MOBILE VIEW  | 

VALPROIC ACID

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Valproic acid is a branched-chain carboxylic acid that has antiepileptic activity against a variety of seizure types and a low incidence of sedation and other central nervous system effects in therapeutic doses. Valproic acid dissociates in the gastrointestinal tract to the valproate ion. Valproate is available in the United States as divalproex sodium and valproic acid.

Specific Substances

    A) VALPROIC ACID
    1) 2-Propylpentanoic acid
    2) 2-Propylvaleric acid
    3) Abbott 44089
    4) Acetic acid, dipropyl-
    5) Acido valproico (Spanish)
    6) Depakene(R) (Abbott)
    7) Depakine
    8) Dipropylacetic acid
    9) Di-n-propylacetic acid
    10) n-Dipropylacetic acid
    11) Di-n-propylessigsaure (German)
    12) n-DPA
    13) Epilim
    14) Kyselina 2-propylpentanoic acid
    15) 2-Propylpentanioc acid
    16) 2-Propylvaleric acid
    17) Valproate
    18) Valpromide
    19) Valproato (Spanish)
    RELATED COMPOUND -- SODIUM VALPROATE
    1) Abbott 44090
    2) Depakene
    3) Depakine
    4) Depakane
    5) Depakote(R) (Abbott)
    6) Dipropylacetate sodium
    7) Divalproex sodium (Depakote(R))
    8) DPA sodium
    9) Epilim
    10) Ergenyl
    11) Eurekene
    12) KW-066
    13) Labazene
    14) Pentanoic acid, 2-propyl-, sodium salt
    15) 2-propylpentanoic acid sodium salt
    16) 2-Propylvaleric acid sodium salt
    17) Sodium bispropylacetate
    18) Sodium Dipropylacetate
    19) Sodium alpha, alpha-dipropylacetate
    20) Sodium n-dipropylacetate
    21) Sodium 2-propylpentanoate
    22) Sodium 2-propylvalerate
    23) Sodium valproate
    24) Valproic acid, sodium salt

Available Forms Sources

    A) FORMS
    1) Valproic acid is available in the following formulations:
    a) Liquid filled soft gelatin 250 mg capsules (orange-colored) in bottles of 100 capsules; oral syrup, 250 mg/5 mL (Prod Info DEPAKENE(R) oral capsules, oral solution, 2007)
    b) Oral capsule, delayed release: 125, 250, 500 mg (Prod Info STAVZOR(R) delayed release oral capsules, 2008)
    B) USES
    1) Valproic acid is approved as monotherapy or adjunctive therapy for the prophylactic treatment of complex partial seizures occurring alone or in association with other seizure types (Prod Info DEPAKENE(R) oral capsules, oral solution, 2007). It is also used for treatment of mania associated with bipolar affective disorder, and for prophylaxis of migraine headaches. An unapproved use of valproic acid includes treatment of neuropathic pain (Chappell et al, 1999).
    2) Recent studies have shown that valproic acid may be associated with anti-cancer activity, suppressing not only tumor growth and metastasis, but also inducing tumor differentiation in vitro and in vivo. These findings open novel aspects for the treatment of tumor patients (Blaheta & Cinatl, 2002; Blaheta & Cinatl, 2002).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Anticonvulsant for a broad spectrum of seizure disorders. Also used as a mood stabilizer in the treatment of bipolar affective disorder, used to treat chronic pain and as prophylaxis for migraine headaches.
    B) PHARMACOLOGY: Inhibits voltage-gated sodium channels (membrane stabilizing effects). Inhibits T-type calcium channels (excessive conductance causes seizures). Competitive antagonist of N-methyl-D-aspartic acid (NMDA) {excitatory} neuroreceptor. Inhibits GABA transaminase, increasing GABA (inhibitory neurotransmitter) concentrations in the brain.
    C) TOXICOLOGY: Causes CNS depression in overdose by extension of the pharmacologic mechanisms. Valproic acid depletes hepatic carnitine stores by forming valproylcarnitine, which inhibits the carnitine transported on the plasma membrane. Fatty acids cannot be metabolized due to lack of carnitine, resulting in chronic fatty liver. Valproic acid also depletes coenzyme A (CoA) stores in the liver by trapping CoA in the mitochondria by the valproic acid beta-oxidation metabolites. Depletion of CoA affects the activation of the carbamyl phosphate synthetase I (CPS I), which is needed for incorporating ammonia into the urea cycle, leading to hyperammonemia.
    D) EPIDEMIOLOGY: Poisoning is common with moderate severity.
    E) WITH THERAPEUTIC USE
    1) ADVERSE EFFECTS: COMMON: Anorexia, nausea, alopecia, peripheral edema, rash, sedation, weight gain, and teratogenicity. IDIOSYNCRATIC: Pancreatitis, hepatotoxicity, thrombocytopenia, hyperammonemia, and encephalopathy.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Primary effect is CNS depression, generally lethargy and sedation, vomiting and tachycardia.
    2) SEVERE TOXICITY: Patients typically develop more severe CNS depression, coma, miotic pupils, tachycardia, hypotension, QTc prolongation, and respiratory depression following severe poisoning. Seizures and cerebral edema are less common. Laboratory abnormalities may include hypernatremia, hypocalcemia and hyperammonemia. Bone marrow suppression may develop 3 to 5 days after massive overdose, and usually resolves spontaneously. Pancreatitis, acute hepatotoxicity, renal insufficiency, and acute lung injury are uncommon, but have been reported. Valproate-induced hyperammonemic encephalopathy can develop after overdose or therapeutic use. It is characterized by deterioration of mental status (i.e., lethargy, confusion, coma) AND an elevated ammonia concentration; it may also be associated with hepatotoxicity.
    0.2.3) VITAL SIGNS
    A) WITH THERAPEUTIC USE
    1) Hypothermia has been reported with therapeutic valproic acid use.
    B) WITH POISONING/EXPOSURE
    1) Both fever and hypothermia have been reported following valproic acid overdose. Hypotension may occur following severe overdose.
    0.2.20) REPRODUCTIVE
    A) Valproic acid is a known teratogen to humans. Maternal use of valproic acid alone or in combination with other antiepileptic drugs has been found to cause valproic acid syndrome in infants. Facial dysmorphology, congenital heart defects, spina bifida, cleft lip and palate, and developmental delays are some of the teratogenic effects seen.
    B) In 2009, the FDA notified healthcare professionals and patients about an increased risk of congenital malformations (ie, neural tube defects, craniofacial defects, cardiovascular malformations, and other body system malformations) in infants born of women exposed to valproate sodium and related products during pregnancy.
    C) The FDA has classified valproic acid as pregnancy category X and its use in pregnant women is contraindicated for the prevention of migraine headaches. However, valproate products continue to have a pregnancy category of D for treatment of epilepsy and manic episodes associated with bipolar disorder. Valproate products should only be used in pregnant women with epilepsy or bipolar disorder if other medications are not effective or are otherwise unacceptable in treating the condition.
    D) Valproic acid is excreted in breast milk with low concentrations and is not contraindicated during breastfeeding.
    0.2.21) CARCINOGENICITY
    A) At the time of this review, no data were found as to the carcinogenic effects of valproic acid in humans.

Laboratory Monitoring

    A) Valproate concentration should be measured (therapeutic level 50 to 100 mcg/mL) and repeated every 4 to 6 hours until downward trend is established.
    B) Serum concentrations greater than 450 mcg/mL is associated with drowsiness/obtundation; greater than 850 mcg/mL is associated with coma.
    C) Monitor blood gases, liver enzymes, ammonia concentration, lactic acid, electrolytes, blood sugar, renal function and ECG in patients with significant toxicity.
    D) Valproate causes false elevation of urine ketones.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) DECONTAMINATION
    1) PREHOSPITAL: Ipecac-induced emesis is not recommended in the prehospital setting because of the potential for aspiration. Activated charcoal may be considered in the prehospital setting by a healthcare professional only, if the patient is asymptomatic, a recent ingestion, and there are no contraindications. However, transportation to an emergency center should NOT be delayed to administer activated charcoal.
    2) HOSPITAL: Administer activated charcoal if the patient presents early after a significant ingestion if the patient is awake and can protect their airway, or the airway is protected. Administer a second dose of charcoal in patients who have ingested a sustained release formulation, or in patients with rising serum concentrations despite therapy. MULTIDOSE ACTIVATED CHARCOAL: Consider if valproic acid level is continuously rising and gastrointestinal function is intact (Note: patients with severe toxicity often have depressed gastrointestinal motility and may not tolerate multiple dose charcoal).
    B) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Symptomatic and supportive care in all patients. Monitor for progression of sedation. Repeat valproic acid levels every 4 to 6 hours and consider multidose activated charcoal if the level is continuously rising.
    C) MANAGEMENT OF SEVERE TOXICITY
    1) Resuscitation, symptomatic and supportive care in all patients. Early intubation in patient with declining level of consciousness. Hypotension: Treat with IV fluids, if no response start vasopressors. Consider hemodialysis in patients with severe toxicity who are not responding to supportive care. Consider carnitine in patients with coma, hepatotoxicity, elevated ammonia concentration, or a valproate concentration above 450 mcg/mL.
    D) AIRWAY MANAGEMENT
    1) Endotracheal intubation should be considered early if the patient presents with altered level of consciousness, hemodynamic instability, or multiorgan toxicity.
    E) ANTIDOTE
    1) NALOXONE has been used to reverse valproic acid induced CNS depression and coma, with variable success. GENERAL: Risk of naloxone administration is low; it is reasonable to administer in patients with significant CNS depression.
    2) L-CARNITINE: Orphan drug approved by FDA for the treatment of L-carnitine deficiency secondary to valproic acid toxicity. Appears effective in increasing survival in patients with valproate-induced hepatotoxicity and encephalopathy after therapeutic administration. INDICATION: Patients with coma, hepatotoxicity, hyperammonemia, or serum valproate concentration greater than 450 mcg/mL. While there are no controlled studies to support its use after acute overdose, there are several reports suggesting it lowers serum ammonia concentrations, and it is associated with few adverse effects. DOSE: IV 100 mg/kg over 30 minutes (maximum: 6 g) followed by 15 mg/kg IV every 4 hours until clinical improvement.
    F) ENHANCED ELIMINATION PROCEDURE
    1) Hemodialysis and hemoperfusion are reserved for severe toxicity with failure of improvement or deterioration despite supportive management, especially with concomitant severe metabolic disturbance, and/or serum valproic acid concentration of greater than 1000 mg/L. Because protein binding is saturated at a high serum concentration (resulting in higher free valproate concentrations), hemodialysis appears to be useful with severe overdose.
    G) PATIENT DISPOSITION
    1) HOME CRITERIA: Asymptomatic patients with unintentional ingestion of less than 50 mg/kg can be observed at home.
    2) OBSERVATION CRITERIA: Symptomatic patients, those with deliberate ingestions, and those with unintentional ingestions of 50 mg/kg or more should be referred to a medical facility for evaluation and treatment. Obtain serial valproate concentrations every 2 to 3 hours. Patients should be observed until valproate concentrations are clearly declining on at least two sequential measurements, and symptoms have resolved. IMMEDIATE RELEASE: Patients should be observed for a minimum of 6 hours after immediate release valproate ingestions. EXTENDED RELEASE: Monitor patients for a minimum of 12 hours following extended release preparations and should be admitted if symptoms develop.
    3) ADMISSION CRITERIA: Patients with rising valproate concentrations and those developing CNS depression or other clinical or laboratory evidence of toxicity should be admitted. Patients with persistent altered mental status, abnormal vital signs, acidosis, renal or hepatic involvement should be admitted to an intensive care setting.
    4) CONSULT CRITERIA: Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity, or in whom the diagnosis is not clear.
    H) PITFALLS
    1) Inadequate observation for respiratory depression.
    2) Insufficient duration of monitoring, particularly after ingestion of an extended release formulation. After a large ingestion, absorption can be slow and erratic. Coma may be delayed for up to 12 hours.
    3) Failure to repeat serial valproic acid concentrations until it is declining. Initial valproic acid concentration can be within therapeutic range even after a large ingestion.
    I) PHARMACOKINETICS
    1) Valproic acid is well absorbed (almost 100%). IMMEDIATE RELEASE: Peak serum concentration occurs 6 hours after immediate release. The Tmax for valproate is 4 hours following oral dosing. Highly protein bound (90%); volume of distribution is 0.1 to 0.2 L/kg; hepatic metabolism; and elimination half-life is 6 to 18 hours. EXTENDED RELEASE: The median time to maximum plasma valproate concentration (Cmax) after an extended release product can range from 4 to 17 hours. Peak serum concentration can be up to 24 hours after an extended-release product. The mean terminal half-life ranges from 9 to 16 hours following an oral dose regimen of 250 to 1000 mg. Hepatic metabolism. INTRAVENOUS: The Tmax for IV valproate sodium (sodium salt of valproic acid) is reached at the end of a 1 hour infusion.
    2) Therapeutic serum concentration: between 50 and 100 mg/L (350 to 700 micromol/L).
    J) TOXICOKINETICS
    1) Absorption may be slow and erratic after overdose, particularly for an extended release formulation. Protein binding is saturated in overdose (higher proportion of unbound valproate); protein binding can decrease as lows as 15% as serum concentrations rise to greater than 1000 mg/L (likely leading to greater clinical toxicity). Active metabolites contribute to toxicity. Prolonged elimination half-life in overdose (30 hours). Peak plasma concentration was delayed for 17 hours after ingestion of divalproex sodium and chlorpheniramine in overdose.
    2) Serum concentrations of greater than 450 mg/L (3125 micromol/L) are likely to result in moderate or major adverse clinical effects; concentrations of greater than 850 mg/L (5900 micromol/L) are likely to result in coma and metabolic acidosis.
    K) DIFFERENTIAL DIAGNOSIS
    1) The following should be considered: other anticonvulsants, opioid overdose, sedative hypnotics, alcohol ingestion, other drug-induced hepatitis, or herbal hepatitis.

