnormalities again resolved within two weeks of discontinuing venlafaxine (Phillips et al, 2006).
    C) HEPATIC NECROSIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: After ingesting an unknown amount of venlafaxine, a 34-year-old man developed signs and symptom of serotonin syndrome, and developed rhabdomyolysis, renal and cardiac failure and died a day later. The postmortem histology examination of the liver showed confluent coagulative necrosis, occurring mainly as an acinar zone 3 pattern (notably perivenular and centrilobular) with midzone involvement to a lesser extent. Macrovesicular steatosis with a scattered inflammatory infiltrate, indicating acute hepatitis, was also observed (Shaw & Sheard, 2005).

Genitourinary

    3.10.1) SUMMARY
    A) WITH THERAPEUTIC USE
    1) Sexual dysfunction has been reported in a small number of patients receiving therapeutic doses of venlafaxine.
    3.10.2) CLINICAL EFFECTS
    A) ABNORMAL SEXUAL FUNCTION
    1) WITH THERAPEUTIC USE
    a) Sexual dysfunction has been reported in a small number of patients receiving venlafaxine 30 to 375 mg/day in divided doses. Erectile failure, delayed orgasm, anorgasmia, impotence, and abnormal ejaculation have occurred (Anon, 1993; Schweizer et al, 1988).
    B) ACUTE RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: After ingesting an unknown amount of venlafaxine, a 34-year-old man developed signs and symptom of serotonin syndrome, and developed rhabdomyolysis, renal and cardiac failure and died a day later (Shaw & Sheard, 2005).

Hematologic

    3.13.1) SUMMARY
    A) WITH THERAPEUTIC USE
    1) Abnormal platelet function has been reported with therapeutic use.
    3.13.2) CLINICAL EFFECTS
    A) PLATELET DISORDER
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 27-year-old woman noted abnormal bruising and hematomas in response to minimal trauma 6 weeks after starting venlafaxine and lorazepam for anxiety. Platelet count, PT and aPTT were all normal. Venlafaxine was discontinued and symptoms resolved. Symptoms recurred within 2 weeks of restarting venlafaxine therapy. Platelet count, PT and aPTT levels remained normal. Tests of platelet aggregation were performed on the patient’s blood samples. In vitro addition of venlafaxine inhibited the normal platelet responses to collagen, ADP, and epinephrine (Sarma & Horne, 2006).

Dermatologic

    3.14.1) SUMMARY
    A) WITH THERAPEUTIC USE
    1) Sweating has occurred with venlafaxine use.
    2) Rare cases of Stevens-Johnson syndrome and psoriasiform rashes have been reported with venlafaxine use.
    3.14.2) CLINICAL EFFECTS
    A) EXCESSIVE SWEATING
    1) WITH THERAPEUTIC USE
    a) Schweizer et al reported a 25% incidence of increased sweating in patients receiving venlafaxine 75 to 375 mg/day, compared to no increased sweating in patients receiving placebo. In a premarketing dose comparison trial of venlafaxine, 12.4% of patients receiving 225 mg/day incurred sweating as compared to 19.3% of patients receiving 375 mg/day (Schweizer et al, 1991; Prod Info Effexor(R) oral tablets, 2010).
    b) CASE REPORTS: Profuse sweating has been reported in two patients following oral venlafaxine therapy for the treatment of depression. Symptoms resolved following discontinuation of the venlafaxine (Adesanya & Varma, 1997; Garber & Gregory, 1997).
    1) One of the patients was restarted on venlafaxine 75 mg twice daily, with the addition of benztropine 0.5 mg twice daily. The diaphoresis did not recur, and the venlafaxine was increased to 75 mg 3 times daily with no subsequent side effects (Garber & Gregory, 1997).
    B) STEVENS-JOHNSON SYNDROME
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 38-year-old woman experienced fever, truncal rash, and oral and vaginal ulcerations 12 days after starting venlafaxine at 37.5 mg daily. The rash was purpuric with central vesiculation. Skin biopsy findings were consistent with Stevens-Johnson syndrome or erythema multiforme. Venlafaxine was discontinued after 2 days. Rash improved over three days with intravenous steroid treatment. The patient's other medications included topiramate, alprazolam, temazepam, and estradiol/levonorgestrel. These medications were also withheld. However, they were restarted following clinical improvement without recurrence of rash (Weiss et al, 2004).
    C) PSORIASIFORM RASH
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 57-year-old man developed hyperkeratosis of the palms and soles, as well as subungual hyperkeratosis over 6 months. He had been treated with venlafaxine for the prior 10 months. No other etiology could be determined. Lesions did not respond to therapy with oral retinoid or topical urea, salicylate or caryolysin. Symptoms resolved spontaneously over 4 to 5 weeks after discontinuation of venlafaxine (Dalle et al, 2006).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) MYOSITIS
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 30-year-old man experienced pain in the left thigh 2 months after starting venlafaxine therapy for anxiety. Creatinine kinase level was 7700 Units/L while tests for autoimmune disease were normal. MRI of the thigh revealed increased T2-weighted signal intensity within the semimembranosus and semitendinosus muscles. Symptoms improved within three weeks of discontinuing venlafaxine. Biopsy noted intact muscles fibers with a modest mononuclear infiltrate (Jewell et al, 2004).
    B) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: A review of 235 cases of venlafaxine overdose found creatinine kinase levels greater than 1000 Units/L in 9 patients (3.8%). Analysis of the patients who did not have seizures (n=214), found creatinine kinase levels greater than 1000 Units/L in 6 patients (3.6%) (Wilson et al, 2007).
    b) CASE REPORT: A 38-year-old man developed ventricular tachycardia, seizures, and rhabdomyolysis after ingesting venlafaxine and lamotrigine (Peano et al, 1996).
    c) CASE SERIES: Two cases of a delayed rise in plasma creatine kinase (CK) occurred following intentional ingestions of venlafaxine. Case 1 was a 24-year-old woman who ingested 8.85 g with wine and developed dysrhythmias and grand-mal seizures. On hospital day 3, CK peaked at 4346 IU/L with a normal CK-MB fraction; she was discharged home on hospital day 5 without complications. Case 2 was a 21-year-old woman who ingested 1.05 g venlafaxine with 1.6 g citalopram and an unknown amount of co-codamol (acetaminophen and codeine phosphate) and became unconscious with seizures requiring intubation and mechanical ventilation. CK peaked at 10,475 IU/L on hospital day 5; she was discharged home after 6 days (Oliver et al, 2002).
    d) CASE REPORT: After ingesting an unknown amount of venlafaxine, a 34-year-old man developed signs and symptom of serotonin syndrome, and developed rhabdomyolysis (CK 111,000 Units/L), renal and cardiac failure and died a day later (Shaw & Sheard, 2005).
    e) CASE REPORT: Serotonin syndrome and rhabdomyolysis occurred in a 21-year-old woman after ingesting 7.8 g of the extended-release venlafaxine. Her plasma CK levels, 5 hours and 41 hours after ingestion, were 43 Units/L and 42,340 Units/L, respectively. She developed somnolence and disorientation with a Glasgow Coma Scale of 4-6-3, nausea, vomiting, diaphoresis, and sinus tachycardia 160 to 170 bpm, and tonic-clonic seizures. Following supportive care, her CK level decreased to 1276 Units/L (Hanekamp et al, 2005).
    f) CASE REPORT: After ingesting 4.5 g of venlafaxine, a 25-year-old woman developed a generalized seizure, sinus tachycardia (150 bpm), incomplete right bundle branch block, QRS duration of 100 msec, prolonged QTc duration (0.46 seconds), and rhabdomyolysis (CK 52,600 Units/L). This patient had a single brief seizure and was never comatose, suggesting that venlafaxine might cause rhabdomyolysis directly (Pascale et al, 2005).
    g) COMBINATION OVERDOSE: A 44-year-old woman ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g, and was found unconscious at home. After admission, a tonic-clonic seizure, rigidity, hyperreflexia, and reflex myocloni were observed. Laboratory results revealed sodium level of 129 mmol/L, creatinine kinase of 1915 Units/L, and creatine kinase-MB of 31 Units/L. Despite supportive therapy, including 8 hours of hemodialysis, her rigidity and hyperreflexia did not improve. Following treatment with an intravenous bolus (150 mL, 2.5 mL/kg) of 20% lipid emulsion, all symptoms resolved completely. On day 6, she was transferred to a psychiatric ward. Before hemodialysis, plasma concentrations of lamotrigine, diazepam, and venlafaxine were 42.4 mg/L (therapeutic range, 1 to 4 mg/L), 560 mcg/L (therapeutic range: 200 to 500 mcg/L), and 1254 mcg/L (therapeutic range, 30 to 150 mcg/L), respectively (Dagtekin et al, 2011).
    h) COMBINATION OVERDOSE: A 30-year-old woman was thought to have intentionally ingested a one month supply of 150 mg venlafaxine, 300 mg bupropion, and 3 mg lorazepam that were prescribed 3 days earlier. Soon after admission the patient developed tachycardia, fever (40.6 degrees C), a prolonged QRS complex, respiratory distress, serotonin syndrome and rhabdomyolysis. Her creatine kinase and alanine transaminase levels peaked at 107,895 Units/L and 2453 Units/L, respectively, approximately 43 and 102 hours after exposure. Cyproheptadine and lorazepam were given. Clinically, she improved over the next 2 days. She was extubated on day 7 and the patient was found to have persistent choreoathetosis with frontal release signs, impaired recent memory, language and executive function. A MRI performed 7 weeks after exposure showed bilateral pallidal necrosis (Lin et al, 2012).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPOGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 42-year-old woman with a history of depression intentionally ingested 9 g of venlafaxine. She arrived at the ED 4 hours later and was somnolent with tachycardia, mydriasis and tremor. Her initial blood glucose was 2.6 mmol/L (46.8 mg/dL); all other labs were within normal limits. She received a 50 mL bolus of 50% glucose and was decontaminated with gastric lavage and activated charcoal. Immediately following decontamination, she developed a grand mal seizure. A continuous glucose infusion (10% glucose) was started for ongoing intermittent hypoglycemia (0.9 to 3.2 mmol/L) which persisted for 40 hours after exposure. Plasma insulin and C-peptide levels were found to be within normal range during periods of hypoglycemia. Fourteen hours after ingestion, her venlafaxine concentration was 14.7 mg/L (therapeutic, 0.07 to 0.27 mg/L); a toxicology screen was negative for ethanol and other drugs. It is suggested that a venlafaxine overdose can produce prolonged normoinsulinemic hypoglycemia that is resistant to glucose therapy and it is due to increased glucose uptake in peripheral tissues and/or inhibition of hepatic gluconeogenesis (Brvar et al, 2015).
    b) CASE REPORT: A 29-year-old man with a history of depression and posttraumatic stress disorder took 22.5 g venlafaxine and 5 g oxazepam. Initial vital signs were normal, but his bedside glucose was 2.7 mmol/L (reference range: 4.4 to 5.5 mmol/L). The patient required intubation secondary to decreased consciousness. He required supplemental dextrose infusion for 30 hours after ingestion because of recurrent hypoglycemia. The patient gradually improved and regained consciousness on day 3 and was extubated on day 5. Serum insulin and C peptide concentrations were high. Coingestion of an oral hypoglycemic was not completely excluded in this patient. Although the pathogenetic mechanism remained unknown, the authors suggested a serotonergic effect on insulin secretion by venlafaxine and recommended close glucose monitoring following intoxication (Meertens et al, 2007).