Range Of Toxicity

    A) TOXICITY: MILD: Greater than 200 mg/kg risk of CNS depression; MODERATE: Greater than 400 mg/kg risk of multiogran system toxicities; and SEVERE: Greater than 750 mg/kg potentially lethal.
    B) THERAPEUTIC DOSE: ADULTS and CHILDREN 10 years of age and older: Initial dose: 10 to 15 mg/kg/day, titrate as needed; maximum dose: 60 mg/kg/day.

Summary Of Exposure

    A) USES: Anticonvulsant for a broad spectrum of seizure disorders. Also used as a mood stabilizer in the treatment of bipolar affective disorder, used to treat chronic pain and as prophylaxis for migraine headaches.
    B) PHARMACOLOGY: Inhibits voltage-gated sodium channels (membrane stabilizing effects). Inhibits T-type calcium channels (excessive conductance causes seizures). Competitive antagonist of N-methyl-D-aspartic acid (NMDA) {excitatory} neuroreceptor. Inhibits GABA transaminase, increasing GABA (inhibitory neurotransmitter) concentrations in the brain.
    C) TOXICOLOGY: Causes CNS depression in overdose by extension of the pharmacologic mechanisms. Valproic acid depletes hepatic carnitine stores by forming valproylcarnitine, which inhibits the carnitine transported on the plasma membrane. Fatty acids cannot be metabolized due to lack of carnitine, resulting in chronic fatty liver. Valproic acid also depletes coenzyme A (CoA) stores in the liver by trapping CoA in the mitochondria by the valproic acid beta-oxidation metabolites. Depletion of CoA affects the activation of the carbamyl phosphate synthetase I (CPS I), which is needed for incorporating ammonia into the urea cycle, leading to hyperammonemia.
    D) EPIDEMIOLOGY: Poisoning is common with moderate severity.
    E) WITH THERAPEUTIC USE
    1) ADVERSE EFFECTS: COMMON: Anorexia, nausea, alopecia, peripheral edema, rash, sedation, weight gain, and teratogenicity. IDIOSYNCRATIC: Pancreatitis, hepatotoxicity, thrombocytopenia, hyperammonemia, and encephalopathy.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Primary effect is CNS depression, generally lethargy and sedation, vomiting and tachycardia.
    2) SEVERE TOXICITY: Patients typically develop more severe CNS depression, coma, miotic pupils, tachycardia, hypotension, QTc prolongation, and respiratory depression following severe poisoning. Seizures and cerebral edema are less common. Laboratory abnormalities may include hypernatremia, hypocalcemia and hyperammonemia. Bone marrow suppression may develop 3 to 5 days after massive overdose, and usually resolves spontaneously. Pancreatitis, acute hepatotoxicity, renal insufficiency, and acute lung injury are uncommon, but have been reported. Valproate-induced hyperammonemic encephalopathy can develop after overdose or therapeutic use. It is characterized by deterioration of mental status (i.e., lethargy, confusion, coma) AND an elevated ammonia concentration; it may also be associated with hepatotoxicity.

Vital Signs

    3.3.1) SUMMARY
    A) WITH THERAPEUTIC USE
    1) Hypothermia has been reported with therapeutic valproic acid use.
    B) WITH POISONING/EXPOSURE
    1) Both fever and hypothermia have been reported following valproic acid overdose. Hypotension may occur following severe overdose.
    3.3.3) TEMPERATURE
    A) HYPOTHERMIA
    1) WITH POISONING/EXPOSURE
    a) Hypothermia has been reported in epileptic adult patients following acute overdoses of sodium valproate (Connacher et al, 1987; Hintze et al, 1987).
    b) CASE REPORT: A 22-year-old man developed a core rectal temperature of 27 degrees Celsius following an overdose of sodium valproate. The patient required aggressive rewarming including external rewarming therapy and invasive rewarming with warmed bladder lavage. The patient fully recovered with no neurologic deficit by day 2. Although the authors concluded that the etiology of hypothermia was likely multifactorial, sodium valproate was thought to significantly contributed to the development of severe hypothermia in this case (Robinson & Abbott, 2005).
    2) WITH THERAPEUTIC USE
    a) Hypothermia has been reported in children following short-term therapeutic dosing (Nagarajan et al, 2001).
    B) WITH POISONING/EXPOSURE
    1) FEVER
    a) Fever has been reported following valproic acid overdose (Bigler, 1985; Hintze et al, 1987).
    3.3.4) BLOOD PRESSURE
    A) WITH POISONING/EXPOSURE
    1) HYPOTENSION
    a) Hypotension has been reported with severe overdoses (Tank & Palmer, 1993; Farrar et al, 1993) Anderson & Ritland, 1995; (Williams & Clark, 1995; Kuperschmidt et al, 1998).

Heent

    3.4.3) EYES
    A) WITH POISONING/EXPOSURE
    1) MIOSIS
    a) Miosis has been reported following acute overdose (Steiman et al, 1979; Alberto et al, 1989; Hicks & McFarlane, 2001).
    2) OPTIC NERVE ATROPHY
    a) Optic nerve atrophy has been reported following an acute overdose (Bigler, 1985).
    b) CASE REPORT: Neurologic sequelae were reported in a 24-year-old man following an ingestion of more than 100 grams of sodium valproate who presented late to the emergency department (18 hours postingestion).
    1) The patient was comatose and febrile for 10 days despite antibiotic therapy. After 2 months there was severe reduction of vision due to optic nerve atrophy (Bigler, 1985).
    3) NYSTAGMUS
    a) A 16-year-old boy was reported to have reactive pupils of 4 mm and horizontal nystagmus following an overdose of 38 delayed-release tablets of 250 mg each (Wasserman et al, 2001).
    4) SALIVARY GLANDS
    a) THERAPEUTIC USE: A 26-year-old woman developed bilateral sialadenosis (benign, noninflammatory enlargement) of the parotid glands following 4 years of valproic acid therapy for seizures. Enlargement persisted despite discontinuation of valproic acid, eventually requiring surgical excision (Mauz et al, 2005).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) CARDIAC ARREST
    1) WITH POISONING/EXPOSURE
    a) Cardiorespiratory arrest has been reported following the ingestion of an unknown amount of sodium valproate (plasma valproate concentration 13,672 mcmol/L) (Gourru, 1981).
    b) CASE REPORT: Cardiorespiratory arrest was reported in a 20-month-old boy who unintentionally ingested 15 g of sodium valproate. Peak serum concentration was reported to be 1061 micrograms/milliliter within three hours of the time of ingestion (Janssen et al, 1985). Death was thought to be caused by severe bronchial pneumonia.
    B) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Hypotension, often refractory to fluid resuscitation and vasopressor therapy, has been reported with severe overdoses (Singh et al, 2004; Tank & Palmer, 1993; Farrar et al, 1993; Andersen & Ritland, 1995; Williams & Clark, 1995; Kuperschmidt et al, 1998; Johnson et al, 1999; Christianson et al, 2001).
    b) INCIDENCE: In a series of 133 patients with valproate only overdose reported to a group of poison centers, 4 (3%) developed hypotension (Spiller et al, 2000).
    c) CASE REPORT: A 40-year-old epileptic woman became hypotensive (BP 60/40 mmHg) following an overdose of sodium valproate. The hypotension was difficult to correct despite infusion of intravenous fluids, careful central venous pressure monitoring, and administration of dopamine and dobutamine. Valproate concentration shortly after onset of hypotension was 17,200 micromoles/liter (Connacher et al, 1987).
    C) PROLONGED QT INTERVAL
    1) WITH POISONING/EXPOSURE
    a) Significant QT interval prolongation appears to be a common effect of acute valproic acid (VPA) poisoning. Kupferschmidt et al (1999) retrospectively examined 16 cases of acute valproic acid poisonings, with no other coingestants. Twelve of the 16 patients were reported to have significantly longer QTc during the acute stage than at baseline. Nine of the 16 had acute QT values greater than 450 msec (6 cases greater than 500 msec). The authors suggested that VPA-related QT prolongation may be due to diminished potassium currents in cardiomyocytes.
    b) CASE REPORT: Following ingestion of an unknown amount of valproic acid (VPA), a 29-year-old woman was admitted 30 hours later to the ED. She was unresponsive, hypotensive (systolic BP 70 mmHg), and an ECG revealed peaked T-waves with a prolonged QTc interval of 593 msec. The plasma VPA concentration at 36 hours postingestion was 660 mg/L and the patient also had hypocalcemia, hyperkalemia and acute renal insufficiency. QTc normalized within 56 hours. The patient was discharged on day 18 with only mild neurological deficits (Kuperschmidt et al, 1998).
    D) TACHYARRHYTHMIA
    1) WITH POISONING/EXPOSURE
    a) Tachycardia has been reported following an overdose (Meyer et al, 2005; Camilleri et al, 2005; Ibister et al, 2003; Spiller et al, 2000).
    1) INCIDENCE: In a series of 133 patients with valproate only overdose reported to a group of poison centers, 24 (17%) developed tachycardia (Spiller et al, 2000).
    2) In a small case series of 15 patients with valproate only overdose, 5 patients developed tachycardia. There were no reports of dysrhythmias or hypotension (Ibister et al, 2003).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) RESPIRATORY FAILURE
    1) WITH POISONING/EXPOSURE
    a) Respiratory arrest requiring artificial ventilation is common in patients with severe overdose (Eyer et al, 2005; Minari et al, 2002; Christianson et al, 2001; Williams & Clark, 1995; Graudins & Aaron, 1996; Andersen & Ritland, 1995; Gourru, 1981; Fuller et al, 1981; p 87) .
    b) CASE REPORT: Apneic episodes developed in a 26 month old after ingesting 4.5 grams. Naloxone appeared to reverse the apnea (Farrar et al, 1993).
    B) PULMONARY ASPIRATION
    1) WITH POISONING/EXPOSURE
    a) INCIDENCE: In a series of 133 patients with valproate only overdose reported to a group of poison centers, 8 (6%) developed aspiration pneumonitis (Spiller et al, 2000).
    C) PNEUMONIA
    1) WITH THERAPEUTIC USE
    a) Apnea, pulmonary hemorrhage, and bronchopneumonia have been associated with chronic valproate administration (Gerber et al, 1979). A case of thrombocytopenia-induced fatal pulmonary hemorrhage was reported in a 30-year-old woman on chronic valproate monotherapy, with a history of a viral illness 3 weeks earlier (Sleiman et al, 2000). Initial serum valproate concentration was 124 micrograms/mL (normal 50 to 100 micrograms/mL). The authors suggested that viral infections may be associated with thrombocytopenia in patients on valproate therapy.

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) CENTRAL NERVOUS SYSTEM DEFICIT
    1) WITH POISONING/EXPOSURE
    a) Confusion, somnolence, acute toxic encephalopathy with behavior disturbances, worsened seizure control, and coma have been associated with ingestions of overdose amounts of sodium valproate. When ingestions of sustained-release or enteric-coated divalproex sodium are taken, delayed toxicity may occur, with CNS depression occurring as long as 8 to 13 hours postingestion (Weiner et al, 2013; Steiman et al, 1979; Simon & Penry, 1975; Eeg-Olofsson & Linkskog, 1982; Fuller et al, 1981; Gourru, 1981; Garnier & Fournier, 1982; p 87; Chadwick et al, 1979; Andersen & Ritland, 1995; Graudins & Aaron, 1996; Lee et al, 1998; Johnson et al, 1999; Montero, 1999; Brubacher et al, 1999; Hicks & McFarlane, 2001; Thole et al, 2001; Kroll & Nand, 2002; Minari et al, 2002).
    b) One reference states coma occurs at doses of about 20 mg/kg or more (Garnier & Fournier, 1982).
    c) CASE REPORT: A 41-year-old epileptic man was admitted comatose with an initial valproic plasma concentration of 1308 mg/L (therapeutic range: 50-150 mg/L) (estimated ingestion was more than 100 tablets of 1 g of valproic acid). Laboratory studies included hyperammonemia, elevated liver enzymes, and an elevated creatinine kinase (10,000 U/L). The patient was treated with activated charcoal and L-carnitine and the patient awoke 36 hours after admission. The patient fully recovered and was discharged to home on hospital day 17 (Sikma et al, 2008).
    d) INCIDENCE: In a series of 133 patients with valproate only overdose reported to a group of poison centers, 94 (71%) developed lethargy and 19 (15%) developed coma (Spiller et al, 2000). Coma was more common in patients with peak valproate levels of more than 850 mcg/ml.
    e) ELECTROENCEPHALOGRAM/CASE REPORT: A 20-year-old woman ingested approximately 75 grams of sodium valproate and had a serum concentration of 1500 mcg/mL 3 hours postingestion. The EEG revealed decreased dominant frequency and some theta and delta activity.
    1) At 8 hours the level was 1800 mcg/mL and the EEG was dominated by delta activity of low amplitude. Following hemodialysis and hemoperfusion, levels decreased to 100 mcg/mL (36 hours postingestion) and the EEG had slight activity with low voltage. Four days after the overdose the valproate level was in the normal range (40 to 100 mcg/mL) and the EEG was dominated by delta activity of high amplitude, especially in the frontal lobe. At 6 days the EEG was normal (Pedersen & Juul-Jensen, 1984).
    f) CHRONIC EXPOSURE: A 7-year-old girl, with severe cerebral palsy and severe developmental delay and chronic epilepsy that was treated with valproic acid, developed further development deterioration (ie, more sleepy, poor feeding and decreased responsiveness) over a period of a few weeks. Upon admission, an arterial blood gas showed compensated respiratory acidosis. Thrombocytopenia was also present. Her valproic acid concentration was 700 mcg/mL (therapeutic range, 50 to 100 mcg/mL). Two months prior to admission the patient's valproic acid had been changed from 250 mg/5 mL to a concentration of 200 mg/mL. The patient continued to receive the same volume resulting in a dosage that was approximately 4 times her usual dose. Valproic acid was stopped and the patient gradually became more alert; valproic acid and laboratory concentrations also normalized (Weiner et al, 2013).
    g) VALPROMIDE: Delayed CNS depression and coma have been reported after overdose of valpromide in several patients. Three patients who presented after deliberate overdose were initially asymptomatic with therapeutic concentrations. Two were transferred to a psychiatric facility and developed mental status depression progressing to coma 20 and 44 hours after ingestion associated with valproic acid concentrations of 295 and 256 mg/L (peak serum levels not determined). A third patient developed dyspnea after 10 hours of observation, and progressed to metabolic acidosis, respiratory failure and encephalopathy with a serum valproate concentration of 200 mg/L. All patients recovered (Payen et al, 2004).
    B) TOXIC ENCEPHALOPATHY
    1) WITH THERAPEUTIC USE
    a) Severe encephalopathy associated with hyperammonemia has been reported following chronic therapy, and as a synergistic effect of other anticonvulsants (phenobarbital, phenytoin, and topiramate) administered with valproic acid (Lheureux et al, 2005; Borbath et al, 2000; Hamer et al, 2000).
    b) CASE REPORTS
    1) PEDIATRIC: A 9-year-old boy diagnosed with intermittent explosive disorder was admitted to the hospital for aggressive behavior after being treated fro 7 months with valproate (100 mg daily) and quetiapine. He had a therapeutic valproate serum concentration (113 mcg/mL) and normal liver enzymes, but an elevated serum ammonia concentration (127 mcg/dL) with EEG patterns consistent with diffuse encephalopathy. Symptoms were controlled with chlorpromazine, diazepam, lithium carbonate, and discontinuation of valproate (Yehya et al, 2004).
    2) ADULT: A 50-year-old woman taking carbamazepine 1200 mg daily for partial epilepsy developed acute confusion after valproate was added, initially in a 1000 mg per day dose, which was increased to 1500 mg after three days. A mildly elevated serum ammonia concentration (81 microg/dL) was noted. Her electroencephalogram indicated encephalopathy with diffuse background slowing mixed with high-amplitude slow waves. Serum concentrations of valproic acid and carbamazepine were 49.1 mg/L and 8.6 mg/L, respectively. Both concentrations were nontoxic. The patient improved rapidly after the valproate was discontinued (Chen et al, 2001). Although a safe previous administration of valproic acid did not preclude the occurrence of encephalopathy in this patient. Its suggested that a relatively large initial valproic acid dose could possibly have caused the symptoms observed in this patient.
    2) WITH POISONING/EXPOSURE
    a) Severe encephalopathy associated with hyperammonemia has been reported following acute overdoses (Unal et al, 2007; Borbath et al, 2000; Hamer et al, 2000).
    b) CASE REPORT: A 9-year-old, 50 kg, girl intentionally ingested 196 mg/kg of valproic acid and upon admission 4 hours after ingestion she was stuporous and required immediate intubation and ventilation. Serum valproic acid level was 599.2 mcg/mL (reference range: 50 to 100 mcg/mL). Hyperammonemia (serum ammonia: 111 mcg/dL) was observed and treated with L-carnitine. Hemoperfusion was started within 8 hours of exposure that was repeated on day 2 for 3 hours; serum valproic level rapidly improved with therapy. The patient was also treated with fresh frozen plasma for an increased prothrombin and activated partial thromboplastin time. The child gradually improved and was extubated on day 4. She was discharged to home on day 9 with no permanent sequelae (Colak et al, 2011).
    c) CASE REPORT/INFANT: A 26-day-old full term infant (4500 g) was unintentionally given 300 mg valproate (intended dose 30 mg/kg twice daily); symptoms began within 12 hours. Within 36 hours, the pupils were fixed and dilated. A CT scan of the head showed cerebral edema, and a serum ammonia of 279 mcg/dL (normal 15 to 56 mcg/dL) was also present. Despite aggressive care, the patient died of a cardiorespiratory arrest secondary to severe brain edema 42 hours after exposure (Unal et al, 2007).
    C) CEREBRAL EDEMA
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Cerebral edema and coma have been reported in fatal cases; the etiology is unclear (Unal et al, 2007; Camilleri et al, 2005; Eyer et al, 2005; Poklis et al, 1998), but may be due to a direct toxic effect of VPA or its metabolites and hyperammonemia, increasing intracellular osmolarity, promoting an influx of water, cerebral edema, and increased intracranial pressure (Camilleri et al, 2005; Thabet et al, 2000; Eyer et al, 2005).
    b) CASE REPORTS
    1) PEDIATRIC
    a) INFANT: A 26-day-old full term infant (4500 g) was unintentionally given 300 mg valproate (intended dose 30 mg/kg twice daily); symptoms began within 12 hours. Within 36 hours, the pupils were fixed and dilated. A CT scan of the head showed cerebral edema, and serum ammonia of 279 mcg/dL (normal 15 to 56 mcg/dL) was also present. Despite aggressive care, the patient died of cardiorespiratory arrest secondary to severe brain edema 42 hours after exposure (Unal et al, 2007).
    b) TODDLER: Cerebral edema occurred in a previously healthy 26-month-old girl who ingested 625 mg/kg of valproic acid. Evidence of elevated pressure was observed 48 hours after ingestion. Resolution occurred by day 5 following placement of ventriculostomy, hyperventilation to maintain cerebral perfusion pressure at 60 to 70 mmHg, and administration of mannitol 25 g (one dose) and dopamine 1 to 8 mcg/kg/minute (Dupuis et al, 1990).
    c) TEENAGER: Following an intentional overdose of 20 grams VPA (330 mg/kg), a 15-year-old girl was admitted with coma, metabolic acidosis, hyperammonemia, and hyperlactacidemia. Valproate serum concentration was reported to be 1550 mg/L at hour 10. The patient died 4 days after ingestion due to cerebral edema with irreversible coma (Thabet et al, 2000).
    2) ADULT
    a) CASE REPORT: A 29-year-old man with bipolar affective disorder sustained permanent injury when he developed cerebral edema, herniation, and infarct after intentionally ingesting 4 g of valproic acid (VPA) and 75 mg of diazepam (10 hours prior to ingesting VPA). He presented to the emergency department 5 hours after ingestion of VPA with agitation, an unsteady gait, and vomiting. At that time, his vital signs and neurologic examination were normal. Initial lab analyses showed a VPA level of 5469 micromol/L (therapeutic: 350 to 700 micromol/L), serum ammonia of 348 micromol/L (normal: 0 to 50 micromol/L), and a pH of 7.34. CT scan confirmed diffuse cerebral edema with early herniation. Nine hours after ingestion, his Glasgow coma scale score deteriorated to 8/15. He was intubated, gastric contents were aspirated, and he was treated with a single 50 g dose of activated charcoal followed by 4 doses of L-carnitine (900 mg each, 1 hour apart), and mannitol. Thirty-eight hours after ingestion, VPA levels were therapeutic. A dilated right pupil, absent right papillary reflex, reduced visual acuity with skew deviation of the right eye, and right-sided weakness were observed on day 5. At that point, MRI findings confirmed cerebral infarct and ECG findings were consistent with encephalopathy. Ammonia levels normalized on day 6 and on day 8 he was extubated. After 150 days of hospitalization including extensive rehabilitation he was discharged to an assisted living facility with residual right-sided weakness and a modified Rankin scale score of 3 (moderate disability; requiring some help, but able to walk without assistance) (Rupasinghe & Jasinarachchi, 2011; van Swieten et al, 1988).
    b) CASE REPORT: After ingesting 20 g (333 mg/kg) of valproic acid extended release, 60 mg of risperidone, and 3 g of venlafaxine extended release, a 19-year-old man developed hyperammonemia (peak ammonia concentration 1,191 mcg/dL at 65 hours postingestion). The valproic acid concentration peaked at 305.4 mcg/dL. Despite decontamination with activated charcoal with sorbitol and lactulose, the patient's condition deteriorated. The brain CT scan revealed cerebral edema and possible tentorial herniation. At approximately 120 hours after ingestion, cerebral blood flow studies showed an absence of cerebral perfusion, and the patient was pronounced brain dead (Camilleri et al, 2005).
    c) CASE REPORT: An adult presented with drowsiness 6 hours after ingesting 60 g of sodium valproate. By 9 hours postingestion, the patient became apneic and comatose. Peak serum valproate concentration was 1361 mcg/mL and peak ammonia concentration was 541 mcg/dL 10 hours postingestion. Cerebral edema was evident on CT scan by day 2. Multiorgan failure developed and the patient died 8 days after exposure (Eyer et al, 2005).
    d) CASE REPORT: A 29-year-old epileptic man developed massive cerebral edema following an acute overdose of sodium valproate. The diagnosis of cerebral edema was supported by computerized tomography (CT) and electroencephalography (EEG). A normal CT was reported 9 days later after successful treatment of cerebral edema with sodium thiopental, glycerol, and glucocorticoids (Hintze et al, 1987). It is uncertain whether the cerebral edema was the result of hypoxia or valproate intoxication.
    D) DISORDER OF THE PERIPHERAL NERVOUS SYSTEM
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 26-year-old man, with a history of bipolar disease, intentionally ingested an unknown amount of valproic acid and was admitted a few hours later with sopor without focal neurologic deficits. There were no signs of trauma and a CT of the brain was negative. Laboratory studies were normal. A valproic acid serum level was 2896 micromol/L (therapeutic range: 350 to 690 micromol/L). His cognitive function gradually improved over several days. The patient reported weakness in his right arm on day 3 and he had evidence of weakness of flexion and abduction of the right arm and loss of sensation in the skin over the lateral upper right arm. Radiographic and MR studies showed no evidence of fracture or neurologic dysfunction of the shoulder or cord or nerve root of the spine. Paresis of the right axillary nerve was detected on physical exam and electrodiagnostic studies. A physical therapy program resulted in gradual improvement (Marusic et al, 2014).
    E) SEIZURE
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A report of paradoxical increasing electrical seizures in a 25-year-old with a hypothalamic hamartoma is reported shortly after initiation with valproic acid therapy. A positive correlation was shown between the prevalence of spike and wave activity and serum valproate concentrations (Stecker & Kita, 1998).
    2) WITH POISONING/EXPOSURE
    a) Seizures may occur after intoxication (Chadwick et al, 1979).
    b) CASE REPORT: Seizures and myoclonic jerking developed in a 44-year-old woman after overdose with valproic acid and trazodone (Williams & Clark, 1995).
    c) CASE REPORT: Generalized seizures, progressive coma, and anuria in a 2-month-old girl were associated with serum sodium valproate concentration of 1299 mcg/mL (Roodhooft et al, 1990).
    F) DROWSY
    1) WITH POISONING/EXPOSURE
    a) In a series of 15 patients with valproate only overdose, 2 patients experienced drowsiness after ingesting 300 to 400 mg/kg. No other neurological deficits were reported (Ibister et al, 2003).
    G) NEUROLOGICAL FINDING
    1) WITH THERAPEUTIC USE
    a) Increased serum drug concentrations may cause unusual neurological effects. The following may be due to increased blood ammonia concentration.
    1) Chronic high doses of valproic acid (2.2 to 2.5 gm/daily) in two adults produced drowsiness, confusion, irritability, apathy, withdrawn behavior, and visual hallucinations (Chadwick et al, 1979).
    2) Drowsiness, twitching of limbs, deep coma, sedation, emotional disturbances, weakness, lethargy, ataxia, mental stimulation, excitement, aggressiveness, hyperactivity, headache, and tremor have been reported following chronic valproate administration (Lewis, 1978; Gerber et al, 1979) .
    H) ABNORMAL BEHAVIOR
    1) WITH THERAPEUTIC USE
    a) NORMAL BLOOD CONCENTRATIONS: Behavioral changes were seen in 56/88 pediatric patients receiving sodium valproate monotherapy.
    1) These included irritability, longer and deeper sleep, superficial sleep, hyperactivity, increased alertness, lassitude, drowsiness, increased sociability, calmness, increased sadness, happiness, and aggression.
    2) It was emphasized that stimulant-like reactions were as frequent as depressive effects (Herranz, 1982).
    I) MULTIPLE SYSTEM ATROPHY
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Valproic acid toxicity, mimicking multiple system atrophy, was reported in a 67-year-old woman taking the drug for a total of 8 years. Over the last 5 years she developed dysarthria, progressive action tremor, worsened bradykinesia, and ataxia. The patient was diagnosed with multiple system atrophy and finally was wheelchair bound, ataxic and incontinent. Three months after stopping valproic acid, symptoms began to improve. At three year follow-up, her neurologic examination was normal (Shill & Fife, 2000).
    J) EXTRAPYRAMIDAL DISEASE
    1) WITH THERAPEUTIC USE
    a) Parkinson's syndrome has been associated with chronic valproate therapy. In a series of 36 patients treated with valproate, 27 (75%) had clinical evidence of Parkinsonism (Armon et al, 1996). Of these patients 19 (53%) complained of instability, 16 (44%) reported tremor, 30 (80%) demonstrated cognitive impairment, 22 (62%) had bradykinesia, 20 (55%) had abulia and 16 (44%) had upper motor neuron signs (Armon et al, 1996). Most patients improved after valproate was discontinued.
    b) An extrapyramidal syndrome, unresponsive to antiparkinson medication (benztropine), developed in a 52-year-old man with schizophrenia who was given sodium valproate 1 to 2 g/d (Lautin, 1979).
    K) CEREBELLAR DISORDER
    1) WITH THERAPEUTIC USE
    a) CASE SERIES: In a series of 16 patients treated with valproate in whom cranial CTs were performed, 11 (69%) demonstrated new or progressive cerebral atrophy (Armon et al, 1996). The atrophy improved in the two patients in whom cranial CTs were repeated after valproate was discontinued.