Reproductive

    3.20.1) SUMMARY
    A) Venlafaxine and desvenlafaxine are classified as FDA pregnancy category C. Current data does not demonstrate an increased risk of major malformations following venlafaxine use in pregnancy. However, neonates exposed to serotonin and norepinephrine reuptake inhibitors (SNRIs) or SSRIs late in the third trimester have developed serious complications that could be indicative of a toxic effect of the drug, a drug discontinuation syndrome, and in some cases, findings were consistent with serotonin syndrome. An increased risk for social-behavioral abnormalities at 2 to 6 years of age was reported in children exposed to SSRIs or SNRIs in utero who developed neonatal abstinence syndrome (NAS) at birth. Venlafaxine and its active metabolite, desvenlafaxine, are excreted in human breast milk; however, no adverse effects have been noted in nursing infants. Neither venlafaxine nor desvenlafaxine affected fertility in rats at doses that were close to the human dose.
    3.20.2) TERATOGENICITY
    A) NECROTIZING ENTEROCOLITIS
    1) A case report described development of necrotizing enterocolitis in dichorial, diamniotic, twin infants on the sixth day of life following maternal venlafaxine use of 75 mg/day for depression throughout pregnancy until delivery. The mother, who experienced uneventful first and second trimesters, was hospitalized during week 31 due to vaginal bleeding. A Chlamydia trachomatis infection was diagnosed for which she received azithromycin for 4 days. She received betamethasone 12 g twice in 24 hours to augment fetal lung development. The twin infants were delivered via cesarean section at 33+2 weeks. Both were intubated on day 1 of life. Twin A was successfully extubated on day 2. On day 6, signs of necrotizing enterocolitis, abdominal distension, bloody stool, signs of peritonitis, and gastric residuals were observed in the infants. Subsequently, oral feeding was withheld and IV amikacin and amoxicillin were given to both. Following antibiotics, feeding was well-tolerated in twin A. However, twin B continued to deteriorate and underwent surgery on day 10. Bowel necrosis was observed. Therefore, terminal ileum, right colon, and the proximal transverse colon were resected, and enterostomy was performed. He underwent a second surgery for stomal stenosis on day 22 of life. At 5 months of age, the stoma was closed. Histological examination confirmed a complete luminal obliteration of the remaining transverse colon and the proximal section of the descending colon for which an intestinal anastomosis was created. He was discharged 10 days after the stoma closure (Treichel et al, 2009).
    B) LACK OF EFFECT
    1) A meta-analysis of pregnancy risk associated with newer antidepressants, including venlafaxine, found no overall increased risk of major malformations in newborns (Einarson & Einarson, 2005).
    2) One study of 150 women exposed to venlafaxine during pregnancy showed no difference in the risk of major malformations between the venlafaxine group and women taking SSRIs or women taking nonteratogenic drugs (Einarson et al, 2001).
    C) ANIMAL STUDIES
    1) RATS, RABBITS: In studies of rats and rabbits treated with oral venlafaxine 2.5 times and 4 times, respectively, the human dose on a mg/m2 basis, there was no evidence of teratogenicity (Prod Info Effexor XR(R) oral extended release capsules, 2012; Prod Info EFFEXOR(R) oral tablets, 2007).
    2) RATS, RABBITS: Animal studies of pregnant rabbits and rats treated during organogenesis with oral desvenlafaxine succinate up to 15 times and 30 times, respectively, the human dose of 100 mg/day on a mg/m(2) basis showed that there was no evidence of teratogenicity; although fetal weights were reduced and skeletal ossification was delayed in rats at the highest, maternally toxic dose of 300 mg/kg/day. Additionally, pup weights decreased and pup mortality increased during the first 4 days in the offspring of pregnant rats treated with the highest dose of desvenlafaxine succinate throughout gestation and lactation (Prod Info KHEDEZLA oral extended-release tablets , 2013).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) The manufacturer has classified venlafaxine as FDA pregnancy category C (Prod Info Effexor XR(R) oral extended release capsules, 2012; Prod Info EFFEXOR(R) oral tablets, 2007).
    2) The manufacturer has classified desvenlafaxine is classified as FDA pregnancy category C (Prod Info KHEDEZLA oral extended-release tablets , 2013).
    B) SPONTANEOUS ABORTION
    1) A nested case-controlled study showed an increased risk of spontaneous abortion with SSRI use, and notably, venlafaxine or paroxetine use alone, during pregnancy. Data collected from the Quebec Pregnancy Registry between January 1998 and December 2003 on women who filled at least 1 antidepressant prescription during pregnancy and had a clinically detected spontaneous abortion by the twentieth week of gestation (n=284) showed an increased risk of spontaneous abortion (adjusted odds ratio (OR), 1.68; 95% confidence interval (CI), 1.38 to 2.06) when compared with randomly selected registry controls (4 matched controls per case) without antidepressant use. Tracked antidepressant categories included SSRIs, tricyclic antidepressants, serotonin-norepinephrine reuptake inhibitors, combined use of 2 or more antidepressant classes, or others. Venlafaxine use (adjusted OR 2.11; 95% CI, 1.34 to 3.3) or paroxetine use (adjusted OR 1.75; 95% CI, 1.31 to 2.34) alone were independently associated with a higher risk of spontaneous abortion. The highest daily doses of venlafaxine or paroxetine during pregnancy were associated with the greatest spontaneous abortion risk; of the women taking venlafaxine (n=33) or paroxetine (n=84) who spontaneously aborted, an adjusted analysis showed 50% averaged daily doses greater than 150 mg of venlafaxine and 25.5% averaged daily doses of more than 25 mg of paroxetine. Use of sertraline, fluoxetine, citalopram, fluvoxamine, or combined use of 2 or more SSRIs during pregnancy did not correspond with a significant increase in risk of spontaneous abortion (Nakhai-Pour et al, 2010).
    2) A multicenter, prospective, controlled study comparing the results of pregnant women who were being treated with venlafaxine (n=150), an SSRI (n=150), or a nonteratogenic drug (n=150) during the first trimester found that there was not a significant increase in major malformations in those patients taking venlafaxine. Of the 150 patients in the venlafaxine group, all were treated with venlafaxine in the first trimester and 34 of the patients were treated with venlafaxine throughout their pregnancy. Of the patients treated with venlafaxine, 105 patients took 75 mg/day of the immediate-release formulation and 45 patients took 37.5 to 300 mg/day. There were up (0.7%; p=0.93). There was not a significant difference in pregnancy outcomes among the three groups. An increase in spontaneous abortions was found in the venlafaxine group (12%) compared with the SSRI group (10.7%) and the nonteratogenic drugs group (7.3%), but it did not reach statistical significance (Einarson et al, 2001a).
    C) NEONATAL EFFECTS
    1) SUMMARY: Neonates exposed in utero to SSRIs and venlafaxine exhibited a higher rate of behavioral changes than controls. Effects included tremors, agitation, spasms, hypotonia, irritability, sleep disturbances, apnea/bradycardia and tachypnea. Exposed neonates also had a longer median length of hospitalization. Effects were transient, resolving within 3 to 5 days (Ferreira et al, 2007).
    D) SEIZURES
    1) CASE REPORT: Serotonin and norepinephrine reuptake inhibitor (SNRI) withdrawal, complicated with recurrent seizures, was reported in the neonate of a 31-year-old woman administered venlafaxine 150 mg and 75 mg on alternate days for the treatment of anxiety and depression. Initially, the neonate was born at term via vaginal delivery following an uneventful pregnancy and there were no signs of withdrawal or toxicity other than mild respiratory depression. The mother continued on venlafaxine therapy and chose to breastfeed the infant. After the third day of life, short episodes of jerks were observed in the upper extremities followed by myoclonic fits on the fourth day of life. Despite an initial improvement following treatment with phenobarbitone, he continued to have seizures. Treatment with supplemental fosphenytoin was implemented which resulted in normalization of the EEG. However, the infant became hypotonic and his reflexes diminished. Treatment with phenobarbitone and fosphenytoin were discontinued. Breastfeeding was discontinued due to the mother's continued venlafaxine use. Following further supportive care, including treatment with phenobarbitone, his condition gradually improved and he was discharged after 21 days. Treatment with antiepileptic agents was discontinued after 7 weeks, at which time neurologic function was completely restored and EEG recordings were normalized (Haukland et al, 2013).
    2) CASE REPORTS: Two cases of seizures were reported in neonates born to mothers using venlafaxine during pregnancy. Seizures occurred within 24 hours of birth and were self-limited. No other etiology could be found in either case. Both children had subsequently normal growth and development at one year follow-up (Pakalapati et al, 2006).
    3) CASE REPORT: Myoclonic seizures occurred in a 29-week-old preterm infant on her second day of life. She was born to a 26-year-old primigravida via emergency C-section due to maternal preeclampsia; the mother had been taking venlafaxine 150 mg daily and insulin for type 1 diabetes throughout her pregnancy. At 41 hours, the infant developed myoclonic jerks in all 4 extremities along with tremulous and severe agitation. Slight hypocalcemia was the only abnormal laboratory study which was promptly corrected but seizures continued to occur. No other source for seizure activity could be found. Treatment included an IV barbiturate (ie, phenobarbitone) and phenytoin therapy for up to 9 days for persistent seizures. By day 10, seizure activity had resolved. Follow-up at the corrected age of 5 months showed appropriate development with a normal neurologic examine and no seizure activity (Ansary et al, 2014).
    E) LONG TERM NEURODEVELOPMENT
    1) In a prospective, single-blind, cohort study, full-term infants who developed neonatal abstinence syndrome (NAS) at birth had similar cognitive abilities compared with full term infants without NAS at birth when reevaluated at 2 to 6 years of age. However, infants with NAS at birth were at an increased risk for social-behavioral abnormalities at 2 to 6 years of age. The study was designed to assess the long-term neurodevelopment of children exposed in utero to fluoxetine, paroxetine, citalopram, sertraline, fluvoxamine, or venlafaxine. Children with NAS at birth (n=30; Finnegan score of 4 or greater) were compared to children without NAS (n=52; Finnegan score 0 to 3); both groups were similar in mean cognitive ability (106.9 +/- 14 versus 100.5 +/- 14.6, respectively; p=0.12) and developmental scores (98.9 +/- 11.4 versus 95.7 +/- 9.9, respectively; p=0.21). Cognitive ability was based on scores from the Wechsler Preschool and Primary Scale of Intelligence II, the Stanford-Binet Intelligence Scales, or the Bayley Scale of Infant Development II. The NAS infants had an increased risk of social-behavior abnormalities (odds ratio (OR) 3.03, 95% CI, 1.07 to 8.6, p=0.04) based on the Denver Developmental Screening Test II (DDST-II) and NAS after birth was associated with advanced maternal age (OR 1.12, 95% CI, 1 to 1.25, p=0.04). In addition, there was a trend towards small head circumference in the NAS group when compared with the children without NAS (n=6 (20%) versus n=3 (6%), respectively; p=0.068) (Klinger et al, 2011).
    F) QT PROLONGATION
    1) A study of prospectively collected data suggests antenatal use of selective serotonin-reuptake inhibitor (SSRI) antidepressants is associated with QTc interval prolongation in exposed neonates. Between January 2000 and December 2005, researchers compared 52 neonates exposed to SSRI antidepressants (paroxetine (n=25), citalopram (n=13), fluoxetine (n=12), fluvoxamine (n=1), and venlafaxine (n=1)) in the immediate antenatal period to 52 matched neonates with no exposure. Prolonged QTc is defined as an interval of greater than 460 milliseconds (msec) (the widely used upper limit cited by authorities in both pediatric cardiology and neonatology). A pediatric cardiologist blinded to drug exposure, interpreted all electrocardiograms (ECGs) using standard statistical analyses. ECG recordings revealed markedly prolonged mean QTc intervals in exposed neonates compared to unexposed neonates (mean; 409 +/- 42 msec versus 392 +/- 29 msec, p=0.02). The mean uncorrected QT interval was 7.5% longer among exposed neonates (mean; 280 +/- 31 msec versus 261 +/- 25 msec, p less than 0.001). Ten percent (n=5) of exposed neonates had a notable increase in QTc interval prolongation (greater than 460 msec) compared to none of the unexposed neonates. The longest QTc interval observed was 543 msec (Dubnov-Raz et al, 2008).
    G) SEROTONIN SYNDROME
    1) Neonates exposed to serotonin and norepinephrine reuptake inhibitors (SNRIs) or SSRIs late in the third trimester have developed complications necessitating extended hospitalizations, respiratory support, and tube feeding. These complications can occur immediately upon delivery. Respiratory distress, cyanosis, apnea, seizures, temperature instability, feeding difficulty, vomiting, hypoglycemia, hypertonia, hypotonia, hyperreflexia, tremor, jitteriness, irritability, and constant crying have been reported. These clinical findings could be the result of a toxic effect of the drug or a drug discontinuation syndrome. In some cases, clinical findings have been consistent with serotonin syndrome (Prod Info KHEDEZLA oral extended-release tablets , 2013; Prod Info EFFEXOR XR(R) extended-release oral capsules, 2008; Prod Info EFFEXOR(R) oral tablets, 2007).
    H) ANIMAL STUDIES
    1) The no-effect dose for rat pup mortality with desvenlafaxine was 10 times the human dose. However, when pregnant rats were treated with desvenlafaxine succinate throughout gestation and lactation at the highest dose of 300 mg/kg/day, pup weights decreased and pup mortality increased during the first 4 days of lactation (Prod Info KHEDEZLA oral extended-release tablets , 2013).
    2) When pregnant rats were treated with venlafaxine 2.5 times the human dose on a mg/m(2) basis throughout gestation and lactation, pup weights decreased and the incidence of stillborn pups and pup mortality increased during the first five days of lactation; the cause of death is unknown. The no-effect dose for rat pup mortality with venlafaxine was 0.25 times the human dose on a mg/m(2) basis (Prod Info Effexor XR(R) oral extended release capsules, 2012; Prod Info EFFEXOR(R) oral tablets, 2007).
    3) LACK OF EFFECT: When pregnant rats were treated with 30 times the human dose, post-weaning growth and reproductive performance of the progeny were not affected (Prod Info KHEDEZLA oral extended-release tablets , 2013).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) In one study, serum levels were not detected in breastfed infants (6 paroxetine and 6 sertraline) of paroxetine or sertraline-treated mothers. However, venlafaxine serum levels were detected in breastfed infants (n=3) of venlafaxine-treated mothers, amounting to a mean of 10.2% (range 5.3 to 19%) of the maternal drug level for the sum of venlafaxine plus O-desmethylvenlafaxine (ODV; desvenlafaxine). There was no evidence of adverse effects in infants as reported by the mothers (Berle et al, 2004).
    2) Venlafaxine and its metabolite, O-desmethylvenlafaxine (ODV; desvenlafaxine), were detected in six infant blood samples collected over a 12-hour dose interval at steady-state following a maternal venlafaxine dose of 255 mg/day in a study of 6 women taking venlafaxine and their 7 nursing infants. Venlafaxine was detected in the blood of one infant at a low concentration of 5 mcg/L, while ODV was present in four infants in concentrations ranging from 3 to 38 mcg/mL. Mean milk-to-plasma ratios for venlafaxine and ODV were 2.5 (range 2 to 3.2) and 2.74 (range 2.3 to 3.2), respectively. No adverse effects were noted in the infants (Ilett et al, 2002).
    3) Detectable levels of the metabolite O-desmethylvenlafaxine (ODV; desvenlafaxine) were reported in three infants exposed to venlafaxine through breast milk. Both venlafaxine and ODV were concentrated in the milk (milk-to-plasma concentration ratio of 4:1 and 3:1, respectively). Total infant exposure was 7.6% of the weight-adjusted maternal dose. No adverse effects were observed in the nursing infants (Ilett et al, 1998).
    B) ANIMAL STUDIES
    1) Pup weights decreased and pup mortality increased during the first 4 days of lactation in the offspring of pregnant rats treated with of 300 mg/kg/day (30 times the human dose of 100 mg/day on an mg/m(2) basis) of desvenlafaxine succinate throughout gestation and lactation (Prod Info KHEDEZLA oral extended-release tablets , 2013)
    3.20.5) FERTILITY
    A) ANIMAL STUDIES
    1) Venlafaxine did not effect fertility in male or female rats treated with oral doses 2 times the maximum human dose of 225 mg/day on a mg/m2 basis (Prod Info Effexor XR(R) oral extended release capsules, 2012; Prod Info EFFEXOR(R) oral tablets, 2007). However, fertility reductions did occur when male and female rats received O-desmethylvenlafaxine (a metabolite of venlafaxine) before and during mating and gestation at doses about 2 to 3 times the 225 mg/day human dose (Prod Info Effexor XR(R) oral extended release capsules, 2012).
    2) Desvenlafaxine resulted in reduced fertility in both male and female rats treated with oral doses of 300 mg/kg/day (approximately 30 times a human dose of 100 mg/day on a mg/m2 basis). However, there was no effect on fertility in male and female rats at oral doses of 100 mg/kg/day (approximately 10 times the human dose) (Prod Info KHEDEZLA oral extended-release tablets , 2013).

Carcinogenicity

    3.21.2) SUMMARY/HUMAN
    A) At the time of this review, the manufacturer does not report any carcinogenic potential in humans.
    3.21.4) ANIMAL STUDIES
    A) LACK OF EFFECT
    1) No evidence of tumorigenicity was seen in an 18 month study in mice given venlafaxine by oral gavage 120 mg/kg (approximately 16 times the maximum recommended human dose on a mg/kg basis). Additionally, no evidence of tumorigenicity was observed in a 24 month study in rats given 120 mg/kg daily by oral gavage (Prod Info Effexor(R) oral tablets, 2010).

Genotoxicity

    A) Venlafaxine and O-desmethylvenlafaxine (ODV), major metabolite, were not mutagenic in the following assays: Ames reverse mutation assay in Salmonella bacteria; CHO/HGPRT mammalian cell forward gen mutation assay. Additionally, venlafaxine was not found to be mutagenic in the in vitro BALB/c-3T3 mouse cell transformation assay, the sister chromatid exchange in cultured CHO cells, or in the in vitro chromosomal aberration assay in rat bone marrow, while ODV did not show evidence of mutagenicity in the in vitro CHO cell chromosomal aberration assay. ODV demonstrated clastogenic changes in the in vivo chromosomal aberration assay in male rat bone marrow at doses 200 times the maximum human daily dose on a mg/kg basis (50 times the maximum human daily dose on a mg/m(2) basis). The no effect dose was 67 times the human dose on a mg/kg basis (17 times the human dose on a mg/m(2) basis) (Prod Info Effexor(R) oral tablets, 2010).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor vital signs, institute continuous cardiac monitoring and obtain an ECG.
    B) In moderate to severe toxicity serum electrolytes (including glucose), CBC, and creatinine kinase should be obtained.
    C) EEG monitoring may be necessary for patients that require paralysis.
    D) Monitor glucose following a significant exposure. Infrequent reports of prolonged/recurrent hypoglycemia have occurred following overdose.
    E) Venlafaxine may cause false positive results for phencyclidine or tramadol on drug screening assays.
    4.1.2) SERUM/BLOOD
    A) THERAPEUTIC CONCENTRATIONS
    1) Serum concentrations of venlafaxine are not routinely available. In patients experiencing improved depression symptoms during venlafaxine therapy, median serum concentration was 54 mcg/L. Serum concentrations varied between 10.1 mcg/L and 200 mcg/L. The median serum concentration of venlafaxine plus its active metabolite, O-desmethylvenlafaxine, was 193 mcg/L (Charlier et al, 2002).
    B) LABORATORY INTERFERENCE
    1) Venlafaxine in high concentrations may give false positive results for phencyclidine on the Abbot AxSYM fluorescent polarized immunoassay (Bond et al, 2001). False positive results for phencyclidine have also been reported for the Alpha Laboratories INSTANT-VIEW Multi-Drug Screen Urine Test. These findings occurred with therapeutic dosing of venlafaxine (Santos et al, 2007).
    2) Venlafaxine produced a false positive result for tramadol on a liquid chromatography/mass spectrometry assay (Allen, 2006).
    4.1.4) OTHER
    A) OTHER
    1) MONITORING
    a) Monitor blood pressure for possible hypotension or hypertension.
    b) Monitor temperature for possible hyperthermia.
    c) Monitor fluid status and electrolytes as indicated in patients with profuse sweating.
    d) ECG should be monitored for possible tachycardia.

Methods

    A) CHROMATOGRAPHY
    1) Venlafaxine and its major active metabolite, O-desmethylvenlafaxine, have been measured in plasma by high-performance liquid chromatography with ultraviolet detection (Mazur et al, 2003; Klamerus et al, 1992; Matoga et al, 2001).
    2) In one case report, gas chromatography coupled with mass spectrometry (GC-MS) was used to measure venlafaxine serum levels. The limit of detection and the limit of quantification for both venlafaxine and its metabolite were 10 and 25 mcg/L, respectively (Pascale et al, 2005).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Overdose patients with delirium, seizures, rhabdomyolysis, or ventricular dysrhythmias should be admitted until toxicity resolves. All patients with cardiac dysrhythmias or serotonin syndrome should be admitted to an ICU.
    6.3.1.2) HOME CRITERIA/ORAL
    A) An inadvertent ingestion of a single agent of less than 5.5 mg/kg venlafaxine in an asymptomatic child may be managed at home.
    B) In a retrospective series of 14 pediatric cases of venlafaxine ingestion with doses ranging from 18.75 to 75 mg (2.1 to 5.5 mg/kg), only one patient developed minor effects (lethargy) (Herrington & Gorman, 1996). Children ingesting 5.5 mg/kg or less can be managed at home.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) A medical toxicologist or poison control center should be consulted for any patient with moderate or severe toxicity.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Patients who are symptomatic, those who have taken deliberate ingestions, and children with inadvertent ingestions of more than 5.5 mg/kg venlafaxine should be sent to a healthcare facility for observation. IMMEDIATE RELEASE: Observe patients for at least 6 hours (Tmax of venlafaxine is 2 hours for venlafaxine and 3 hours for the active metabolite, O-desmthylvenlafaxine (ODV)) as symptoms will likely develop within this time period. EXTENDED RELEASE: Patients that have ingested an extended release product (Tmax of venlafaxine (150 mg) extended release capsule is 5.5 hours for venlafaxine and 9 hours for the active metabolite ODV) have the potential to manifest delayed symptoms and should be observed for 11 to 18 hours. Patients should be observed until vital signs are normal and symptoms have resolved.