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) PANCREATITIS
    1) WITH THERAPEUTIC USE
    a) Some cases of hemorrhagic pancreatitis with rapid progression from initial symptoms to death have been described. Onset of pancreatitis may be shortly after initiation of valproic acid therapy or following several years of use (Chapman et al, 2001; (Anon, 2000)). Patients on valproate polytherapy may be at a higher risk of developing pancreatitis (Yazdani et al, 2002).
    b) CASE SERIES: Fourteen cases of valproate-associated pancreatitis are reviewed. None of the cases experienced other valproate toxic effects and pancreatitis was not dose-related. It developed as early as one week and as late as 4.5 years after starting treatment. Two of 14 patients died. Of 7 rechallenged with VPA, 6 had recurrence of pancreatitis (Wyllie et al, 1984).
    c) Development of VPA-associated pancreatitis is a relative contraindication to further treatment. However, routine monitoring of serum amylase is not necessary in asymptomatic patients.
    d) Other cases of fatal and nonfatal valproate induced pancreatitis have been described (Evans et al, 1995; Pinkston & Walker, 1997).
    e) CASE REPORT: A 23-year-old man, on hemodialysis for endstage renal disease secondary to hemolytic uremic syndrome, developed pancreatitis following a 3 month course of valproic acid (2500 mg/day) and phenobarbital (200 mg/day). After stopping valproic acid and administering symptomatic therapy, the pancreatitis resolved. Phenobarbital was continued, with no further pancreatic symptoms (Plaza et al, 1999).
    f) CASE REPORT/SUDDEN DEATH: A 4.5 year old girl, with a history of epilepsy and receiving valproic acid (375 mg three times daily) and topiramate (25 mg twice daily) therapy for approximately four months, developed acute abdominal pain with uncontrollable vomiting and then sudden death (Mileusnic et al, 2002). Postmortem findings were consistent with hemorrhagic pancreatitis due to valproic acid therapy.
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: Hemorrhagic pancreatitis was found on postmortem examination of a 40-year-old epileptic woman following an overdose of sodium valproate (Connacher et al, 1987).
    b) CASE REPORT - Elevated amylase and lipase concentrations developed in a 27-year-old woman after severe valproate overdose (Andersen & Ritland, 1995).
    c) CASE REPORT: An adult developed pancreatitis and cerebral edema and eventually died after a large valproate overdose of unknown size. Autopsy revealed hemorrhagic necrotic pancreatitis (Eyer et al, 2005).
    B) NAUSEA AND VOMITING
    1) WITH THERAPEUTIC USE
    a) Vomiting as well as nausea, and abdominal pain have been reported (Egger & Brett, 1981; Lewis, 1978; Herranz, 1982).
    2) WITH POISONING/EXPOSURE
    a) Nausea and vomiting may occur and may be prolonged following ingestions of sustained-release products (Wasserman et al, 2001).
    b) In a series of 15 patients with valproate only overdose, 4 developed vomiting (Ibister et al, 2003).
    C) BEZOAR
    1) WITH POISONING/EXPOSURE
    a) Due to the enteric-coated formulation of valproic acid, large overdoses may potentially result in tablet bezoars (Graudins & Aaron, 1996).
    D) INCREASED APPETITE
    1) WITH THERAPEUTIC USE
    a) Anorexia and increased appetite as well as weight gain have been reported (Bruni & Wilder, 1979).
    E) DIARRHEA
    1) WITH THERAPEUTIC USE
    a) Diarrhea has been reported, as has transient enuresis (Gerber et al, 1979; Suchy et al, 1979).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) LIVER DAMAGE
    1) WITH POISONING/EXPOSURE
    a) Garnier reported 516 acute valproate overdoses, in which liver function was normal (Garnier et al, 1982). Risk factors for developing hepatotoxicity during therapeutic use include age under 2 years, previous liver disease, developmental delay, coincident metabolic disorders and severe seizures requiring multiple drugs or ketogenic diets for control (Lheureux et al, 2005). Valproic acid-induced hepatotoxicity is thought to be due to carnitine depletion, based on limited clinical evidence (Lheureux & Hantson, 2009).
    b) CASE REPORT: A 33-year-old man on combined valproic acid and phenytoin therapy for seizures developed severe AST elevations (over 100 times normal upper limit) (McLaughlin et al, 2000).
    c) CASE REPORT: Elevated aminotransferase concentrations developed in 2 women after valproate overdose (Andersen & Ritland, 1995).
    B) HYPERAMMONEMIA
    1) WITH THERAPEUTIC USE
    a) SUMMARY: Valproic acid-induced hyperammonemia may develop after an acute overdose or following chronic therapy (Lheureux & Hantson, 2009; Halaby et al, 2013).
    b) Severe encephalopathy associated with hyperammonemia has been reported following early therapeutic dosing and as a synergistic effect of other anticonvulsants (phenobarbital, phenytoin, and topiramate) with valproic acid (Lheureux et al, 2005; Raskind & El-Chaar, 2000; Borbath et al, 2000; Hamer et al, 2000; Ellaway et al, 1999). Hyperammonemia (plasma ammonia >80 mcg/dL) has also been reported in the psychiatric setting following initiation of valproic acid therapy (Eubanks et al, 2008; Panikkar & Gilman, 1999).
    c) CLINICAL SYNDROMES: Hyperammonemia in the setting of valproate use or overdose can be associated with any of the following clinical syndromes:(Halaby et al, 2013; Chan et al, 2007):
    1) hyperammonemia without significant CNS or liver toxicity;
    2) valproic acid-induced hyperammonemic encephalopathy (VHE);
    3) valproic acid-induced hepatotoxicity
    d) PREVALENCE: In prospective studies of valproate overdose, the prevalence of hyperammonemia ranges from 70% to 100%, although 50% of patients will not have associated CNS or hepatic toxicity (Lheureux & Hantson, 2009).
    e) CASE REPORTS: A 31-year-old man with a new onset of agitation and aggressive behavior was newly diagnosed with bipolar type I disorder and started on lithium, valproic acid and quetiapine. No improvement was noted after 3 weeks of treatment. Despite a normal valproic acid level, his ammonia plasma level was elevated (1.37 mg/L, reference range 0.1 and 05. mg). Hepatic function was normal. Valproic acid was stopped and confusion cleared within 24 hours along with a gradual decrease in his ammonia level over 48 hours (Halaby et al, 2013).
    1) In the second case, a 21-year-old man, with a history of bipolar disorder type II for 2 years, was admitted for medical detoxification secondary to opioid and cannabis abuse. He remained on valproic acid (500 mg twice daily) and was started on analgesics, tranquilizers and sedatives (ie, diazepam and quetiapine) for potential withdrawal symptoms. About 4 days after the start of therapy, he became confused and disoriented. His valproic acid level was only slightly elevated (55 mcg/mL (reference range, 50 to 100 mcg/mL). Other laboratory studies included normal hepatic function, a negative toxicology screen for illicit drugs, and an ammonia level of 4.8 mg/L (4 times the upper limit of normal). Two days after stopping valproic acid his mentation was normal and his ammonia level was within normal limits (Halaby et al, 2013). The possible increased risk of hyperammonemia due to the coadministration of valproic acid and quetiapine remains unclear.
    f) CASE REPORT: Ten days following initiation of valproic acid (10 mg/kg/day), a 51-year-old woman presented with a rapidly declining level of consciousness (Glasgow coma score 5/15). EEG showed triphasic waves consistent with hepatic encephalopathy. Serum valproic acid and liver enzyme concentrations were normal; blood arterial ammonia concentration was significantly increased (234 micromole/L) 10 hours after presentation. Following discontinuation of valproic acid and administration of levocarnitine, her neurological condition improved within 18 hours (Borbath et al, 2000).
    g) CASE REPORT: A 33-year-old woman with bipolar disorder was started on daily divalproex sodium 1500 mg daily for 2 days and was noted to be lethargic by her caregivers. Her initial ammonia concentration was 283 mcmol/L (reference range: 2-30), along with a valproate serum concentration of 120 mg/L (reference range: 50-100). The patient received an initial 3 g IV of carnitine followed by carnitine 990 mg three times daily and lactulose 30 cc every 6 hours. Mental status improved within several hours of therapy. The concentration dropped to 25 on hospital day 4, the patient was discharged with a valproate concentration of less than 10 (Eubanks et al, 2008).
    h) CASE REPORT: In another study, a 32-year-old man on daily valproate therapy alone developed hyperammonic encephalopathy (Ziyeh et al, 2002). Valproic acid serum concentrations were within normal limits (68 mg/L) on admission, and laboratory and sonographic studies were negative for hepatic disease. However, evaluation with MRI indicated a pattern that was compatible with a toxic-metabolic encephalopathy. In addition, MR spectroscopic findings were also consistent with hepatic encephalopathy based on severe depletion of myoinositol and choline along with glutamate excess. The exact mechanism for choline depletion has yet to be determined, but may be based on a disturbance of osmotic homeostasis at the cellular level.
    i) PEDIATRIC: A 9-year-old boy was treated over 7 months with valproate for intermittent explosive disorder. During hospital admission for aggressive behavior he had a serum concentration of 99 mcg/mL on a dose of 100 mg valproate daily. The patient was also treated with quetiapine. Over one week, the patient developed worsening irritability and aggression, despite addition of lithium carbonate. Valproate serum concentration was 113 mcg/mL with an elevated serum ammonia concentration of 127 mcg/dL. Liver enzymes were normal. An EEG showed a pattern consistent with diffuse encephalopathy. Serum valproate concentration dropped to 55 mcg/mL 4 days after cessation of medication. It is unclear if the withdrawal of valproate was effective, because the patient's behavioral problems required chlorpromazine and diazepam for symptom control (Yehya et al, 2004).
    2) WITH POISONING/EXPOSURE
    a) Severe encephalopathy associated with hyperammonemia has been reported following acute overdoses (Unal et al, 2007; Raskind & El-Chaar, 2000; Borbath et al, 2000; Hamer et al, 2000; Ellaway et al, 1999). Hyperammonemia has been reported even in the absence of hepatic failure following acute overdose (Rupasinghe & Jasinarachchi, 2011; Thabet et al, 2000).
    b) CASE REPORT: Hyperammonemia developed after acute overdose in a 20-year-old man with a partial deficiency of the enzyme carbamyl phosphate synthetase I. It was postulated that valproate induced further inhibition of the enzyme, preventing conversion of ammonia to urea (Felgenhauer et al, 1993).
    c) CASE REPORT: Severe hyperammonemia (623 mcg/dL) and increased serum aminotransferase concentrations were reported in an 18-year-old man following an overdose of 45 grams of sodium valproate. The patient recovered following supportive care (Lee et al, 1998).
    d) CASE REPORT: After ingesting 20 g (333 mg/kg) of valproic acid extended release, 60 mg of risperidone, and 3 g of venlafaxine extended release, a 19-year-old man developed hyperammonemia (peak ammonia concentration 1,191 mcg/dL at 65 hours postingestion). The valproic acid concentration peaked at 305.4 mcg/dL. Despite treatment with activated charcoal with sorbitol, and lactulose, the patient's condition deteriorated. The brain CT scan revealed cerebral edema and possible tentorial herniation. At approximately 120 hours after ingestion, cerebral blood flow studies showed an absence of cerebral perfusion, and the patient was pronounced brain dead (Camilleri et al, 2005).
    e) The exact mechanism of valproic acid induced hyperammonemia is unclear. Metabolites of valproic acid may inhibit CPS I, the first enzyme needed for ammonia elimination via the urea cycle. Production of these metabolites may be increased due to valproic acid induced deficiency of carnitine. Other studies have implicated increased renal production of ammonia in patients treated with valproic acid (Lheureux et al, 2005; Sztajnkrycer, 2002).
    C) INJURY OF LIVER
    1) WITH THERAPEUTIC USE
    a) Jaundice, drug-induced hepatitis, hepatocellular necrosis, transient elevated liver enzymes (SGPT/ALT, SGOT/AST), and fatal cholestatic hepatitis have been associated with chronic valproate administration (Lewis, 1978; Bruni & Wilder, 1979; Gerber et al, 1979; Addison & Gordon, 1980; Thruston et al, 1981; Suchy et al, 1979; Pinkston & Walker, 1997). Microvesicular steatosis is a characteristic lesion of VPA-induced hepatotoxicity (Thabet et al, 2000).
    b) CASE SERIES: Dreifuss et al (1987) reviewed all US cases of fatal hepatotoxicity coincident with valproate anticonvulsant therapy reported between 1978-1984 (Dreifuss et al, 1987). Thirty-seven fatalities were reported.
    1) All but one patient had other medical problems (mental retardation, developmental delay, congenital abnormalities, and other neurological diseases).
    2) The primary risk of fatal hepatic dysfunction (1/500) was found to be in children 0 to 2 years old receiving valproate as polytherapy. The risk declined with age and was low in patients receiving valproate as monotherapy (1/37,000).
    3) No hepatic fatalities occurred in patients above the age of 10 years receiving valproate as monotherapy.
    c) INCIDENCE/PEDIATRICS: Fatal hepatotoxicity is reported in 1/800 children under the age of 2 years following antiepileptic therapy with valproic acid (Raskind & El-Chaar, 2000). It is suggested that valproic acid may induce a carnitine deficiency in young children and result in nonspecific symptoms of deficiency, hepatotoxicity, and hyperammonemia. Carnitine supplementation may help prevent the onset of hepatotoxicity.
    D) LIVER ENZYMES ABNORMAL
    1) WITH THERAPEUTIC USE
    a) Elevated liver enzymes, and hyperammonemia have been reported following chronic administration of valproate (Lewis, 1978; Bruni & Wilder, 1979; Gerber et al, 1979; Addison & Gordon, 1980; Coulter & Allen, 1981; Rawat et al, 1981).