Monitoring

    A) Monitor vital signs, institute continuous cardiac monitoring and obtain an ECG.
    B) In moderate to severe toxicity serum electrolytes (including glucose), CBC, and creatinine kinase should be obtained.
    C) EEG monitoring may be necessary for patients that require paralysis.
    D) Monitor glucose following a significant exposure. Infrequent reports of prolonged/recurrent hypoglycemia have occurred following overdose.
    E) Venlafaxine may cause false positive results for phencyclidine or tramadol on drug screening assays.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) In general, there is no role for prehospital decontamination because of the risk of seizures and subsequent aspiration.
    6.5.2) PREVENTION OF ABSORPTION
    A) SUMMARY: Activated charcoal should be considered in asymptomatic or minimally symptomatic patients who present within a few hours of ingestion, or in symptomatic patients who have a secure airway. Gastric lavage may be considered for large overdoses who present within approximately 1 hour of ingestion, though airway protection should be considered prior to the procedure. Gastroscopy has been used rarely to remove extended release formulations from the stomach. COMBINED THERAPY: The combination of single-dose activated charcoal and whole bowel irrigation has been shown to decrease peak blood concentrations and may be beneficial in decreasing the risk of seizures and cardiovascular toxicity, following airway management, in a large overdose predominated by CNS depression.
    B) ACTIVATED CHARCOAL
    1) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    C) GASTRIC LAVAGE
    1) INDICATIONS: Consider gastric lavage with a large-bore orogastric tube (ADULT: 36 to 40 French or 30 English gauge tube {external diameter 12 to 13.3 mm}; CHILD: 24 to 28 French {diameter 7.8 to 9.3 mm}) after a potentially life threatening ingestion if it can be performed soon after ingestion (generally within 60 minutes).
    a) Consider lavage more than 60 minutes after ingestion of sustained-release formulations and substances known to form bezoars or concretions.
    2) PRECAUTIONS:
    a) SEIZURE CONTROL: Is mandatory prior to gastric lavage.
    b) AIRWAY PROTECTION: Place patients in the head down left lateral decubitus position, with suction available. Patients with depressed mental status should be intubated with a cuffed endotracheal tube prior to lavage.
    3) LAVAGE FLUID:
    a) Use small aliquots of liquid. Lavage with 200 to 300 milliliters warm tap water (preferably 38 degrees Celsius) or saline per wash (in older children or adults) and 10 milliliters/kilogram body weight of normal saline in young children(Vale et al, 2004) and repeat until lavage return is clear.
    b) The volume of lavage return should approximate amount of fluid given to avoid fluid-electrolyte imbalance.
    c) CAUTION: Water should be avoided in young children because of the risk of electrolyte imbalance and water intoxication. Warm fluids avoid the risk of hypothermia in very young children and the elderly.
    4) COMPLICATIONS:
    a) Complications of gastric lavage have included: aspiration pneumonia, hypoxia, hypercapnia, mechanical injury to the throat, esophagus, or stomach, fluid and electrolyte imbalance (Vale, 1997). Combative patients may be at greater risk for complications (Caravati et al, 2001).
    b) Gastric lavage can cause significant morbidity; it should NOT be performed routinely in all poisoned patients (Vale, 1997).
    5) CONTRAINDICATIONS:
    a) Loss of airway protective reflexes or decreased level of consciousness if patient is not intubated, following ingestion of corrosive substances, hydrocarbons (high aspiration potential), patients at risk of hemorrhage or gastrointestinal perforation, or trivial or non-toxic ingestion.
    D) WHOLE BOWEL IRRIGATION (WBI)
    1) Consider the use of whole bowel irrigation after ingestion of extended release products. Gastric bezoar has been reported with these products (Djogovic et al, 2007).
    2) DRUG-INDUCED ILEUS
    a) NEOSTIGMINE: Neostigmine (2.5 mg IV) was found to be effective in treating a drug-induced ileus in 2 adult patients who intentionally ingested extended-release venlafaxine along with other agents (ie, valproate and dothiepin). Each patient was decontaminated with whole-bowel irrigation, but the treatment was found to be ineffective (ie, new onset of abdominal distension, lack of bowel sounds, lack of rectal effluent). Following neostigmine an almost immediate response was observed in one patient, the other patient required a second dose; whole bowel irrigation was continued for up to 48 hours in one patient until pill fragments were no longer visible. Both patients clinically improved with no permanent sequelae (Chan et al, 2005).
    a) WHOLE BOWEL IRRIGATION/INDICATIONS: Whole bowel irrigation with a polyethylene glycol balanced electrolyte solution appears to be a safe means of gastrointestinal decontamination. It is particularly useful when sustained release or enteric coated formulations, substances not adsorbed by activated charcoal, or substances known to form concretions or bezoars are involved in the overdose.
    1) Volunteer studies have shown significant decreases in the bioavailability of ingested drugs after whole bowel irrigation (Tenenbein et al, 1987; Kirshenbaum et al, 1989; Smith et al, 1991). There are no controlled clinical trials evaluating the efficacy of whole bowel irrigation in overdose.
    b) CONTRAINDICATIONS: This procedure should not be used in patients who are currently or are at risk for rapidly becoming obtunded, comatose, or seizing until the airway is secured by endotracheal intubation. Whole bowel irrigation should not be used in patients with bowel obstruction, bowel perforation, megacolon, ileus, uncontrolled vomiting, significant gastrointestinal bleeding, hemodynamic instability or inability to protect the airway (Tenenbein et al, 1987).
    c) ADMINISTRATION: Polyethylene glycol balanced electrolyte solution (e.g. Colyte(R), Golytely(R)) is taken orally or by nasogastric tube. The patient should be seated and/or the head of the bed elevated to at least a 45 degree angle (Tenenbein et al, 1987). Optimum dose not established. ADULT: 2 liters initially followed by 1.5 to 2 liters per hour. CHILDREN 6 to 12 years: 1000 milliliters/hour. CHILDREN 9 months to 6 years: 500 milliliters/hour. Continue until rectal effluent is clear and there is no radiographic evidence of toxin in the gastrointestinal tract.
    d) ADVERSE EFFECTS: Include nausea, vomiting, abdominal cramping, and bloating. Fluid and electrolyte status should be monitored, although severe fluid and electrolyte abnormalities have not been reported, minor electrolyte abnormalities may develop. Prolonged periods of irrigation may produce a mild metabolic acidosis. Patients with compromised airway protection are at risk for aspiration.
    E) COMBINED THERAPY: ACTIVATED CHARCOAL AND WHOLE BOWEL IRRIGATION
    1) In one study, the combination of activated charcoal and whole bowel irrigation has been shown to decrease peak blood concentrations and may be beneficial in decreasing the risk of seizures and cardiovascular toxicity, following airway management, in a large overdose predominated by CNS depression. The combined use of single-dose activated charcoal (SDAC) and whole bowel irrigation (WBI) reduced the absorption of a dose of venlafaxine by 29% and the decrease in the maximum plasma concentration was larger when SDAC and WBI were administered together. However, there was no apparent change in drug clearance. The reduction in peak concentration may be clinically significant in limiting toxicity (eg, seizures) associated with venlafaxine toxicity. The half-life observed for venlafaxine in overdose was 12.9 hours (previously reported range: 10 to 15 hours) in this study (Kumar et al, 2009).
    F) GASTROSCOPY
    1) Consider the use of gastroscopy following an overdose with extended release products. Basket extraction was necessary in one patient to remove a bezoar (Djogovic et al, 2007).
    G) CONCRETIONS
    1) Venlafaxine pellets may coat the lining of the gastrointestinal tract in overdose and may produce prolonged clinical symptoms and delayed drug clearance (Hudson & Djogovic, 2007)
    a) CASE REPORT: A 47-year-old woman ingested 15 g of venlafaxine and developed multiple seizures and mechanical ventilation was required secondary to aspiration that progressed to acute respiratory distress syndrome. The patient remained hypotensive with a depressed level of consciousness. On day 5, a gastroscopy detected venlafaxine pellets coating the entire stomach and gastric lavage was performed for 24 hours to clear the pellets. A repeat gastroscopy showed no significant change and the pellets were removed endoscopically; the retained pellets likely altered normal pharmacokinetics (Hudson & Djogovic, 2007).
    6.5.3) TREATMENT
    A) SUPPORT
    1) Treatment is symptomatic and supportive. Seizures may occur and should be treated aggressively.
    B) MONITORING OF PATIENT
    1) Monitor vital signs, institute continuous cardiac monitoring and obtain an ECG.
    2) In moderate to severe toxicity serum electrolytes (including glucose), CBC, and creatinine kinase should be obtained.
    3) EEG monitoring may be necessary for patients that require paralysis.
    4) Monitor glucose following a significant exposure. Infrequent reports of prolonged/recurrent hypoglycemia have occurred following overdose.
    5) Venlafaxine may cause false positive results for phencyclidine or tramadol on drug screening assays.
    C) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    D) VENTRICULAR ARRHYTHMIA
    1) Treatment of ventricular dysrhythmias may include sodium bicarbonate, lidocaine, or amiodarone.
    a) VENTRICULAR DYSRHYTHMIAS SUMMARY
    1) Obtain an ECG, institute continuous cardiac monitoring and administer oxygen. Evaluate for hypoxia, acidosis, and electrolyte disorders (particularly hypokalemia, hypocalcemia, and hypomagnesemia). Lidocaine and amiodarone are generally first line agents for stable monomorphic ventricular tachycardia, particularly in patients with underlying impaired cardiac function. Amiodarone should be used with caution if a substance that prolongs the QT interval and/or causes torsades de pointes is involved in the overdose. Unstable rhythms require immediate cardioversion.
    2) BICARBONATE
    a) Sodium bicarbonate may be useful to treat QRS prolongation or dysrhythmias after venlafaxine overdose. A reasonable starting dose is 1 to 2 mEq/kg intravenous bolus, repeated as necessary. Monitor arterial blood gases to maintain a pH of 7.45 to 7.55.
    b) CASE REPORT: A 44-year-old woman overdosed with venlafaxine (3 g), clonazepam (20 mg), lormetazepam (24 mg) and thioridazine (10 mg). She presented with incomplete right bundle branch block, and subsequently developed hypotension followed by atrial fibrillation with wide QRS complexes. Sinus rhythm with normal QRS complexes resumed within 15 minutes of infusion of 100 mL 1M sodium bicarbonate (Combes et al, 2001).
    3) LIDOCAINE
    a) LIDOCAINE/INDICATIONS
    1) Ventricular tachycardia or ventricular fibrillation (Prod Info Lidocaine HCl intravenous injection solution, 2006; Neumar et al, 2010; Vanden Hoek et al, 2010).
    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) AMIODARONE
    a) AMIODARONE/INDICATIONS
    1) Effective for the control of hemodynamically stable monomorphic ventricular tachycardia. Also recommended for pulseless ventricular tachycardia or ventricular fibrillation in cardiac arrest unresponsive to CPR, defibrillation and vasopressor therapy (Link et al, 2015; Neumar et al, 2010). It should be used with caution when the ingestion involves agents known to cause QTc prolongation, such as fluoroquinolones, macrolide antibiotics or azoles, and when ECG reveals QT prolongation suspected to be secondary to overdose (Prod Info Cordarone(R) oral tablets, 2015).
    b) AMIODARONE/ADULT DOSE
    1) For ventricular fibrillation or pulseless VT unresponsive to CPR, defibrillation, and a vasopressor therapy give an initial dose of 300 mg IV followed by 1 dose of 150 mg IV. For stable ventricular tachycardias: Infuse 150 milligrams over 10 minutes, and repeat if necessary. Follow by a 1 milligram/minute infusion for 6 hours, then a 0.5 milligram/minute. Maximum total dose over 24 hours is 2.2 grams (Neumar et al, 2010).
    c) AMIODARONE/PEDIATRIC DOSE
    1) Infuse 5 milligrams/kilogram as a bolus for pulseless ventricular tachycardia or ventricular fibrillation; may repeat twice up to 15 mg/kg. Infuse 5 milligrams/kilogram over 20 to 60 minutes for perfusing tachycardias. Maximum single dose is 300 mg. Routine use with other drugs that prolong the QT interval is NOT recommended (Kleinman et al, 2010).
    d) ADVERSE EFFECTS
    1) Hypotension and bradycardia are the most common adverse effects (Neumar et al, 2010).
    E) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) 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) DOPAMINE
    a) DOSE: Begin at 5 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). If hypotension persists, dopamine may need to be discontinued and a more potent vasoconstrictor (eg, norepinephrine) should be considered (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    b) CAUTION: If ventricular dysrhythmias occur, decrease rate of administration (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    3) NOREPINEPHRINE
    a) 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).
    b) DOSE
    1) 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).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    4) If hypotension persists consider insulin and dextrose infusion, or intravenous fat emulsion therapy. Consider extracorporeal support (cardiopulmonary bypass, extracorporeal membrane oxygenation, aortic balloon pump) in patients who remain unstable despite maximal medical therapy.
    F) INSULIN
    1) Infusion of insulin and dextrose has been used to reverse hypotension after overdose of other xenobiotics, and has been used in a patient with hypotension and left ventricular failure after severe venlafaxine overdose.
    2) In adults, a bolus dose 10 units insulin and 25 grams dextrose is followed by an insulin infusion, with a dose ranging from 0.1 unit/kilogram/hour to 1.0 unit/kilogram/hour, and dextrose (50% w/v) infusion, the dose ranging from 5 grams/hour to 15 grams/hour, via a central venous catheter (Yuan et al, 1999; Boyer et al, 2002). Institute continuous cardiac monitoring, and monitor blood glucose and blood pressure frequently.
    3) CASE REPORT: A 45-year-old woman ingested 3150 mg immediate release and 2100 mg extended release venlafaxine. She developed tachycardia, hypotension, left ventricular failure (ejection fraction 18%), oliguria, and increased lactate concentrations. Because of the presence of tachycardia, insulin and dextrose were used as first line agents to treat hypotension. She received an insulin bolus of 1 Unit/kg followed by an infusion of 1 Unit/kg/hr for 18 hours, which was associated with increased urine output and decreased arterial lactate. On the third day after overdose her LV ejection fraction was 65% and she recovered completely (Batista et al, 2013).
    G) 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 REPORT
    a) COMBINATION OVERDOSE: A 44-year-old woman ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g, and was found unconscious at home. After admission, a tonic-clonic seizure, rigidity, hyperreflexia, and reflex myocloni were observed. Laboratory results revealed sodium level of 129 mmol/L, creatinine kinase of 1915 Units/L, and creatine kinase-MB of 31 Units/L. Despite supportive therapy, including 8 hours of hemodialysis, her rigidity and hyperreflexia did not improve. Following treatment with an intravenous bolus (150 mL, 2.5 mL/kg) of 20% lipid emulsion, all symptoms resolved completely. On day 6, she was transferred to a psychiatric ward. Before hemodialysis, plasma concentrations of lamotrigine, diazepam, and venlafaxine were 42.4 mg/L (therapeutic range: 1 to 4 mg/L), 560 mcg/L (therapeutic range: 200 to 500 mcg/L), and 1254 mcg/L (therapeutic range: 30 to 150 mcg/L), respectively (Dagtekin et al, 2011).
    H) 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).
    I) HYPERTENSIVE EPISODE
    1) Monitor vital signs regularly. For mild/moderate hypertension without evidence of end organ damage, pharmacologic intervention is generally not necessary. Sedative agents such as benzodiazepines may be helpful in treating hypertension and tachycardia in agitated patients, especially if a sympathomimetic agent is involved in the poisoning.
    2) For hypertensive emergencies (severe hypertension with evidence of end organ injury (CNS, cardiac, renal), or emergent need to lower mean arterial pressure 20% to 25% within one hour), sodium nitroprusside is preferred. Nitroglycerin and phentolamine are possible alternatives.
    3) SODIUM NITROPRUSSIDE/INDICATIONS
    a) Useful for emergent treatment of severe hypertension secondary to poisonings. Sodium nitroprusside has a rapid onset of action, a short duration of action and a half-life of about 2 minutes (Prod Info NITROPRESS(R) injection for IV infusion, 2007) that can allow accurate titration of blood pressure, as the hypertensive effects of drug overdoses are often short lived.
    4) SODIUM NITROPRUSSIDE/DOSE
    a) ADULT: Begin intravenous infusion at 0.1 microgram/kilogram/minute and titrate to desired effect; up to 10 micrograms/kilogram/minute may be required (American Heart Association, 2005). Frequent hemodynamic monitoring and administration by an infusion pump that ensures a precise flow rate is mandatory (Prod Info NITROPRESS(R) injection for IV infusion, 2007). PEDIATRIC: Initial: 0.5 to 1 microgram/kilogram/minute; titrate to effect up to 8 micrograms/kilogram/minute (Kleinman et al, 2010).
    5) SODIUM NITROPRUSSIDE/SOLUTION PREPARATION
    a) The reconstituted 50 mg solution must be further diluted in 250 to 1000 mL D5W to desired concentration (recommended 50 to 200 mcg/mL) (Prod Info NITROPRESS(R) injection, 2004). Prepare fresh every 24 hours; wrap in aluminum foil. Discard discolored solution (Prod Info NITROPRESS(R) injection for IV infusion, 2007).
    6) SODIUM NITROPRUSSIDE/MAJOR ADVERSE REACTIONS
    a) Severe hypotension; headaches, nausea, vomiting, abdominal cramps; thiocyanate or cyanide toxicity (generally from prolonged, high dose infusion); methemoglobinemia; lactic acidosis; chest pain or dysrhythmias (high doses) (Prod Info NITROPRESS(R) injection for IV infusion, 2007). The addition of 1 gram of sodium thiosulfate to each 100 milligrams of sodium nitroprusside for infusion may help to prevent cyanide toxicity in patients receiving prolonged or high dose infusions (Prod Info NITROPRESS(R) injection for IV infusion, 2007).
    7) SODIUM NITROPRUSSIDE/MONITORING PARAMETERS
    a) Monitor blood pressure every 30 to 60 seconds at onset of infusion; once stabilized, monitor every 5 minutes. Continuous blood pressure monitoring with an intra-arterial catheter is advised (Prod Info NITROPRESS(R) injection for IV infusion, 2007).
    8) NITROGLYCERIN/INDICATIONS
    a) May be used to control hypertension, and is particularly useful in patients with acute coronary syndromes or acute pulmonary edema (Rhoney & Peacock, 2009).
    9) NITROGLYCERIN/ADULT DOSE
    a) Begin infusion at 10 to 20 mcg/min and increase by 5 or 10 mcg/min every 5 to 10 minutes until the desired hemodynamic response is achieved (American Heart Association, 2005). Maximum rate 200 mcg/min (Rhoney & Peacock, 2009).
    10) NITROGLYCERIN/PEDIATRIC DOSE
    a) Usual Dose: 29 days or Older: 1 to 5 mcg/kg/min continuous IV infusion. Maximum 60 mcg/kg/min (Laitinen et al, 1997; Nam et al, 1989; Rasch & Lancaster, 1987; Ilbawi et al, 1985; Friedman & George, 1985).

Enhanced Elimination

    A) SUMMARY
    1) No studies have addressed the utilization of extracorporeal elimination techniques in poisoning with this agent.

Case Reports

    A) ADULT
    1) A 41-year-old woman ingested 4.5 g venlafaxine, 500 mg diphenhydramine, and 50 mg thiothixene and subsequently developed severe CNS depression requiring intubation. On presentation to the ED, she was also hypertensive (BP 168/101 mmHg) and tachycardic (106 bpm). Serum electrolytes were normal. ECG revealed sinus tachycardia and nonspecific T wave changes. An arterial blood gas showed a pH of 7.34, PaCO2 38, and PaO2 377. Decontamination with activated charcoal and sorbitol was administered following normal saline gastric lavage. The patient was responsive to verbal commands approximately one hour after admission to the ED. Supportive care was continued and she was extubated 9 hours postingestion with subsequent full recovery(Fantaskey & Burkhart, 1995).

Summary

    A) VENLAFAXINE
    1) ADULTS: Doses larger than 1500 mg have been associated with seizures. Life-threatening cardiovascular toxicity manifested by ventricular dysrhythmias are associated with doses larger than 8000 mg.
    2) PEDIATRIC: Doses less than 5.5 mg/kg in children are unlikely to result in toxicity.
    B) DESVENLAFAXINE
    1) ADULT: Limited data. In a pre-marketing study, 4 adults ingested desvenlafaxine succinate (4000 mg [desvenlafaxine alone], 900, 1800 and 5200 mg [in combination with other drugs] and each patient recovered. Adverse events reported and likely associated with a 5-day overdose of greater than 600 mg daily, included: headache, nausea/vomiting, diarrhea, agitation, dizziness, paresthesia, and tachycardia.
    2) PEDIATRIC: An 11-month-old ingested 600 mg of desvenlafaxine succinate and recovered completely with treatment.
    C) THERAPEUTIC DOSE: VENLAFAXINE: ADULT: 37.5 to 75 mg/day orally; MAX dose of 375 mg/day of immediate-release and 225 mg of extended-release. DESVENLAFAXINE: ADULT: 50 mg daily; PEDIATRIC: Not approved for use.

Therapeutic Dose

    7.2.1) ADULT
    A) VENLAFAXINE
    1) Initial: 75 mg/day in 2 to 3 divided doses (Prod Info Effexor(R) oral tablets, 2010).
    2) Maintenance: 75 to 225 mg/day in 2 to 3 divided doses is generally effective for patients with moderate depression. In more severely depressed patients, doses up to 375 mg/day in 3 divided doses have been reported. MAXIMUM DOSE: 375 mg/day (Prod Info Effexor(R) oral tablets, 2010).
    B) VENLAFAXINE EXTENDED-RELEASE
    1) Initial dose: 75 mg once daily. MAXIMUM DOSE: 225 mg/day (Prod Info Effexor XR (R) extended-release oral capsules, 2010).
    C) DESVENLAFAXINE
    1) 50 mg orally once daily (Prod Info KHEDEZLA oral extended-release tablets , 2013; Prod Info PRISTIQ(R) oral extended-release tablets, 2013).
    7.2.2) PEDIATRIC
    A) Venlafaxine has not been approved for use in pediatric patients (Prod Info Effexor(R) oral tablets, 2010; Prod Info Effexor XR (R) extended-release oral capsules, 2010).
    B) Desvenlafaxine has not been approved for use in pediatric patients (Prod Info KHEDEZLA oral extended-release tablets , 2013; Prod Info PRISTIQ(TM) oral extended-release tablets, 2008).

Minimum Lethal Exposure

    A) VENLAFAXINE
    1) SUMMARY: In postmarketing safety studies, overdosage of extended-release venlafaxine may be associated with an increased risk of fatal outcome as compared with other SSRI antidepressants, but lower than that observed with tricyclic antidepressants. There have been reports of fatal toxicity with venlafaxine alone at doses as low as approximately 1 g (Cobalt Pharmaceuticals Inc, 2008).
    2) CASE REPORT: A 40-year-old man intentionally ingested 19 g of extended release venlafaxine (90 150-mg venlafaxine and 75 75-mg venlafaxine extended-release tablets) and was admitted within 45 minutes of exposure, and was alert with mild tachycardia. An initial ECG showed tachycardia with no evidence of ischemia. Decontamination included activated charcoal and whole bowel irrigation. Toxicology screening was negative. Over the course of several hours the patient developed 2 seizures and was admitted to the intensive care. Approximately 9 hours after exposure, the patient was increasingly lethargic and developed refractory ventricular fibrillation (VF). The authors suggest that sodium channel toxicity was responsible for the fatal dysrhythmia. Although the patient developed no evidence of ventricular ectopy, the ECG showed a new onset of QRS widening (158 msec) and QTc prolongation (564 msec) shortly before the patient's death (Bosse et al, 2008).
    3) CASE REPORT: Following an intentional ingestion of 30 g extended release venlafaxine, a 39-year-old woman developed hypotension, decreased level of consciousness, tachycardia and perforated bowel. Venlafaxine and O-desmethylvenlafaxine levels, measured by high-performance liquid chromatography, peaked 24 hours after ingestion at 21.82 and 3.33 mg/L, respectively. The patient died on hospital day 43 (Mazur et al, 2003).
    4) CASE REPORTS: Four healthy women with no history of cardiovascular disease were admitted with acute left ventricular (LV) failure following overdose of venlafaxine. Three patients died and one completely recovered. The amount of venlafaxine ranged from 3150 to 13500 mg (extended-release preparations in 2 cases). LV ejection fraction on echocardiography was between 15% and 18% in 3 patients. A 38-year-old woman ingested 4200 mg and developed serotonin syndrome and LV failure and was improving, but died on day 11 of refractory hypoxemia. The second patient, a 35-year-old woman ingested 90 (150 mg extended-release) tablets and developed generalized seizures and LV failure (15% ejection fraction). At 17 hours, despite aggressive care the patient died of a sudden cardiac arrest. The final patient, a 65-year-old woman ingested 7000 mg of immediate-release venlafaxine and was comatose within 7 hours and died 7 hours later. The patient developed low cardiac output (1 L/min) and died of refractory cardiogenic shock; an echocardiogram was not performed (Batista et al, 2013).

Maximum Tolerated Exposure

    A) VENLAFAXINE
    1) ADULT
    a) CASE SERIES
    1) In a prospective review of consecutive patients presenting with a venlafaxine alone overdose between January 1997 and December 2007, 369 occasions of venlafaxine overdoses (median dose: 1500 mg; Interquartile Range (IQR): 600 to 3000 mg; range: 75 to 13500 mg) in 273 patients were observed with no reports of dysrhythmias and only minor ECG changes. Tachycardia was observed in 54% of patients and mild hypertension was reported in 40% of patients; severe hypotension and hypertension were uncommon. Conduction disturbances were found in 7 patients with 5 patients having a preexisting history of conduction disturbances. Alterations in QRS and QT interval were considered mild (the median maximum QRS width was 85 ms (IQR: 80 to 90 ms; range: 70 to 145 ms; 24 patients had a QRS of greater than or equal to 120 ms). The findings suggest that a mild exposure does not result in cardiotoxicity; however, doses greater than 8 g may produce significant cardiotoxicity (Isbister, 2009).
    b) SEVERE EXPOSURE (8 GRAMS OR GREATER)
    1) CASE REPORT: A 24-year-old woman who ingested 8.85 g venlafaxine developed seizures, cardiac dysrhythmias and late onset rhabdomyolysis, but recovered fully. The maximum plasma venlafaxine level recorded was 10.5 mg/L (Oliver et al, 2002).
    2) CASE REPORT: A 45-year-old woman ingested 10.9 g of venlafaxine and 35 mg of clonazepam and subsequently developed somnolence, severe hypotension, myoclonic jerking, and intraventricular block (Rosen et al, 1997).
    3) CASE REPORT: A 33-year-old woman ingested 8.4 g of venlafaxine and developed right axis deviation, a prolonged QT interval (469 msec) and a prolonged QRS complex (129 msec). In addition, she experienced a grand mal seizure. It is suggested that cardiotoxicity is more likely in patients with the poor CYP2D6 metabolizer phenotype (Blythe & Hackett, 1999).
    c) MILD TO MODERATE EXPOSURES (LESS THAN 8 GRAMS)
    1) CASE REPORT: COMBINATION OVERDOSE: A 30-year-old woman was thought to have intentionally ingested a one month supply of 150 mg venlafaxine (estimate 4.5 g), 300 mg bupropion, and 3 mg lorazepam that were prescribed 3 days earlier. The patient developed tachycardia, fever (40.6 degrees C), a prolonged QRS complex with a deep slurred S wave, respiratory distress, serotonin syndrome and rhabdomyolysis. Her creatine kinase and alanine transaminase levels peaked at 107,895 Units/L and 2453 Units/L, respectively, approximately 43 and 102 hours after exposure. She was extubated on day 7 and the patient was found to have persistent choreoathetosis with frontal releasing signs, impaired recent memory, language and executive function. A MRI performed 7 weeks after exposure showed bilateral pallidal necrosis (Lin et al, 2012).
    2) CHRONIC ABUSE/CASE REPORT: A 35-year-old man, with a history of schizophrenia and generalized anxiety disorder, presented to the ED seeking to refill a prescription for venlafaxine to suppress his severe anxiety. He reportedly said that his prior prescription was "stolen", but venlafaxine had been discontinued 1 month previously by his primary psychiatrist due to abuse. Upon chart review, the patient was being treated with risperidone for his schizophrenia, but his compliance was inconsistent. He denied issues with psychotic, depressive, or manic symptoms and claimed he was misdiagnosed as a schizophrenic. He also reported a prior history of substance abuse (ie, alcohol, benzodiazepines, cocaine, and marijuana), but refused a toxicology screen. The patient was admitted involuntarily to the psychiatric unit and was continued on risperidone and started on sertraline for generalized anxiety. As the patient improved and rapport was built, he reported taking 10 to 15 tablets of 225 mg (2250 to 3375 mg/day) extended-release venlafaxine daily and stated it made him feel "full of energy" and "alive" and helped him to feel focused. He would often chew the tablets to make the drug work faster. During his hospitalization, symptoms of withdrawal were absent and his vital signs remained normal (Namdari, 2013).
    3) CASE REPORT: A 46-year-old woman intentionally ingested 3150 mg of immediate-release and 2100 mg of extended-release venlafaxine and developed tonic-clonic seizures and hypotension. An echocardiogram showed acute left ventricular failure with an ejection fraction of 18%. With inotropic support, LV function improved by day 3 (ejection fraction 65%) and the patient recovered completely (Batista et al, 2013).
    4) CASE REPORT: A 27-year-old man intentionally ingested 6.3 g of extended-release venlafaxine resulting in respiratory failure and eosinophilic pneumonia. He was intubated and treated with empiric antibiotics, vasopressors, and corticosteroids. Seven days later he was discharged from the ICU without further event (Paparrigopoulos et al, 2011).
    5) CASE REPORT: An adult patient who ingested an estimated 6.75 g recovered with no sequelae (Prod Info Effexor(R) oral tablets, 2010).
    6) CASE REPORT: Serotonin syndrome and rhabdomyolysis occurred in a 21-year-old woman after ingesting 7.8 g of the extended-release venlafaxine. Her plasma CK levels, 5 hours and 41 hours after ingestion, were 43 Units/L and 42,340 Units/L, respectively. She developed somnolence and disorientation with a Glasgow Coma Scale of 4-6-3, nausea, vomiting, diaphoresis, and sinus tachycardia 160 to 170 beats per minute, and tonic-clonic seizures. Following supportive care, her CK level decreased to 1276 Units/L (Hanekamp et al, 2005).
    7) CASE REPORT: A 38-year-old man developed chest pain without ECG or cardiac enzyme changes following an ingestion of 4050 mg of crushed extended-release venlafaxine capsules (Sattar et al, 2003).
    8) CASE REPORT: A 34-year-old woman experienced a generalized seizure approximately 1 hour after ingesting 1400 to 1500 mg of venlafaxine (Leaf, 1998).
    9) CASE REPORT: A 29-year-old man ingested 2.8 g of venlafaxine and experienced seizures 2 and 4 hours after ingestion (Coorey & Wenck, 1998).
    10) CASE REPORT: A 41-year-old woman suffered severe CNS depression following ingestion of 4.5 g venlafaxine. Recovery was complete (Fantaskey & Burkhart, 1995).
    d) PEDIATRIC
    1) CASE SERIES: In a retrospective series of 14 pediatric cases of venlafaxine ingestion with doses ranging from 18.75 to 75 mg (2.1 to 5.5 mg/ kg), only one patient developed minor effects (lethargy) (Herrington & Gorman, 1996). Children ingesting 5.5 mg/kg or less can be managed at home.
    B) DESVENLAFAXINE
    1) ADULT
    a) Limited data. In a premarketing study, 4 adults ingested desvenlafaxine succinate (4000 mg [desvenlafaxine alone], 900, 1800 and 5200 mg [in combination with other drugs] and each patient recovered. Adverse events reported and likely associated with a 5-day overdose of greater than 600 mg daily, included: headache, nausea/vomiting, diarrhea, agitation, dizziness, paresthesia, and tachycardia (Prod Info PRISTIQ(TM) oral extended-release tablets, 2008).
    2) PEDIATRIC
    a) An 11-month-old inadvertently ingested 600 mg of desvenlafaxine succinate and recovered completely with treatment (Prod Info PRISTIQ(TM) oral extended-release tablets, 2008).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) VENLAFAXINE
    a) Therapeutic serum levels of venlafaxine have not been defined. After single oral doses of 25, 75, and 150 mg, mean peak serum concentrations were 37, 102, and 163 nanograms/mL, respectively, in one study. Corresponding levels of o-desmethylvenlafaxine, its major active metabolite, were 61, 168, and 325 nanograms/mL following these doses (Klamerus et al, 1992).
    b) After doses of 25, 75, and 150 mg every 8 hours for 3 days, mean peak serum levels were 53, 167, and 393 nanograms/mL; corresponding levels of O-desmethylvenlafaxine were 148, 397, and 686 nanograms/mL (Klamerus et al, 1992). With multiple doses, steady state concentrations of venlafaxine and O-desmethylvenlafaxine are achieved within three days and both drugs demonstrate linear kinetics for doses between 75 and 450 mg per day (Prod Info Effexor(R) oral tablets, 2010).
    c) After a single 150 mg dose, peak plasma levels of venlafaxine normally occur 2.4 hours after oral dosing, reaching 33 to 172 mcg/L (Pascale et al, 2005).
    d) In patients experiencing improved depression symptoms during venlafaxine therapy, median serum level was 54 mcg/L. Serum levels varied between 10.1 and 200 mcg/L. The median serum level of venlafaxine plus its active metabolite, O-desmethylvenlafaxine, was 193 mcg/L (Charlier et al, 2002).
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) SURVIVAL CONCENTRATIONS
    a) COMBINATION OVERDOSE: In one case, a woman who ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g developed coma, seizures, rigidity, and hyperreflexia. Plasma concentrations of lamotrigine, diazepam, and venlafaxine were 42.4 mg/L (therapeutic range, 1 to 4 mg/L), 560 mcg/L (therapeutic range, 200 to 500 mcg/L), and 1254 mcg/L (therapeutic range, 30 to 150 mcg/L), respectively. Following treatment with intravenous lipid emulsion, all symptoms resolved completely (Dagtekin et al, 2011).
    b) Peak plasma levels following acute ingestions of 2.75 and 2.5 grams were 6.24 and 2.35 mcg/mL respectively, and the peak plasma levels of O-desmethylvenlafaxine were 3.37 and 1.30 mcg/mLrespectively (Prod Info Effexor(R) oral tablets, 2010).
    c) Peak plasma levels were 12,000 and 1,200 nanograms/mL, respectively, for venlafaxine and O-desmethylvenlafaxine after overdose of 5.6 grams (Kokan & Dart, 1996).
    d) A 24-year-old woman ingested 8.85 g venlafaxine was found to have a plasma venlafaxine level of 10.5 mg/L. She recovered fully (Oliver et al, 2002).
    e) CASE REPORT: After ingesting 4.5 g of venlafaxine, a 25-year-old woman developed a generalized seizure, sinus tachycardia (150 bpm), incomplete right bundle branch block, QRS duration of 100 msec, prolonged QTc duration (0.46 seconds), and rhabdomyolysis (CK 52,600 Units/L). Following supportive care, she was discharged to the psychiatry department on day 5. The serum concentration of venlafaxine and desmethylvenlafaxine were 251 mcg/L and 319 mcg/L, respectively, 37 hours after ingestion (Pascale et al, 2005).
    f) CASE REPORT: A woman ingested 5 g venlafaxine and was found unconscious with seizures and developed hypotension. Electrocardiogram showed a right bundle branch block with a QRS duration of 144 ms and 428 ms corrected QT intervals. Plasma venlafaxine concentration was 25.8 micromol/L (gas chromatography; therapeutic range 0.25 to 0.85, quantification limit 0.2). By day 3, the patient had improved following an epinephrine infusion and supportive care and was extubated successfully (Megarbane et al, 2007).
    2) POSTMORTEM CONCENTRATIONS
    a) CASE REPORT: An adult male intentionally ingested 19 g of venlafaxine extended-release tablets (90 150-mg venlafaxine extended-release and 75 75-mg venlafaxine extended-release) and died approximately 9 hours after exposure of refractory ventricular fibrillation. His venlafaxine blood concentration obtained about 5 hours after exposure was 9000 ng/mL and the O-desmethylvenlafaxine blood concentration (metabolite) was 3000 ng/mL. A postmortem concentration obtained from a peripheral site was 5500 ng/mL. No other substances were found on postmortem exam (Bosse et al, 2008).
    b) CASE REPORTS: Two women were admitted with CNS depression, tachycardia and hypotension following large ingestions of venlafaxine. Both died of cardiac arrest within 24 hours of admission. Autopsy was performed at 48 hours after death in both patients; the first patient had an estimated ingestion of 14.7 g, with a venlafaxine concentration of 95 mcg/mL (metabolite O-desmethyl-venlafaxine 8 mcg/mL). The second patient also had a large ingestion (exact amount unknown) with a venlafaxine concentration of 79 mcg/mL (metabolite O-desmethyl-venlafaxine 17.7 mcg/mL) (Hojer et al, 2008).
    c) Postmortem venlafaxine level was 89,000 nanograms/mL in one case (Dahl et al, 1996).
    d) A 45-year-old woman died after ingestion of 8.4 g venlafaxine. Venlafaxine level was 14 mg/L on presentation to the hospital and 16 mg/L postmortem (Banham, 1998).
    e) Postmortem venlafaxine concentrations of 53 mg/L and 78 mg/L have been reported in two other cases. Venlafaxine undergoes postmortem distribution from tissues into blood (Jaffe et al, 1999).
    f) EXTENDED RELEASE: Following an intentional ingestion of 30 g of extended release venlafaxine, a 39-year-old woman died on hospital day 43. Venlafaxine and o-desmethylvenlafaxine levels peaked 24 hours after ingestion at 21.82 mg/L and 3.33 mg/L, respectively (Mazur et al, 2003).
    g) CASE REPORT: After ingesting an unknown amount of venlafaxine, a 34-year-old man developed signs and symptom of serotonin syndrome, and developed rhabdomyolysis, renal and cardiac failure and died a day later. The postmortem histology examination of the liver showed confluent coagulative necrosis, occurring mainly as an acinar zone 3 pattern (notably perivenular and centrilobular) with midzone involvement to a lesser extent. Macrovesicular steatosis with a scattered inflammatory infiltrate indicating acute hepatitis was observed. The venlafaxine concentration just before death was 18.6 mg/L (therapeutic range 0.07 and 0.27 mg/L); liver concentration was 46 mg/kg (Shaw & Sheard, 2005).