    1) NOTE: Increased blood ammonia concentrations have been seen with doses of 52 to 60 mg/kg/day and 500 mg twice daily chronically.
    b) CASE REPORT: A 33-year-old man on a combination of valproic acid and phenytoin therapy for seizures developed severe AST elevations (over 100 times normal upper limit) (McLaughlin et al, 2000).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) RENAL FAILURE SYNDROME
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Idiosyncratic acute renal failure, with acute tubular necrosis, and rhabdomyolysis were reported in conjunction with multiorgan system failure in a 52-year-old man diagnosed with valproic acid toxicity. Renal function returned to normal after hydration therapy and stopping valproic acid therapy (Pinkston & Walker, 1997).
    b) CASE REPORT: A 14-year-old handicapped girl with epilepsy was found to have a fever and hypokalemia after treatment with valproic acid for 7 years. Lab results revealed Fanconi syndrome. A renal biopsy showed interstitial nephritis. An alternative seizure drug was substituted and clinical symptoms improved (Yoshikawa et al, 2002).
    2) WITH POISONING/EXPOSURE
    a) Following a massive overdose, persistent metabolic acidosis and renal failure, without evidence of hepatic injury, have been reported (Christianson et al, 2001).
    b) CASE REPORT: A 2-month-old girl presented with generalized seizures, progressive coma, and anuria associated with a serum sodium valproate concentration of 1299 mcg/mL (Roodhooft et al, 1990).
    B) FANCONI SYNDROME
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A transient nephropathy syndrome similar to Fanconi syndrome (i.e., phosphaturia, kaliuria, proteinuria) was reported in a 15-year-old boy 2 days following the ingestion of 115 250-mg tablets. It resolved by the third day and the patient made a complete recovery (Su et al, 2002).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH THERAPEUTIC USE
    a) Acidosis has also been reported following therapeutic use of valproic acid.
    2) WITH POISONING/EXPOSURE
    a) Metabolic acidosis and elevated lactate concentrations, which may be persistent, have been reported in patients with severe overdose and hypotension (Farrar et al, 1993; Tank & Palmer, 1993; Lee et al, 1998; Johnson et al, 1999; Christianson et al, 2001; Kroll & Nand, 2002). Kupferschmidt et al (1998) reported an adult with hypotension and metabolic acidosis (pH 7.20) with an elevated anion gap (34 mmol/L) after ingestion of an unknown quantity of valproic acid (Kuperschmidt et al, 1998).
    b) INCIDENCE: In a series of 133 patients with valproate only overdose reported to a group of poison centers, 8 (6%) developed acidosis (Spiller et al, 2000). Acidosis was more common in patients with peak valproate concentrations of greater than 850 mcg/mL.
    c) CASE REPORT: Metabolic acidosis was reported in a 20-month-old who unintentionally ingested 1.5 grams of sodium valproate (Janssen et al, 1985).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) MYELOSUPPRESSION
    1) WITH POISONING/EXPOSURE
    a) Severe bone marrow depression, requiring transfusions, has been reported following severe acute valproic acid ingestions (Koelliker et al, 1999).
    b) CASE REPORT: Two women developed severe thrombocytopenia (platelets less than 20,000/mm3) and anemia after severe valproate overdose (Andersen & Ritland, 1995). One of them also developed leukopenia (WBC 1,200/mm3). Bone marrow suppression was most severe 4 to 5 days postingestion.
    c) CASE REPORT: A 43-year-old woman developed thrombocytopenia (platelet count 87,000/mm(3)) following an ingestion of approximately 19 grams of a slow-release form of valproate (Johnson et al, 1999).
    B) METHEMOGLOBINEMIA
    1) WITH POISONING/EXPOSURE
    a) Symptomatic methemoglobinemia has been described after acute valproate ingestion in a 3-year-old child. The child was admitted to the ED approximately 6 hours postingestion with initial oxygen saturation by pulse oximetry of 75% on an FiO2 of 1.00. Arterial blood was dark brown in color. Significant metabolic acidosis (pH 7.23) was noted. Initial methemoglobin concentration was 38.8%. Complete resolution of methemoglobinemia occurred after administration of a total of 4 mg/kg of methylene blue over 90 minutes (Lynch & Tobias, 1998).
    C) THROMBOCYTOPENIC DISORDER
    1) WITH THERAPEUTIC USE
    a) Inhibition of secondary platelet aggregation, prolonged bleeding times, and thrombocytopenia have been reported following therapeutic use (Lewis, 1978; Bruni & Wilder, 1979; Gerber et al, 1979; Addison & Gordon, 1980; Hintze et al, 1987; Gidal et al, 1994). In most cases, abnormalities resolve with dose reduction alone; discontinuation of drug use is usually not necessary (Acharya & Bussel, 2000).
    b) Significant risk factors for developing thrombocytopenia or a clinically significant drop in platelets are dosage greater than 1000 mg/day and age older than 65 years (Conley et al, 2001).
    c) A case of thrombocytopenia-induced fatal pulmonary hemorrhage was reported in a 30-year-old woman receiving valproate monotherapy (Sleiman et al, 2000). It has been suggested that viral infections may be associated with thrombocytopenia in individuals receiving valproate therapy.
    d) A 67-year-old woman, with Lennox-Gasteau syndrome, developed severe, isolated thrombocytopenia after being placed on a combination of carbamazepine and valproate for the treatment of generalized tonic-clonic seizures. The patient received carbamazepine 150 mg/day for 7 days, 600 mg/day on day 8, and received 1200 mg/day by day 9. On day 10, valproate 300 mg/day was added because of nonconvulsive status epilepticus. The valproate was discontinued 5 days later because the patient developed urticaria and a maculopapulous exanthema. The thrombocyte count was 262 GIGA/L (normal: 150-360 GIGA/L) on day 5 and had dropped to 5 GIGA/L by day 15, at which time carbamazepine was also discontinued. The patient received two thrombocyte transfusions and her thrombocyte count was within normal limits 3 days after the carbamazepine was discontinued. It could not be determined whether it was carbamazepine alone or the combination of carbamazepine and valproate that was responsible for the severe thrombocytopenia (Finsterer et al, 2001).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 39-year-old woman with epilepsy ingested approximately 50,000 mg of valproic acid (VPA) with an initial serum concentration above 110 mcg/mL (above the level detectable by the laboratory) and a platelet count of 261 k/cumm. She was decontaminated and admitted to ICU for persistent drowsiness. On day 4, the patient developed a sudden drop in her platelet count to 46 k/cumm, with a VPA level of 12 mcg/mL. By the eighth day, her serum VPA level was 22.7 mcg/mL with a platelet count of 55 k/cumm. By day 11, her platelet count had normalized and she was discharged to home (Lin et al, 2008). The authors suggest that inadvertent or intentional exposures can result in delayed thrombocytopenia and monitoring may be required for approximately a week after exposure.
    b) INCIDENCE: In a series of 133 patients with valproate only overdose reported to a group of poison centers, 11(8%) developed thrombocytopenia (less than 150,000/mm3) (Spiller et al, 2000).
    D) LEUKOPENIA
    1) WITH THERAPEUTIC USE
    a) CASE SERIES: In a on year prospective study involving 45 patients receiving valproic acid, neutropenia developed in 12 and thrombocytopenia in 15, but both disorders were transient and self-limiting despite continued treatment (Barr et al, 1982).
    b) CASE REPORT: A later case report described severe neutropenia in a child which necessitated withdrawal of the drug (Symon & Russell, 1983).
    c) CASE SERIES: In a study of 1,251 hospitalized patients receiving valproate therapy, 6 developed moderate to severe leukopenia (WBC less than 4,000/mm3); 2 of these patients were also taking carbamazepine (Tohen et al, 1995).
    2) WITH POISONING/EXPOSURE
    a) INCIDENCE: In a series of 133 patients with valproate only overdose reported to a group of poison centers, 4 (3%) developed leukopenia (Spiller et al, 2000).
    E) PANCYTOPENIA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Fatal pancytopenia developed in a 3-year-old child administered high-dose valproate therapy (Rajantie et al, 1992).
    F) APLASTIC ANEMIA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Red cell aplasia in one child, on 2 occasions appeared to be associated with sodium valproate administration. The aplasia promptly resolved after drug withdrawal (MacDougall, 1982).
    G) HEMATOLOGY FINDING
    1) WITH THERAPEUTIC USE
    a) In a cohort study, involving 29,357 recipients of anticonvulsive therapy receiving 684,706 prescriptions of 4 different drugs, one being valproate, serious blood dyscrasias were rarely found in these patients. Among the 4 drugs, rates did not appear different. An overall rate of blood dyscrasias was reported to be 3 to 4 per 100,000 prescriptions for all 4 drugs (Blackburn et al, 1998).
    H) BLOOD COAGULATION DISORDER
    1) WITH THERAPEUTIC USE
    a) Below-normal concentrations of von Willebrand Factor activity was observed in 6 of 29 (21%) children who had been taking valproic acid for at least 6 months for the treatment of epilepsy. The 6 children were regarded as having "acquired von Willebrand's Disease." No correlation was found between von Willebrand factor activity and dose or blood concentration of valproic acid or duration of therapy. The authors cautioned that when surgery is necessary, factor VIII von Willebrand factor concentrations should be supplemented (Serdaroglu et al, 2002).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) ALOPECIA
    1) WITH THERAPEUTIC USE
    a) Skin rashes and hair loss may occur (Lewis, 1978; Bruni & Wilder, 1979; Gerber et al, 1979). Five of 250 patients developed curly hair at dosage of 1 g/day. In 3/5 patients, this effect followed temporary alopecia (Jearons, 1977).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: Alopecia developed in a 37-year-old woman after massive acute ingestion of valproic acid. Initial serum valproate concentration was 1579 mcg/mL (Palatnick et al, 1989).
    B) BULLOUS ERUPTION
    1) WITH POISONING/EXPOSURE
    a) Superficial bullous lesions associated with a severe valproic acid ingestion in a 4-year-old child was reported. A serum valproic acid concentration of 804 mcg/mL was noted approximately 22 hours after ingestion. During the 4 days following ingestion, progression of the bullous skin lesions occurred and new regions of fluid-filled bullae developed (Koelliker et al, 1999).
    C) DERMATITIS
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Pellagra-like syndrome developed in a young boy caused by valproate-induced niacin deficiency (Gillman & Sandyk, 1984).
    D) STEVENS-JOHNSON SYNDROME
    1) WITH THERAPEUTIC USE
    a) Stevens Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) have been described occasionally during the early stages of valproic acid therapy. In a case-control study of patients taking various antiepileptic agents, 73 cases of SJS or TEN were identified; of these, 13 were due to valproic acid ingestion. SJS/TEN generally occurred during the first 8 weeks of therapy (Rzany et al, 1999).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) Rhabdomyolysis may occur following a massive acute overdose. Rhabdomyolysis, with a serum CPK greater than 32,000 U/L, was reported in a 4-year-old child following a severe valproic acid overdose with prolonged coma (Koelliker et al, 1999).
    B) MUSCULOSKELETAL FINDING
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Valproate-induced systemic lupus erythematosus (SLE) was reported in a 30-year-old woman with partial trisomy of chromosome 9 treated for one year with valproate and ethosuximide polytherapy. She developed arthralgias, muscle weakness, fatigue and fever. Lab results showed increased sedimentation rate, hypergammaglobulinemia and high antinuclear antibodies titers. Antihistone antibodies and HLA-DR4 antigen were detected. All symptoms of SLE were resolved after the valproate was discontinued (Gigli et al, 1996).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPERGLYCEMIA
    1) WITH THERAPEUTIC USE
    a) Hyperglycemia has been reported following chronic administration of valproate.
    B) HYPOGLYCEMIA
    1) WITH THERAPEUTIC USE
    a) IN UTERO EXPOSURE
    1) CASE REPORT: An infant born at 35 weeks gestation secondary to placenta previa was exposed in utero (mother with a history of complex partial epilepsy) to valproic acid (600 mg/day) and phenytoin (200 mg/day) developed jitteriness and hypoglycemia (32 mg/dL) on the second day of life. A glucose infusion was started and the jitteriness resolved. Due to suspected withdrawal, serum valproic acid (37.8 mcg/mL {normal: 50 to 100 mcg/mL} and phenytoin (6.37 mcg/dL {normal: 10 to 20 mcg/mL} levels were measured. A second episode of hypoglycemia (38 mg/dL) occurred on day 6, and IV glucose was continued until day 10 when the infant was fully tolerating oral feedings. No further episodes of hypoglycemia were reported and the infant was discharged to home (Coban et al, 2010).
    2) CASE SERIES: Thirteen of 22 infants exposed in utero (throughout pregnancy) to valproic acid became hypoglycemic (lowest blood glucose concentration reported was 1.0 mmol/L). No episode was reported as symptomatic. No evidence for hyperinsulinemia as a cause for hypoglycemia existed. All infants were treated with glucose infusions. Withdrawal syndrome (irritability, jitteriness, hypertonia, seizures, vomiting) was reported in 10 of the infants, beginning 12 to 24 hours after birth and lasting 2 to 7 days (Ebbesen et al, 2000a).
    2) WITH POISONING/EXPOSURE
    a) In a series of 133 patients with valproate only overdose reported to a group of poison centers, one patient developed hypoglycemia (Spiller et al, 2000).