Pharmacologic Mechanism

    A) VENLAFAXINE
    1) Venlafaxine, a bicyclic antidepressant, is a potentiator of neurotransmitter activity in the CNS. Both venlafaxine and its active metabolite, O-desmethylvenlafaxine, inhibit the neuronal uptake of serotonin, noradrenaline, and dopamine in order of decreasing potency, without inhibiting monoamine oxidase. It does not show the degree of anticholinergic, sedative, or cardiovascular effects other antidepressants have been shown to exhibit.
    a) In radioligand binding studies venlafaxine shows moderate affinity for the serotonin transported and low affinity for the norepinephrine transporter despite its potent serotonin and norepinephrine reuptake blocking properties (Beique et al, 1998).
    2) Animal studies have demonstrated no affinity for central muscarinic- cholinergic, dopaminergic, histaminic, opioid (mu), benzodiazepine, or alpha1-adrenergic receptors for venlafaxine or O-desmethylvenlafaxine. Venlafaxine has been shown to inhibit neuronal activity in the locus coeruleus of the brain. Other antidepressant properties include its ability to reverse reserpine hypothermia and to cause pineal beta-adrenergic subsensitivity (Haskins et al, 1985; Klamerus et al, 1992; Muth et al, 1986; Saletu et al, 1992) Yardley et al, 1990).
    3) Venlafaxine is a racemic mixture; while the pharmacologic profile of the levo (-) isomer is similar to that of the racemate, the dextro (+) isomer is primarily a serotonin uptake inhibitor (Klamerus et al, 1992).
    B) DESVENLAFAXINE
    1) Desvenlafaxine succinate is a potent and selective serotonin and norepinephrine reuptake inhibitor. Clinical efficacy is thought to be related to the potentiation of these neurotransmitters in the central nervous system (Prod Info PRISTIQ(TM) oral extended-release tablets, 2008).

Physical Characteristics

    A) DESVENLAFAXINE is a white to off-white crystalline powder that is sparingly soluble in dimethyl sulfoxide; solubility is pH dependent (Prod Info KHEDEZLA oral extended-release tablets , 2013).
    B) DESVENLAFAXINE SUCCINATE is a white to off-white powder that is soluble in water; solubility is pH dependent. The octanol:aqueous system partition coefficient is 0.21 (pH 7) (Prod Info PRISTIQ(R) oral extended-release tablets, 2013).
    C) VENLAFAXINE HYDROCHLORIDE is a white to off-white crystalline solid with a solubility of 572 mg/mL in water (adjusted to 0.2 molar (M) with sodium chloride). The octanol:water (0.2 M sodium chloride) partition coefficient is 0.43 (Prod Info Effexor(R) oral tablets, 2012).

Molecular Weight

    A) DESVENLAFAXINE: 263.38 (Prod Info KHEDEZLA oral extended-release tablets , 2013)
    B) DESVENLAFAXINE SUCCINATE MONOHYDRATE: 399.48 (Prod Info PRISTIQ(R) oral extended-release tablets, 2013)
    C) VENLAFAXINE HYDROCHLORIDE: 313.87 (Prod Info Effexor(R) oral tablets, 2012)

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) Adesanya A & Varma SL: Overdose of venlafaxine - a new antidepressant (letter). Med J Australia 1997; 167:54.
    3) Allen KR: Interference by venlafaxine ingestion in the detection of tramadol by liquid chromatography linked to tandem mass spectrometry for the screening of illicit drugs in human urine. Clin Toxicol (Phila) 2006; 44(2):147-153.
    4) 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.
    5) American College of Medical Toxicology: ACMT position statement: guidance for the use of intravenous lipid emulsion. J Med Toxicol 2016; Epub:Epub-.
    6) American Heart Association: 2005 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2005; 112(24 Suppl):IV 1-203. Available from URL: http://circ.ahajournals.org/content/vol112/24_suppl/. As accessed 12/14/2005.
    7) Anon: FDC Reports: The Pink Sheet 1993; 55:10-11.
    8) Anon: Venlafaxine: a new dimension in antidepressant pharmacotherapy. J Clin Psychiatr 1993a; 54:119-126.
    9) Ansary A, Ibhanesebhor S, & Manjunatha C: Myoclonic seizures in a preterm baby: is this a presentation of venlafaxine withdrawal?. Singapore Med J 2014; 55(4):e57-e59.
    10) Appleton R: Lorazepam vs diazepam in the acute treatment of epileptic seizures and status epilepticus.. Dev Med Child Neuro 1995; 37:682-688.
    11) Baboolal NS: Venlafaxine withdrawal syndrome. Report of seven cases in Trinidad (letter). J Clin Psychopharmacol 2004; 24:229-231.
    12) Ballon JS & Schulman MC: Venlafaxine and the rapid development of anasarca. J Clin Psychopharmacol 2006; 26(1):97-98.
    13) Banham NDG: Fatal venlafaxine overdose(letter). MJA 1998; 169:445-446.
    14) Batista M, Dugernier T, Simon M, et al: The spectrum of acute heart failure after venlafaxine overdose. Clin Toxicol (Phila) 2013; 51(2):92-95.
    15) Beique JC, Lavoie N, & de Montigny C: Affinities of venlafaxine and various reuplate inhibitors for the serotonin and norepinephrine transporters. Eur J Pharmacol 1998; 349:129-132.
    16) Benazzi F: Anticholinergic toxic syndrome with venlafaxine-desipramine combination (letter). Pharmacopsychiat 1998; 31:36-37.
    17) Benazzi F: Venlafaxine-fluoxetine interaction (letter). J Clin Psychopharmacol 1999; 19(1):96-98.
    18) Berle JO, Steen VM, Aamo TO, et al: Breastfeeding during maternal antidepressant treatment with serotonin reuptake inhibitors: infant exposure, clinical symptoms, and cytochrome P450 genotypes. J Clin Psychiatry 2004; 65:1228-1234.
    19) Blass DM & Pearson VE: SIADH with multiple antidepressants in a geriatric patient (letter). J Clin Psychiatry 2000; 61(6):448-449.
    20) Blythe D & Hackett LP: Case report. Cardiovascular and neurological toxicity of venlafaxine. Hum Exp Toxicol 1999; 18:309-313.
    21) Bond GR, Vinks AA, & Steele PE: Venlafaxine (VFX) and a false positive Abbott Axsym immunoassay for phencyclidine (abstract). J Toxicol Clin Toxicol 2001; 39(5):492.
    22) Bosse GM, Spiller HA, & Collins AM: A fatal case of venlafaxine overdose. J Med Toxicol 2008; 4(1):18-20.
    23) Boyd IW: Comment: hyponatremia with venlafaxine (letter). Ann Pharmacother 1998a; 32:981.
    24) Boyd IW: Venlafaxine withdrawal reactions. Med J Aust 1998; 169:91-92.
    25) Boyer EW & Shannon M: The serotonin syndrome. N Eng J Med 2005; 352(11):1112-1120.
    26) Boyer EW, Duic PA, & Evans A: Hyperinsulinemia/euglycemia therapy for calcium channel blocker poisoning. Ped Emerg Care 2002; 18:36-37.
    27) Bridge JA, Iyengar S, Salary CB, et al: Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA 2007; 297(15):1683-1696.
    28) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    29) Brubacher JF, Lurin MJ, & Hirsch S: Serotonin syndrome from venlafaxine- tranylcypromine interaction (abstract). J Toxicol Clin Toxicol 1995; 33:523.
    30) Brvar M, Kozelj G, & Masic LP: Hypoglycemia in venlafaxine overdose: a hypothesis of increased glucose uptake. Eur J Clin Pharmacol 2015; 71(2):261-262.
    31) Buckley NA & Faunce TA: Atypical antidepressants in overdose. Drug Safety 2003; 26(8):539-551.
    32) Caravati EM, Knight HH, & Linscott MS: Esophageal laceration and charcoal mediastinum complicating gastric lavage. J Emerg Med 2001; 20:273-276.
    33) Cardona X, Avila A, & Castellanos P: Venlafaxine-associated hepatitis (letter). Ann Intern Med 2000; 132(5):417.
    34) Carnes CA, Pickworth KK, Votolato NA, et al: Elevated defibrillation threshold with venlafaxine therapy. Pharmacotherapy 2004; 24(8):1095-1098.
    35) Cassidy EM & O'Kearne V: Neuroleptic malignant syndrome after venlafaxine (letter). Lancet 2000; 355:2164-2165.
    36) 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.
    37) Chan B, Whyte I, Dawson A, et al: Use of neostigmine for the management of drug induced ileus in severe poisonings. J Med Toxicol 2005; 1(1):18-22.
    38) Chand PK, Kalyani GS, & Murthy P: Venlafaxine-associated hypomania in unipolar depression. Can J Psychiatry 2004; 49(7):496-.
    39) Charlier C, Pinto E, Ansseau M, et al: Venlafaxine: the relationship between dose, plasma concentration and clinical response in depressive patients. J Psychopharmacol 2002; 16(4):369-372.
    40) 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.
    41) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    42) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    43) Claassen JAHR & Gelissen HPMM: The serotonin syndrome (letter). N Eng J Med 2005; 352(23):2455.
    44) Cobalt Pharmaceuticals Inc: Dear Healthcare Professional Letter for VENLAFAXINE XR(R) (hcl extended release oral capsules). Health Canada. Ottawa, Canada. 2008. Available from URL: http://www.hc-sc.gc.ca/dhp-mps/alt_formats/hpfb-dgpsa/pdf/medeff/venlaflaxine_hpc-cps-eng.pdf.
    45) Combes A, Peytavin G, & Theron D: Conduction disturbances associated with venlafaxinw (letter). Ann Intern Med 2001; 134(2):166-167.
    46) Coorey AN & Wenck DJ: Venlafaxine overdose (letter). Med J Aust 1998; 168:523.
    47) Dagtekin O, Marcus H, Muller C, et al: Lipid therapy for serotonin syndrome after intoxication with venlafaxine, lamotrigine and diazepam. Minerva Anestesiol 2011; 77(1):93-95.
    48) Dahl B, Crouch BI, & Rollins D: Death from venlafaxine overdose (Effexor(R)). J Toxicol Clin Toxicol 1996; 34:557.
    49) Dallal A & Chouinard G: Withdrawal and rebound symptoms associated with abrupt discontinuation of venlafaxine (letter). J Clin Psychopharmacol 1998; 18:343-344.
    50) Dalle S, Becuwe C, Balme B, et al: Venlafaxine-associated psoriasiform palmoplantar keratoderma and subungual hyperkeratosis. Br J Dermatol 2006; 154(5):999-1000.
    51) Daniels RJ: Serotonin syndrome due to venlafaxine overdose. J Accid Emerg Med 1998; 15:333-334.
    52) De Negri M & Baglietto MG: Treatment of status epilepticus in children. Paediatr Drugs 2001; 3(6):411-420.
    53) Djogovic D, Hudson D, & Jacka M: Gastric bezoar following venlafaxine overdose. Clin Toxicol (Phila) 2007; 45(6):735-.
    54) Drent M, Singh S, Gorgels AP, et al: Drug-induced pneumonitis and heart failure simultaneously associated with venlafaxine. Am J Respir Crit Care Med 2003; 167(7):958-961.
    55) Dubnov-Raz G, Juurlink DN, Fogelman R, et al: Antenatal use of selective serotonin-reuptake inhibitors and QT interval prolongation in newborns. Pediatrics 2008; 122(3):e710-e715.
    56) Durback LF & Scharman EJ: Seizures associated with venlafaxine overdose (abstract). J Toxicol Clin Toxicol 1996; 34:557.
    57) Egger C, Muehlbacher M, Nickel M, et al: A case of recurrent hyponatremia induced by venlafaxine. J Clin Psychopharmacol 2006; 26(4):439-.
    58) Einarson A, Fatoye B, & Sarkar M: Pregnancy outcome following gestational exposure to venlafaxine: a multicenter prospective controlled study. Am J Psychiatry 2001; 158:1728-1730.
    59) Einarson A, Fatoye B, Sarkar M, et al: Pregnancy outcome following gestational exposure to venlafaxine: a multicenter prospective controlled study. AM J Psychiatry 2001a; 158:1728-1730.
    60) Einarson TR & Einarson A: Newer antidepressants in pregnancy and rates of major malformations: a meta-analysis of prospective comparative studies. Pharmacoepidemiol.Drug Saf 2005; 14(12):823-827.
    61) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    62) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    63) Fangio P, DeJonghe B, Appere-De-Vecchi C, et al: Acute heart failure associated with venlafaxine poisoning. Am J Emerg Med 2007; 25(2):210-211.
    64) Fantaskey A & Burkhart KK: A case report of venlafaxine toxicity. Clin Tox 1995; 33(4):359-361.
    65) Ferreira E, Carceller AM, Agogue C, et al: Effects of selective serotonin reuptake inhibitors and venlafaxine during pregnancy in term and preterm neonates. Pediatrics 2007; 119(1):52-59.
    66) Fleisch MC, Blauer F, Gubler JG, et al: Eosinophilic pneumonia and respiratory failure associated with venlafaxine treatment. Eur Respir J 2000; 15(1):205-208.
    67) Francesconi G, Orsolini L, Papanti D, et al: Venlafaxine as the 'baby ecstasy'? Literature overview and analysis of web-based misusers' experiences. Hum Psychopharmacol 2015; 30(4):255-261.
    68) Friedman WF & George BL : Treatment of congestive heart failure by altering loading conditions of the heart. J Pediatr 1985; 106(5):697-706.
    69) Garber A & Gregory RJ: Benztropine in the treatment of venlafaxine- induced sweating (letter). J Clin Psychiatr 1997; 58:176-177.
    70) Giang DW & McBride MC : Lorazepam versus diazepam for the treatment of status epilepticus. Pediatr Neurol 1988; 4(6):358-361.
    71) Gillman PK: Ecstasy, serotonin syndrome and the treatment of hyperpyrexia (letter). MJA 1997; 167:109-111.
    72) Gillman PK: Successful treatment of serotonin syndrome with chlorpromazine (letter). MJA 1996; 165:345.
    73) Godkar D, Stensby J, Sinnapunayagam S, et al: Venlafaxine induced acute myocardial infarction with normal coronary arteries. Am J Ther 2009; 16(4):365-366.
    74) Goldberg RJ & Huk M: Serotonin syndrome from trazodone and buspirone (letter). Psychosomatics 1992; 33:235-236.
    75) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    76) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    77) Graham PM: Successful treatment of the toxic serotonin syndrome with chlorpromazine (letter). Med J Australia 1997; 166:166-167.
    78) Gupta AK & Saravay SM: Venlafaxine-induced hyponatremia (letter). J Clin Psychopharmacol 1997; 17:223-225.
    79) Hanekamp BB, Zijlstra JG, Tulleken JE, et al: Serotonin syndrome and rhabdomyolysis in venlafaxine poisoning: a case report. Neth J Med 2005; 63(8):316-318.
    80) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    81) Haskins JT, Moyer JA, & Muth EA: DMI, WY-45,030, WY-45,881 and ciramadol inhibit locus coeruleus activity. Eur J Pharmacol 1985; 115:139-146.
    82) Haukland LU, Kutzsche S, Hovden IA, et al: Neonatal seizures with reversible EEG changes after antenatal venlafaxine exposure. Acta Paediatr 2013; 102(11):e524-e526.
    83) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    84) Herrington LF & Gorman SE: Pediatric ingestion of effexor (venlafaxine) (abstract). J Toxicol Clin Toxicol 1996; 34:558.
    85) Hodgman M, Martin T, & Dean B: Severe serotonin syndrome secondary to venlafaxine and maintenance tranylcypromine therapy (abstract). J Toxicol Clin Toxicol 1995; 33:554.
    86) Hodgman MJ, Martin TG, & Krenzelok EP: Serotonin syndrome due to venlafaxine and maintenance tranylcypromine therapy. Hum Exp Toxicol 1997; 16:14-17.
    87) Hojer J, Hulting J, & Salmonson H: Fatal cardiotoxicity induced by venlafaxine overdosage. Clin Toxicol (Phila) 2008; 46(4):336-337.
    88) Howell C, Wilson AD, & Waring WS: Cardiovascular toxicity due to venlafaxine poisoning in adults: a review of 235 consecutive cases. Br J Clin Pharmacol 2007; 64(2):192-197.
    89) Hudson D & Djogovic D: Alternate venlafaxine kinetics in overdose patients. J Clin Pharm Ther 2007; 32(1):109-.
    90) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    91) Ilbawi MN, Idriss FS, DeLeon SY, et al: Hemodynamic effects of intravenous nitroglycerin in pediatric patients after heart surgery. Circulation 1985; 72(3 Pt 2):II101-II107.
    92) Ilett K, Kristensen J, Hackett P, et al: Distribution of venlafaxine and its O-desmethyl metabolite in human milk and their effects in breastfed infants. Br J Clin Pharmacol 2002; 53(1):17-22.
    93) Ilett KF, Hackett LP, Dusci LJ, et al: Distribution and excretion of venlafaxine and O-desmethylvenlafaxine in human milk. Br J Clin Pharmacol 1998; 45(5):459-62.
    94) Isbister GK : Electrocardiogram changes and arrhythmias in venlafaxine overdose. Br J Clin Pharmacol 2009; 67(5):572-576.
    95) Jaffe PD, Batziris HP, & van der Hoeven P: A study involving venlafaxine overdoses: comparison of fatal and therapeutic concentrations in postmortem specimens. J Forensic Sci 1999; 44(1):193-196.
    96) Jewell DP, Thompson IW, Sullivan BA, et al: Reversible focal myositis in a patient taking venlafaxine. Rheumatology (Oxford) 2004; 43(12):1590-1593.
    97) Johnson H, Bouman WP, & Lawton J: Withdrawal reaction associated with venlafaxine. Br Med J 1998; 317:787.
    98) Kelly CA, Dhaun N, Laing WJ, et al: Comparative toxicity of citalopram and the newer antidepressants after overdose. J Toxicol - Clin Toxicol 2004; 42:67-71.
    99) Khan A, Fabre LF, & Rudolph R: Venlafaxine in depressed outpatients. Psychopharmacol Bull 1991; 27:141-144.
    100) Khazaal Y: Mania after venlafaxine withdrawal in a patient with generalized anxiety disorder. Ann Pharmacother 2007; 41(2):359-360.
    101) Kirshenbaum LA, Mathews SC, & Sitar DS: Whole-bowel irrigation versus activated charcoal in sorbitol for the ingestion of modified-release pharmaceuticals. Clin Pharmacol Ther 1989; 46:264-271.
    102) Klamerus KJ, Maloney K, & Rudolph RL: Introduction of a composite parameter to the pharmacokinetics of venlafaxine and its active O-desmethyl- metabolite. J Clin Pharmacol 1992; 32:716-724.
    103) 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.
    104) Kline SS, Mauro LS, & Scala-Barnett DM: Serotonin syndrome versus neuroleptic malignant syndrome as a cause of death. Clin Pharmac 1989; 8:510-514.
    105) Klinger G, Frankenthal D, Merlob P, et al: Long-term outcome following selective serotonin reuptake inhibitor induced neonatal abstinence syndrome. J Perinatol 2011; 31(9):615-620.