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) ACUTE ALLERGIC REACTION
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: After 5 weeks of treatment with sodium valproate, a 28-year-old woman suddenly developed a generalized maculo-papulous eruption with fever, lymph node enlargement, hypereosinophilia and altered liver function. Drug-induced hypersensitivity to valproic acid is reported as rare (Picart et al, 2000).
    B) AIDS
    1) WITH THERAPEUTIC USE
    a) Valproate therapy may reduce intracellular concentrations of glutathione and inhibit activity of glutathione reductase in human red blood cells. There may be a link between intracellular concentrations of glutathione and the progression of HIV disease. Decreased glutathione concentrations may activate the replication of HIV. In vitro studies with cell lines infected with HIV showed the addition of valproate increased viral expression and replication at therapeutic drug concentrations (Hardy & Nardacci, 1999).

Reproductive

    3.20.1) SUMMARY
    A) Valproic acid is a known teratogen to humans. Maternal use of valproic acid alone or in combination with other antiepileptic drugs has been found to cause valproic acid syndrome in infants. Facial dysmorphology, congenital heart defects, spina bifida, cleft lip and palate, and developmental delays are some of the teratogenic effects seen.
    B) In 2009, the FDA notified healthcare professionals and patients about an increased risk of congenital malformations (ie, neural tube defects, craniofacial defects, cardiovascular malformations, and other body system malformations) in infants born of women exposed to valproate sodium and related products during pregnancy.
    C) The FDA has classified valproic acid as pregnancy category X and its use in pregnant women is contraindicated for the prevention of migraine headaches. However, valproate products continue to have a pregnancy category of D for treatment of epilepsy and manic episodes associated with bipolar disorder. Valproate products should only be used in pregnant women with epilepsy or bipolar disorder if other medications are not effective or are otherwise unacceptable in treating the condition.
    D) Valproic acid is excreted in breast milk with low concentrations and is not contraindicated during breastfeeding.
    3.20.2) TERATOGENICITY
    A) CONGENITAL MALFORMATIONS
    1) In a retrospective review, valproic acid monotherapy during the first trimester resulted in a significantly increased risk of 14 major fetal congenital malformations. Overall, there were 118 infants with malformations out of all exposed pregnancies (n=1565), resulting in a malformation rate of 7.5% (95% confidence interval (CI), 6.3% to 9%). In an adjusted case-control subgroup analysis of infants with malformations, women who received valproic acid during the first trimester (n=122) had a significantly increased risk of delivering an infant with spina bifida (odds ratio (OR), 12.7; 95% CI, 7.7 to 20.7), atrial septal defect (OR, 2.5; 95% CI, 1.4 to 4.4), cleft palate (OR, 5.2; 95% CI, 2.8 to 9.9), hypospadias (OR, 4.8; 95% CI, 2.9 to 8.1), polydactyly (OR, 2.2; 95% CI, 1 to 4.5), or craniosynostosis (OR, 6.8; 95% CI, 1.8 to 18.8) compared with controls with no valproic acid exposure (n=45). Compared with other antiepileptic monotherapies, valproic acid monotherapy resulted in a significantly increased risk of ventricular septal defect, but not craniosynostosis. There was also a significantly increased risk of limb reduction (crude OR, 3.4; 95% CI, 1.6 to 7.2) in infants exposed to valproic acid compared with no exposure (Jentink et al, 2010).
    2) According to data from the North American Antiepileptic Drug (NAAED) Pregnancy Registry, infants born to women with epilepsy treated with valproate monotherapy had an almost 5-fold increased risk of major malformations (ie, neural tube defects, craniofacial defects, cardiovascular malformations, and other body system malformations) compared with those who received a different antiepileptic drug. The major malformation rate was 10.7% (95% CI, 6.3% to 16.9%) among infants born to women who received valproic acid 500 mg to 2000 mg/day during pregnancy (n=149) compared with 2.9% (95% CI, 2% to 4.1%) among infants of women who received any other antiepileptic drug monotherapy (n=1048) (Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015; Prod Info Stavzor(R) oral delayed release capsules, 2013; US Food and Drug Administration, 2009).
    3) Congenital malformations in a study of 715 cases included neural tube defects in 7 (1%), facial cleft in 11 (1.5%), cardiac anomalies in 5 (0.7%), hypospadias and genitourinary defects in 9 (1.3%), gastrointestinal tract defects in 2 (0.3%), and skeletal malformations in 8 (1.1%) (Morrow et al, 2006).
    4) CASE REPORT: An infant exposed in utero to valproic acid 250 mg/day was born at 38-weeks gestation with characteristics of fetal valproate syndrome (including neural tube defects) to a 21-year-old woman with a history of seizures and incomplete prenatal care. Her routine dose was 500 mg/day, but was decreased to 250 mg/day when the pregnancy was diagnosed during the first month. At 36-weeks gestation, an ultrasound (US) revealed craniosynostosis, extremity flexion abnormalities, cardiomegaly, bilateral renal hypoplasia, and hydrocephaly. A fetal echocardiography revealed Ebstein anomaly, secundum atrial septal defect, right ventricle hypertrophy, and pulmonary stenosis. Multiple facial, limb, and abdominal wall defects (ie, frontal bossing, micrognathia, arachnodactyly, and pectus excavatum) were observed upon examination at birth. Abdominal US of the infant at birth showed a right multicystic dysplastic kidney and ophthalmologic examination revealed bilateral optic nerve hypoplasia. A head CT confirmed trigonocephaly, hydrocephaly, and craniosynostosis. The infant was intubated and received supportive care, but died 15 days after birth as a result of respiratory failure (Ozkan et al, 2011).
    B) SPINA BIFIDA
    1) Exposure to valproic acid during pregnancy has been associated with fetal effects, primarily neural tube effects. There are several studies which support the association of neural tube defects (spina bifida) with valproic acid exposure during the first trimester (Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Depakote oral tablets, 2015; Robert et al, 1983; Robert, 1988; Jeavons, 1982; Verloes et al, 1990; Ardinger et al, 1988).
    C) SKELETAL MALFORMATION
    1) Valproic acid may also be associated with a specific syndrome of facial anomalies. One study evaluated 7 children who had been exposed to valproic acid in utero. A consistent facial phenotype was observed in all 7. The facial changes consisted of epicanthal folds which continued to form a crease or groove just under the eye socket, a flat nasal bridge, small upturned folds, long upper lip and down turned angles of the mouth. Major anomalies also consist of congenital heart defects and developmental delay in more than two-thirds of cases (DiLiberti et al, 1984). This valproic acid syndrome has also been noted by other authors (Verloes et al, 1990; Ardinger et al, 1988; Lewis et al, 1998). Another study reported an infant with facial dysmorphology (Dalens et al, 1980).
    2) Two studies of anomalous right pulmonary artery origin confirmed by cardiac catheterization were reported in infants diagnosed with valproate syndrome (Mo & Ladusans, 1999).
    3) The risk of congenital anomalies is greater in infants whose mothers were treated with multiple antiepileptic drugs at the same time. Valproic acid in combination with other primary antiepileptic drugs may be more teratogenic than other combinations of antiepileptic drugs.
    a) Women who took only 1 antiepileptic during pregnancy had a 3% risk factor for giving birth to an infant with congenital anomalies. This compares with a 2% risk in the untreated epileptic mother. This risk increased to 5% in women who took 2 antiepileptics during pregnancy, 10% with 3 antiepileptics taken, and 20% in women who took 4 antiepileptics during pregnancy (Polifka & Friedman, 2002).
    D) EYE ABNORMALITY
    1) Septo-optical dysplasia and hypoplasia of the optic chiasm on MRI have been reported from maternal exposure to valproic acid (McMahon & Braddock, 2001).
    2) CASE STUDY: A group of 46 children, ages 8 months to 16 years 5 months, diagnosed with fetal anticonvulsant syndrome were exposed in utero to valproic acid (29 as monotherapy). The remainder were exposed to other anticonvulsants. Thirty-one of 46 children (67%) had ocular abnormalities. Myopia was the most common (14 of 28 or 50%) in those exposed to valproic monotherapy. Strabismus, astigmatism, and anisometropia were also common (Glover et al, 2002).
    E) CLEFT PALATE
    1) Other fetal abnormalities have included cleft lip and palate with low birth weight, birth marks, abnormally long thin fingers, laryngeal hypoplasia, hydranencephaly, and severe problems in the respiratory system (Pinder et al, 1977; Huot et al, 1987) (Barrera et al, 1994).
    F) FIBRINOGEN PLASMA DECREASED
    1) Cases of neonatal fibrinogen depletion associated with sodium valproate ingestion in epileptic mothers have been reported. In 2 cases, no other medications were taken by the mothers and the pregnancies were described as uneventful. In 1 case, the newborn died within a few hours of afibrinogenemia (Bavoux et al, 1994).
    G) LEARNING DISABILITIES
    1) A case series of 39 children exposed to valproic acid in utero noted a trend towards lower IQ compared with controls, though this trend was not statistically significant. However, the mothers of these children had a statistically significant lower average IQ compared with controls, further confounding the study results (Eriksson et al, 2005).
    2) Children who were exposed to valproate prenatally (n=62) had lower IQ scores at age 6 (IQ, 97; 95% CI, 94 to 101) compared with children who had prenatal exposure to other antiepileptic drugs, including lamotrigine (IQ, 108; 95% CI, 105 to 110), carbamazepine (IQ, 105; 95% CI, 102 to 108), and phenytoin (IQ 108; 95% CI, 104 to 112) (Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015).
    H) BALLER-GEROLD SYNDROME
    1) CASE REPORTS: Three infants with a history of maternal sodium valproate use (1500 to 2000 mg/day) presented with a combination of metopic suture synostosis and preaxial upper limb malformations (absence or hypoplasia of the radius and/or thumb), which could be diagnosed as Baller-Gerold syndrome. All 3 patients underwent surgery for the craniofacial deformities; standard frontocranial reconstruction was performed. Some degree of mental retardation was documented in 2 of the patients (SantosdeOliveira et al, 2006).
    I) ANIMAL STUDIES
    1) RAT, MOUSE, RABBIT, HAMSTER, MONKEY: There is a large volume of data in many animal species. There are several studies which report post-implantation mortality in the rat, mouse, and rabbit. Valproic acid is a teratogen in the rat, mouse, rabbit, hamster, and monkey (RTECS , 1996). Exposure of pregnant mice or rats to valproic doses several times greater than those used in human, but which produce serum valproic acid levels within the human therapeutic range, produces embryonic death and malformations in offspring (Nau & Loscher, 1986; Nau & Scott, 1986; Vorhees, 1987; Ritter et al, 1987; Binkerd et al, 1988; Finnell et al, 1988).
    2) MICE, RATS, HAMSTERS: Exencephaly and skeletal anomalies are among the most common malformations observed in mice. Cardiac, skeletal, and urinary tract anomalies are seen most often in rats. In both species there is a dose-response relationship in the teratogenic effect. Hamsters treated with 5 times the human dose of valproic acid during pregnancy produce offspring with an increased frequency of neural tube defects (Moffa et al, 1984). Spina bifida occulta, exencephaly, and exophthalmia were reported in mice (Emmanouil-Nikoloussi et al, 2004). Conn et al (1982) reported a study on the effects of valproic acid on the fertility of male rats. There was a decrease in the prostate and epididymal weights. The rats also exhibited diminished sperm content and motility and their fertility was decreased. Valproic acid decreased cartilage formation within the growth plate of cultured rat metatarsal bones, leading to shorter overall length (Wu et al, 2004).
    3) MICE: Valproic acid-induced neural tube defects were diminished in mice by coadministration of pantothenic acid; however, the latter did not affect incidence of other external or skeletal malformations (Sato et al, 1995).
    4) RATS, MICE: Rats were less sensitive than mice for induction of structural defects: a dose of 150 mg/kg produced approximately 8.5% malformations in rats and 32% in mice. The most common defect in mice was encephalopathies, while rats were most susceptible to axial skeletal defects (Menegola et al, 1996).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) Valproate products are classified as FDA pregnancy category X in women treated for migraine prophylaxis and pregnancy category of D in women treated for epilepsy or manic episodes associated with bipolar disorder. Valproate products should only be used in pregnant women with epilepsy or bipolar disorder if other medications are not effective or are otherwise unacceptable in treating the condition (Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015; US Food and Drug FDA Drug Safety Valproate).
    2) Valproate should not be administered to women of childbearing age unless absolutely necessary for the treatment of a medical condition. The risks and benefits of this drug should be weighed in women who are pregnant or of childbearing age (Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015).
    3) Women treated with valproate for epilepsy should not abruptly discontinue this drug as it may precipitate seizures that endanger the mother and fetus (Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015).
    4) For women using valproate during pregnancy, dietary folic acid supplementation should be recommended both prior to conception and during pregnancy to reduce the risk of neural tube defects, prenatal diagnostic testing should be offered, and clotting parameters should be carefully monitored in the mother and, if necessary, the fetus (Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015).
    5) Infants born to mothers treated with valproate during pregnancy should have blood glucose levels monitored during the first several hours of life (Ebbesen et al, 2000).
    6) The North American Antiepileptic Drug (NAAED) Pregnancy Registry has been established, and prescribers can register patients by calling 1-888-233-2334 (Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015; Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Stavzor(R) oral delayed release capsules, 2013; US Food and Drug Administration, 2009).
    B) AUTISM SPECTRUM DISORDERS
    1) In a large cohort study, an increased risk of autism spectrum disorders and childhood autism was seen among children with prenatal exposure to valproate (n=508) compared with children with no prenatal valproate exposure (n=655,107). Analyses of population records over a 10-year period identified children of mothers who filled prescriptions of valproate within a range of 30 days before estimated conception to birth. After 14 years of followup, prenatal valproate exposure was associated with adjusted hazard ratios for autism spectrum disorder of 2.9 (95% CI, 1.7 to 4.9) (n=14) and 5.2 (95% CI, 2.7 to 10) for childhood autism (n=9). The adjusted hazard ratios for autism spectrum disorder (1.7 [95% CI, 0.9 to 3.2]; n=11) and childhood autism (2.9 [95% CI, 1.4 to 6]; n=8) in 432 valproate exposed offspring of mothers with epilepsy were lower when compared to the risk of autism spectrum disorder (4.4 [95% CI, 1.4 to 13.6]; n=3) and childhood autism (3.9 [95% CI, 0.5 to 28.9]; n=1) among 76 valproate exposed children of mothers without epilepsy (Christensen et al, 2013).
    C) COGNITIVE IMPAIRMENT
    1) Adjusted analyses of the prospective, observational Neurodevelopmental Effects of Antiepileptic Drugs (NEAD) study found that mean IQ at ages 2, 3, 4.5, and 6 years (n=311) following prenatal exposure to antiepileptic drugs (AEDs) valproate, carbamazepine, lamotrigine, or phenytoin correlated with maternal IQ for all AEDs but valproate and remained consistent over time. Dose-dependent effects were observed for valproate but not for each of the other AEDs. At age 6 years, children with prenatal valproate exposure exhibited IQ reductions of 7 to 10 points compared with children treated with the other AEDs. Using the Bayley Scale of Infant Development (BSID) at age 2 and the Differential Ability Scale (DAS) at ages 3, 4.5, and 6 to assess cognitive development, adjusted mean IQ at age 6 in the carbamazepine group was 106 (n=61; range, 103 to 109); 108 in the lamotrigine group (n=74; range, 105 to 111); 109 in the phenytoin group (n=40; range, 105 to 113), and 98 in the valproate group (n=49; range, 95 to 102). IQ improved as children aged and percentages of cognitively impaired children decreased across all groups. However, children in the valproate group showed significantly lower adjusted mean verbal index scores (97; range, 94 to 100; p value not applicable) compared with children in the carbamazepine (104; range, 102 to 107; p vs valproate 0.0005), lamotrigine (105; range, 102 to 107; p vs valproate 0.0003), and phenytoin (106; range, 102 to 109; p vs valproate 0.0005) groups at age 6. In addition, adjusted mean non-verbal index scores were significantly lower in the valproate group (101; range, 98 to 104; p value not applicable) compared with the lamotrigine group (108; range, 105 to 110; p vs valproate 0.0015) (Meador et al, 2013).
    2) Children who were exposed to valproate medications during pregnancy had statistically significant lower IQ scores at age 6 compared with children who had prenatal exposure to other antiepileptic drugs. The mean IQ score for children exposed to valproate products (n=62) in utero was 97 (95% CI, 94 to 101) as compared with 105 (95% CI, 102 to 108) for carbamazepine (n=94), 108 (95% CI, 105 to 110) for lamotrigine (n=100), and 108 (95% CI, 104 to 112) for phenytoin (n=55). However, these findings should be interpreted with caution, as the effect of periconceptional folate use, which is associated with higher mean IQs in children, was not a primary outcome of the study and information about its use were collected retrospectively (Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015; Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; US Food and Drug FDA Drug Safety Valproate).
    3) In epidemiologic studies, children (ages 3 to 16) born to women who took valproate sodium or related antiseizure drugs (valproic acid and divalproex sodium) throughout pregnancy had lower cognitive test scores compared with children that were exposed in utero to other antiseizure medicines and those with no antiepileptic drug exposure. In the largest of the studies, a prospective cohort trial conducted in the United States and United Kingdom, children with prenatal exposure to valproate tested lower on the Differential Ability Scale (92; 95% confidence interval (CI), 88 to 97) compared with those exposed to lamotrigine (101; 95% CI, 98 to 104), carbamazepine (98; 95% CI, 95 to 102), or phenytoin (99; 95% CI, 94 to 104). Risk associated with less than full-term fetal exposure to valproate sodium has not been determined (U.S. Food and Drug Administration (FDA), 2011).
    4) In children born to mothers who were taking valproic acid monotherapy during pregnancy, 4 of 21 (19%) had a score less than 80 on the full scale intelligence quotient (FIQ) and 2 of 21 (10%) had a score less than 70. The mothers taking valproic acid during pregnancy scored significantly lower in FIQ, verbal IQ, and performance IQ tests although most results fell within the normal range. In this study, mothers taking valproic acid also had significantly fewer years of education compared with mothers taking carbamazepine monotherapy and those without antiepileptic drug treatment (Eriksson et al, 2005).
    D) HYPOGLYCEMIA
    1) CASE REPORT: An infant born at 35 weeks gestation secondary to placenta previa was exposed in utero (mother with a history of complex partial epilepsy) to valproic acid (600 mg/day) and phenytoin (200 mg/day) developed jitteriness and hypoglycemia (32 mg/dL) on the second day of life. A glucose infusion was started and the jitteriness resolved. Due to suspected withdrawal, serum valproic acid (37.8 mcg/mL {normal: 50 to 100 mcg/mL} and phenytoin (6.37 mcg/dL {normal: 10 to 20 mcg/mL} levels were measured. A second episode of hypoglycemia (38 mg/dL) occurred on day 6, and IV glucose was continued until day 10 when the infant was fully tolerating oral feedings. No further episodes of hypoglycemia were reported and the infant was discharged to home (Coban et al, 2010).
    2) CASE SERIES: Thirteen of 22 infants exposed in utero (throughout pregnancy) to valproic acid became hypoglycemic (lowest blood glucose concentration reported was 1 mmol/L). No episode was reported as symptomatic. No evidence for hyperinsulinemia as a cause for hypoglycemia existed. All infants were treated with glucose infusions. Withdrawal syndrome (irritability, jitteriness, hypertonia, seizures, vomiting) was reported in 10 of the infants, beginning 12 to 24 hours after birth and lasting 2 to 7 days (Ebbesen et al, 2000a).
    E) RESPIRATORY DISTRESS
    1) CASE REPORT: An infant exposed in utero to valproic acid 250 mg/day was born at 38-weeks gestation with respiratory distress and characteristics of fetal valproate syndrome (including neural tube defects) to a 21-year old woman with a history of seizures who received incomplete prenatal care. Her routine dose was 500 mg/day, but was decreased to 250 mg/day when the pregnancy was diagnosed during the first month. The infant was intubated and treated with supportive care, but died 15 days after birth as a result of respiratory failure (Ozkan et al, 2011).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) Valproic acid is excreted into breast milk (Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015; Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Stavzor(R) oral delayed release capsules, 2013), with concentrations of about 1% to 15% of maternal serum concentrations; therefore, the drug is NOT contraindicated during breastfeeding (Dickinson et al, 1979; Von Uhruh et al, 1984; Rimmer & Richens, 1985; Briggs et al, 1998; Iqbal et al, 2001). The American Academy of Pediatrics has concluded that use of valproic acid is compatible with breastfeeding (AAP, 1994).
    2) In 2 mother-infant pairs, serum valproate concentrations were 1.5% and 6%, respectively, of the maternal values (Wisner & Perel, 1998).
    3) In a series of 6 breastfeeding mother-infant pairs, one study reported serum valproate concentrations in nursing mothers to be near or within the therapeutic range while serum concentrations in the infants were low, ranging from 0.7 to 1.5 mcg/mL (0.9% to 2.3% of maternal serum concentrations) (Piontek et al, 2000).
    4) CASE REPORT: A 3-month-old breastfed infant, whose mother was treated with sodium valproate (1200 mg/day), presented to the hospital with thrombocytopenic purpura and anemia. Following discontinuation of breastfeeding, the infant recovered (Stahl et al, 1997).
    5) Exercise caution when administering a valproate product to a nursing woman (Prod Info Depacon intravenous injection, 2015; Prod Info DEPAKENE oral capsules, oral solution, 2015; Prod Info Depakote oral tablets, 2015a; Prod Info Depakote ER oral extended-release tablets, 2015; Prod Info DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, 2015).
    3.20.5) FERTILITY
    A) ENDOCRINE DISORDER
    1) In a study of men (n=21) taking valproic acid for epileptic control, 12 men (57%) developed increased serum androgen concentrations, but this was not associated with elevations in circulating LH or insulin concentrations. Mean serum concentration of androstenedione was high (3.7 mcg/L) in these patients (Rattya et al, 2001).