    106) Kokan L & Dart RC: Life-threatening hypotension from venlafaxine overdose (abstract). J Toxicol Clin Toxicol 1996; 34:559.
    107) Kolecki P & Miller M: Isolated venlafaxine-induced serotonin syndrome (abstract). J Toxicol Clin Toxicol 1996; 34:558.
    108) Kumar VV , Oscarsson S , Friberg LE , et al: The effect of decontamination procedures on the pharmacokinetics of venlafaxine in overdose. Clin Pharmacol Ther 2009; 86(4):403-410.
    109) Laitinen P, Happonen JM, Sairanen H, et al: Amrinone versus dopamine-nitroglycerin after reconstructive surgery for complete atrioventricular septal defect. J Cardiothorac Vasc Anesth 1997; 11(7):870-874.
    110) Langford NJ, Martin U, Ruprah M, et al: Alternative venlafaxine kinetics in overdose. J Clin Pharm Ther 2002; 27:465-467.
    111) 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.
    112) Leaf EV: Comment: venlafaxine overdose and seizure (letter). Ann Pharmacother 1998; 32:135-136.
    113) Lessard E, Yessine MA, & Hamelin BA: Influence of CYP2D6 activity on the disposition and cardiovascular toxicity of the antidepressant agent venlafaxine in humans. Pharmacogenetics 1999; 9(4):435-443.
    114) Lin YH, Huang E, & Chien YL: Bilateral pallidal necrosis and cardiac toxicity in a patient with venlafaxine and bupropion overdose. Am J Psychiatry 2012; 169(9):993-994.
    115) Link MS, Berkow LC, Kudenchuk PJ, et al: Part 7: Adult Advanced Cardiovascular Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S444-S464.
    116) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    117) Macbeth R & Rajagopalan M: Venlafaxine withdrawal syndrome. Aust NZ J Med 1998; 28:218.
    118) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    119) Matoga M, Pehourcq F, & Titier K: Rapid high-performance liquid chromatographic measurement of venlafaxine and O-desmethylvenlafaxine in human plasma application to management of acute intoxications. J Chromatography B 2001; 760:213-218.
    120) Mazur JE, Doty JD, & Krygiel AS: Fatality related to a 30 g venlafaxine overdose. Pharmacotherapy 2003; 23:1668-1672.
    121) Meertens JH, Monteban-Kooistra WE, Ligtenberg JJ, et al: Severe hypoglycemia following venlafaxine intoxication: a case report. J Clin Psychopharmacol 2007; 27(4):414-415.
    122) Megarbane B, Bloch V, Deye N, et al: Pharmacokinetic/pharmacodynamic modelling of cardiac toxicity in venlafaxine overdose. Intensive Care Med 2007; 33(1):195-196.
    123) Miller T, McGoodwin L, & Hagemann T: Anticholinergic-type effects following a pediatric venlafaxine overdose. Clin Toxicol (Phila) 2008; 46(4):338-339.
    124) Mills KC: Serotonin syndrome: a clinical update. Med Toxicol 1997; 13:763-783.
    125) Mitchell WG: Status epilepticus and acute repetitive seizures in children, adolescents, and young adults: etiology, outcome, and treatment. Epilepsia 1996; 37(S1):S74-S80.
    126) Muth EA, Haskins JT, & Moyer JA: Antidepressant biochemical profile of the novel bicyclic compound WY-45,030, an ethyl cyclohexanol derivative. Biochem Pharmacol 1986; 35:4493-4497.
    127) Nakhai-Pour HR, Broy P, & Berard A: Use of antidepressants during pregnancy and the risk of spontaneous abortion. CMAJ 2010; Epub:Epub.
    128) Nam YT, Shin T, & Yoshitake J: Induced hypotension for surgical repair of congenital dislocation of the hip in children. J Anesth 1989; 3(1):58-64.
    129) Namdari B: Venlafaxine abuse in a patient with schizophrenia and prior history of substance dependence: a case report. J Addict Dis 2013; 32(4):393-395.
    130) 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.
    131) Ng B, Sanbrook GMC, Malouf AJ, et al: Venlafaxine and bilateral acute angle closure glaucoma. Med J Aust 2002; 176:241.
    132) Nimmagadda SR, Ryan DH, & Atkin SL: Neuroleptic malignant syndrome after venlafaxine (letter). Lancet 2000; 354:289-290.
    133) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    134) Oliver JJ, Kelly C, Jarvie D, et al: Venlafaxine poisoning complicated by a late rise in creatine kinase: two case reports. Hum Exp Toxicol 2002; 21:463-466.
    135) Pakalapati RK, Bolisetty S, Austin MP, et al: Neonatal seizures from in utero venlafaxine exposure. J Paediatr Child Health 2006; 42(11):737-738.
    136) Pan JJ & Shen WW: Serotonin syndrome induced by low-dose venlafaxine. Ann Pharmacother 2003; 37(2):209-211.
    137) Paparrigopoulos T, Tzavellas E, Karaiskos D, et al: Acute eosinophilic pneumonia after venlafaxine overdose. J Clin Psychopharmacol 2011; 31(2):258-259.
    138) Pascale P, Oddo M, Pacher P, et al: Severe rhabdomyolysis following venlafaxine overdose. Drug Monit 2005; 27(5):562-564.
    139) Patridge SJ, MacIver DH, & Solanki T: A depressed myocardium. Clin Toxicol 2000; 38(4):453-455.
    140) Peano C, Wahl M, & Keyes N: Seizures and ventricular tachycardia secondary to venlafaxine and lamotrigine (abstract). J Toxicol Clin Toxicol 1996; 34:558.
    141) 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.
    142) Pfeffer F & Grube M: An organic psychosis due to a venlafaxine-propafenone interaction. Int J Psychiatry Med 2001; 31:427-432.
    143) Phillips BB, Digmann RR, & Beck MG: Hepatitis associated with low-dose venlafaxine for postmenopausal vasomotor symptoms. Ann Pharmacother 2006; 40(2):323-327.
    144) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    145) Prior FH, Isbister GK, Dawson AH, et al: Serotonin toxicity with therapeutic doses of dexamphetamine and venlafaxine. Med J Aust 2002; 176:240-241.
    146) Product Information: ATIVAN(R) injection, lorazepam injection. Baxter Healthcare Corporation, Deerfield, IL, 2003.
    147) Product Information: Cordarone(R) oral tablets, amiodarone HCl oral tablets. Wyeth Pharmaceuticals Inc (per FDA), Philadelphia, PA, 2015.
    148) Product Information: EFFEXOR XR(R) extended-release oral capsules, venlafaxine hcl extended-release oral capsules. Wyeth Pharmaceuticals,Inc, Philadelphia, PA, 2006.
    149) Product Information: EFFEXOR XR(R) extended-release oral capsules, venlafaxine hydrochloride extended-release oral capsules. Wyeth Pharmaceuticals Inc, Philadelphia, PA, 2008.
    150) Product Information: EFFEXOR(R) oral tablets, venlafaxine hcl oral tablets. Wyeth Pharmaceuticals Inc, Philadelphia, PA, 2007.
    151) Product Information: Effexor XR (R) extended-release oral capsules, venlafaxine hydrochloride extended-release oral capsules. Wyeth Pharmaceuticals Inc., Philadelphia, PA, 2010.
    152) Product Information: Effexor XR(R) oral extended release capsules, venlafaxine HCl oral extended release capsules. Wyeth Pharmaceuticals Inc (per FDA), Philadelphia, PA, 2012.
    153) Product Information: Effexor XR(R) oral extended release capsules, venlafaxine HCl oral extended release capsules. Wyeth Pharmaceuticals Inc (per FDA), Philadelphia, PA, 2012a.
    154) Product Information: Effexor(R) oral tablets, venlafaxine HCl oral tablets. Wyeth Pharmaceuticals Inc (per FDA), Philadelphia, PA, 2012.
    155) Product Information: Effexor(R) oral tablets, venlafaxine hydrochloride oral tablets. Wyeth Pharmaceuticals Inc, Philadelphia, PA, 2010.
    156) Product Information: KHEDEZLA oral extended-release tablets , desvenlafaxine oral extended-release tablets. Par Pharmaceutical Companies, Inc. (per FDA), Spring Valley, NY, 2013.
    157) Product Information: Lidocaine HCl intravenous injection solution, lidocaine HCl intravenous injection solution. Hospira (per manufacturer), Lake Forest, IL, 2006.
    158) Product Information: NITROPRESS(R) injection for IV infusion, Sodium Nitroprusside injection for IV infusion. Hospira, Inc., Lake Forest, IL, 2007.
    159) Product Information: NITROPRESS(R) injection, sodium nitroprusside injection. Hospira,Inc, Lake Forest, IL, 2004.
    160) Product Information: PRISTIQ(R) oral extended-release tablets, desvenlafaxine oral extended-release tablets. Wyeth Pharmaceuticals Inc. (per FDA), Philadelphia, PA, 2013.
    161) Product Information: PRISTIQ(TM) oral extended-release tablets, desvenlafaxine oral extended-release tablets. Wyeth Pharmaceuticals Inc, Philadelphia, PA, 2008.
    162) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    163) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    164) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    165) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    166) 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.
    167) Rasch DK & Lancaster L: Successful use of nitroglycerin to treat postoperative pulmonary hypertension. Crit Care Med 1987; 15(6):616-617.
    168) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    169) Rhoney D & Peacock WF: Intravenous therapy for hypertensive emergencies, part 1. Am J Health Syst Pharm 2009; 66(15):1343-1352.
    170) Rosen P, Marek M, & Borys D: Coma, hypotension, and intraventricular block in a patient with venlafaxine and clonazepam overdose (abstract). J Tox- Clin Tox 1997; 35:548.
    171) Saletu B, Grunberger J, & Anderer P: Pharmacodynamics of venlafaxine evaluated by EEG brain mapping, psychometry and psychophysiology. Br J Clin Pharmacol 1992; 33:589-601.
    172) Santos PM, Lopez-Garcia P, Navarro JS, et al: False positive phencyclidine results caused by venlafaxine. Am J Psychiatry 2007; 164(2):349-.
    173) Sarma A & Horne MK: Venlafaxine-induced ecchymoses and impaired platelet aggregation. Eur J Haematol 2006; 77(6):533-537.
    174) Sattar SP, Grant KM, & Bhatia SC: A case of venlafaxine abuse. N Engl J Med 2003; 348:764-765.
    175) Schweizer E, Clary C, & Weise C: An open-label, dose-finding study of WY-45,030, a novel bicyclic antidepressant. Psychopharmacol Bull 1988; 24:195-197.
    176) Schweizer E, Weiss C, & Clary C: Placebo-controlled trial of venlafaxine for the treatment of major depression. J Clin Psychopharmacol 1991; 11:233-236.
    177) 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.
    178) Setz SC, Anderson DA, & Lawler: Acute venlafaxine overdose - a multicenter study (abstract). J Toxicol Clin Toxicol 1995; 33:496.
    179) Shaw MW & Sheard JDH: Fatal venlafaxine overdose with acinar zone 3 liver cell necrosis. Am J Forensic Med Pathol 2005; 26:367-368.
    180) Smith SW, Ling LJ, & Halstenson CE: Whole-bowel irrigation as a treatment for acute lithium overdose. Ann Emerg Med 1991; 20:536-539.
    181) 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.
    182) Sternbach H: The serotonin syndrome. Am J Psychiatr 1991; 148:705-713.
    183) Stone TE, Swanson C, & Feldman MD: Venlafaxine discontinuation syndrome and suicidal ideation: a case series. J Clin Psychopharmacol 2007; 27(1):94-95.
    184) Tenderich G, Dagge A, & Schulz U: Successful use of cardiac allograft from serotonin antagonist intoxication. Transplantation 2000; 72(3):529-530.
    185) Tenenbein M, Cohen S, & Sitar DS: Whole bowel irrigation as a decontamination procedure after acute drug overdose. Arch Int Med 1987; 147:905-907.
    186) Thorsson B, Snook C, & Thorgeirsson G: Survival after prolonged cardiac arrest from venlafaxine (abstract), European Association of Poisons Centres and Clinical Toxicologists. XX International Congress, Amsterdam, The Netherlands, 2000.
    187) Treichel M, Schwendener Scholl K, Kessler U, et al: Is there a correlation between venlafaxine therapy during pregnancy and a higher incidence of necrotizing enterocolitis?. World J Pediatr 2009; 5(1):65-67.
    188) Troy SM, Dilea C, & Martin PT: Bioavailability of once-daily venlafaxine extended release compared with the immediate-release formulation in healthy adult volunteers. Curr Ther Res 1997; 58:492-503.
    189) Turner RC, Nelson JE, Roberts BT, et al: Venlafaxine-associated interstitial pneumonitis. Pharmacotherapy 2005; 25(4):626-629.
    190) Tzallas PJ & Rynn KO: Extrapyramidal side effects secondary to venlafaxine (abstract). J Toxicol Clin Toxicol 1995; 33:518.
    191) Vale JA, Kulig K, American Academy of Clinical Toxicology, et al: Position paper: Gastric lavage. J Toxicol Clin Toxicol 2004; 42:933-943.
    192) 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.
    193) Vanden Hoek TL, Morrison LJ, Shuster M, et al: Part 12: cardiac arrest in special situations: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2010; 122(18 Suppl 3):S829-S861.
    194) Wang CP, Howell SR, & Scatina J: The disposition of venlafaxine enantiomers in dogs, rats, and humans receiving venlafaxine (abstract). Chirality 1992; 4:84-90.
    195) Ware TL, McCloskey K, Ryan MM, et al: Venlafaxine ingestion in a 4-year-old girl. J Paediatr Child Health 2012; 48(11):1047-1048.
    196) Weiner LA, Smythe M, & Cisek J: Serotonin syndrome secondary to phenelzine- venlafaxine interaction. Pharmacother 1998; 18:399-403.
    197) Weiss NT, Jones L, & Chamberlain JC: A possible case of venlafaxine-induced Stevens-Johnson syndrome. J Clin Psychiatry 2004; 65(10):1431-1433.
    198) Wheless JW : Treatment of status epilepticus in children. Pediatr Ann 2004; 33(6):376-383.
    199) White CM, Gailey RA, & Levin GM: Seizure resulting from a venlafaxine overdose. Ann Pharmacother 1997; 31:178-180.
    200) Wilson AD, Howell C, & Waring WS: Venlafaxine ingestion is associated with rhabdomyolysis in adults: a case series. J Toxicol Sci 2007; 32(1):97-101.
    201) Wilson R & Jenkins P: Suspected complication of treatment with venlafaxine (letter). J Clin Psychopharmacol 1997; 17:323.
    202) Woo OF, Vredenburg M, & Freitas P: Seizures after venlafaxine overdose; a case report (abstract). J Toxicol Clin Toxicol 1995; 33:549.
    203) Yuan TH, Kerns WP, & Tomaszewski CA: Insulin-glucose as adjunctive therapy for severe calcium channel antagonist poisoning. Clin Toxicol 1999; 37:463-474.
    204) Zhalkovsky B, Walker D, & Bourgeois JA: Seizure activity and enzyme elevations after venlafaxine overdose (letter). J Clin Psychopharmacol 1997; 17:490-491.
    205) dan-Manes J, Novalbos J, Lopez-Rodriguez R, et al: Lithium and venlafaxine interaction: a case of serotonin syndrome. J Clin Pharm Ther 2006; 31(4):397-400.
    206) 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.
    207) deGuzman MH, Thiagalingam S, Ong PY, et al: Bilateral acute angle closure caused by supraciliary effusions associated with venlafaxine intake. Med J Aust 2005; 182(3):121-123.