Carcinogenicity

    3.21.1) IARC CATEGORY
    A) IARC Carcinogenicity Ratings for CAS99-66-1 (International Agency for Research on Cancer (IARC), 2016; International Agency for Research on Cancer, 2015; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2010; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2010a; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2008; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2007; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2006; IARC, 2004):
    1) Not Listed
    3.21.2) SUMMARY/HUMAN
    A) At the time of this review, no data were found as to the carcinogenic effects of valproic acid in humans.
    3.21.4) ANIMAL STUDIES
    A) CARCINOMA
    1) A study of Wistar rats and B6C3F1 mice showed adenocarcinomas of the uterus and cervix when exposed to calcium valproate. The statistically significant (P<0.01) increase in uterine adenocarcinomas found in females contrasts the absence of this tumor type in a previous rat bioassay study with valproic acid Subcutaneous fibrosarcomas were significantly increased in valproic acid treated males, but no uterine tumors were reported in females. Researchers are unsure why carcinogenic potential in the calcium salt of valproic acid would be expressed in markedly different target organs (Watkins et al, 1992).

Genotoxicity

    A) At the time of this review, little human data regarding potential genotoxicity is available.

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Valproate concentration should be measured (therapeutic level 50 to 100 mcg/mL) and repeated every 4 to 6 hours until downward trend is established.
    B) Serum concentrations greater than 450 mcg/mL is associated with drowsiness/obtundation; greater than 850 mcg/mL is associated with coma.
    C) Monitor blood gases, liver enzymes, ammonia concentration, lactic acid, electrolytes, blood sugar, renal function and ECG in patients with significant toxicity.
    D) Valproate causes false elevation of urine ketones.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Peak concentrations may be delayed 6 to 8 hours or longer after overdose, particularly with enteric coated formulations (Meyer et al, 2005; Kay et al, 2003; Tank & Palmer, 1993). Therapeutic concentrations are considered to be between 50 and 100 mcg/mL. Serum concentrations are not reliable for predicting severity of CNS depression in valproic acid poisoning cases; however, more serious effects (e.g., coma, acidosis, aspiration, respiratory depression) are generally seen at concentrations >850 mcg/mL (Spiller et al, 2000).
    2) Follow serum electrolytes, hepatic enzymes and serum ammonia concentrations in patients with significant symptoms. Hyperammonemia and hyperlactacidemia may be significant following severe overdose.
    B) HEMATOLOGY
    1) Monitor platelet count in symptomatic patients; thrombocytopenia has been reported (Lin et al, 2008).
    C) ACID/BASE
    1) Follow arterial blood gases or pulse oximetry in patients with significant CNS depression.
    4.1.3) URINE
    A) URINALYSIS
    1) False urinary tests for ketones and glucose have been noted following daily therapeutic doses.

Methods

    A) IMMUNOASSAY
    1) Serum valproic acid concentrations should be available from all laboratories doing anticonvulsant screens. An anticonvulsant screen should be obtained on all patients due to the common availability of other anticonvulsant agents to the epileptic patient.
    a) Inter- and intra-assay measurement differences may become clinically significant at very elevated valproic acid concentrations after overdose. Benton et al (2001) reported the CEDIA immunoassay produced consistently higher readings for valproic acid than three other assays it was compared with.
    2) An EMIT(R) homogeneous enzyme immunoassay is available for quantitation of valproic acid in serum or plasma. The assay's range of quantitation is 10 to 150 mcg/mL valproic acid. Clinical studies show excellent correlation between this method and GLC.
    a) In a single patient, on the average, the Emit assay overestimated the true valproic acid concentration by 50%. The overestimate by EMIT reduced to 16% when concentrations of measured metabolites were added to that of valproic acid as determined by GLC (Dupuis et al, 1990).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients with rising valproate concentrations and those developing CNS depression or other clinical or laboratory evidence of toxicity should be admitted. Patients with persistent altered mental status, abnormal vital signs, acidosis, renal or hepatic involvement should be admitted to an intensive care setting.
    6.3.1.2) HOME CRITERIA/ORAL
    A) Asymptomatic patients with unintentional ingestion of less than 50 mg/kg can be observed at home (Manoguerra et al, 2008).
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity, or in whom the diagnosis is not clear.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Symptomatic patients, those with deliberate ingestions, and those with unintentional ingestions of 50 mg/kg or more should be referred to a medical facility for evaluation and treatment (Manoguerra et al, 2008). Monitor serial valproate concentrations every 2 to 3 hours. Patients should be observed until serial valproate concentrations are clearly declining on at least two sequential measurements, and symptoms have resolved. Patients should be observed for a minimum of 6 hours after immediate release valproate ingestions, and for a minimum of 12 hours following delayed release preparations (time to maximum concentration after therapeutic dose of extended release formulation is 4 to 17 hours) (Prod Info Depakote ER oral extended release tablets, 2013).

Monitoring

    A) Valproate concentration should be measured (therapeutic level 50 to 100 mcg/mL) and repeated every 4 to 6 hours until downward trend is established.
    B) Serum concentrations greater than 450 mcg/mL is associated with drowsiness/obtundation; greater than 850 mcg/mL is associated with coma.
    C) Monitor blood gases, liver enzymes, ammonia concentration, lactic acid, electrolytes, blood sugar, renal function and ECG in patients with significant toxicity.
    D) Valproate causes false elevation of urine ketones.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) PREHOSPITAL
    1) Ipecac-induced emesis is not recommended in the prehospital setting because of the potential for aspiration.
    B) NALOXONE
    1) In patients with valproic-induced coma and/or significant respiratory depression, naloxone may be considered in the prehospital setting (Manoguerra et al, 2008).
    C) ACTIVATED CHARCOAL
    1) Activated charcoal may be considered in the prehospital setting by a healthcare professional only, if the patient is asymptomatic, the ingestion is recent, and there are no contraindications. However, transportation to an emergency center should NOT be delayed to administer activated charcoal (Manoguerra et al, 2008).
    2) PREHOSPITAL ACTIVATED CHARCOAL ADMINISTRATION
    a) Consider prehospital administration of activated charcoal as an aqueous slurry in patients with a potentially toxic ingestion who are awake and able to protect their airway. Activated charcoal is most effective when administered within one hour of ingestion. Administration in the prehospital setting has the potential to significantly decrease the time from toxin ingestion to activated charcoal administration, although it has not been shown to affect outcome (Alaspaa et al, 2005; Thakore & Murphy, 2002; Spiller & Rogers, 2002).
    1) In patients who are at risk for the abrupt onset of seizures or mental status depression, activated charcoal should not be administered in the prehospital setting, due to the risk of aspiration in the event of spontaneous emesis.
    2) The addition of flavoring agents (cola drinks, chocolate milk, cherry syrup) to activated charcoal improves the palatability for children and may facilitate successful administration (Guenther Skokan et al, 2001; Dagnone et al, 2002).
    3) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    6.5.2) PREVENTION OF ABSORPTION
    A) SUMMARY
    1) GASTROINTESTINAL DECONTAMINATION RECOMMENDATIONS: In a systematic review of the literature, the Extracorporeal Treatments in Poisoning (EXTRIP) workgroup (includes international experts from the US, Canada, Europe, Australia, Brazil and China) concluded that patients may benefit from a single dose of activated charcoal following a recent ingestion. Currently, the use of multiple dose activated charcoal is not recommended (Ghannoum et al, 2015).
    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).
    3) The absorption of valproate 300 mg was reduced by 65% by the administration of 50 g of activated charcoal 5 minutes postingestion (Neuvonen et al, 1983).
    4) Consider a second dose of activated charcoal in a patient with rising concentrations after initial decontamination or following a large overdose of an enteric-coated or sustained-release valproate formulation.
    C) MULTIPLE DOSE ACTIVATED CHARCOAL
    1) Two case reports suggest that multiple dose activated charcoal may enhance valproate elimination in patients with large overdoses. There are no studies to determine if MDAC improves outcome or shortens the duration of toxicity after valproate overdose. MDAC has also been associated with adverse events including aspiration and bowel obstruction (Sztajnkrycer, 2002). Routine use is NOT recommended.
    2) CASE REPORT: A 15-year-old boy ingested approximately 115 250-mg valproate tablets, and was successfully treated with multiple dose activated charcoal (MDAC) (Su et al, 2002). Activated charcoal 60 g via a nasogastric tube was administered after the patient's neurological status deteriorated (i.e., delirium and pinpoint pupils). Initial serum VPA was 915 mcg/mL, and peaked two hours later at 1183 mcg/mL. The patient was given three additional doses of AC (30 g) every 6 hours. A calculated serum half-life of 6.45 hours was reported while receiving MDAC, as compared to 24.75 hours after MDAC was discontinued.
    3) CASE REPORT: A 26-month-old who ingested at least 4.5 grams (375 milligrams/kilogram) of valproate was treated with continuous nasogastric infusion of activated charcoal 3 grams/hour. Calculated elimination half life was 4.8 hours compared with and expected elimination half life of 10 to 16 hours in children taking therapeutic doses and 21 to 23 hours after overdose (Farrar et al, 1993).
    D) GASTRIC LAVAGE
    1) Gastric lavage should be reserved for patients with very large ingestions, generally 400 milligrams/kilogram or more.
    2) 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.
    3) 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.
    4) 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.
    5) 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).
    6) 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.
    E) WHOLE BOWEL IRRIGATION (WBI)
    1) Whole bowel irrigation was used in the case of a massive overdose of divalproex with prolonged absorption and a delayed peak serum concentration (1380 micrograms/milliliter 17 hours after ingestion) (Graudins & Aaron, 1996). Whole bowel irrigation may be a useful adjunct in the treatment of overdoses of enteric coated valproate formulations or patients with rising concentrations despite activated charcoal therapy.
    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.
    6.5.3) TREATMENT
    A) SUPPORT
    1) General and supportive care is indicated for the patient (Morrow & Routledge, 1989). Most patients with mild/moderate CNS depression may be safely managed with conservative, supportive respiratory and cardiovascular care. Respiratory arrest requiring artificial ventilation has been reported following severe overdose.
    2) Patients ingesting less than 400 milligrams/kilogram are not likely to develop severe toxicity (Ibister et al, 2003). Patients with large ingestions may require resuscitation and intensive supportive care
    B) 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).
    C) NALOXONE
    1) POSITIVE EFFECT: Intravenous administration of NALOXONE (0.01 milligram/kilogram and 2 milligrams, respectively in 2 cases) has reversed coma thought to be secondary to valproate ingestion in a limited number of case reports (Steiman et al, 1979; Alberto et al, 1989; Montero, 1999). It was suggested that naloxone can act by displacing GABA from its receptors and block or inhibit the effect of GABA, which was augmented by valproate (Thanacoody, 2007; Montero, 1999).
    a) ACUTE TOXICITY
    1) In addition, naloxone 2 mg was successfully used to reverse a depressed level of consciousness in a 22-year-old male whose admission valproate concentration was 180.4 micrograms/milliliter; other toxicological studies were negative (Popiel et al, 1989). Two mg naloxone was administered intravenously to a 44-year-old woman who ingested a "handful" of her valproic acid and gabapentin tablets as well as 4 mexilitine tablets and ethanol (Roberge & Francis, 2002). Within one minute the patient awoke and became conversant. A second dose was repeated in one hour due to increasing somnolence with a quick reversal of symptoms. Further support for the efficacy of naloxone in these patients has not yet appeared in the literature.
    b) CHRONIC TOXICITY
    1) A 76-year-old woman with bipolar depression, treated chronically with sodium valproate was admitted with acute confusion and tremulousness. Glasgow Coma Score decreased to 10/15 after admission. Serum valproate concentration was 848 micromol/L (normal 300 to 600 micromol/L). An intravenous bolus of 0.8 mg naloxone resulted in rapid improvement of mental status and blood pressure, and resolution of apneic episodes. A naloxone infusion was continued and symptoms resolved over 24 hours with no sequelae (Thanacoody, 2007) .
    c) NO EFFECT
    1) Naloxone 0.8 milligram intravenously was ineffective in a 40-year-old epileptic woman following a sodium valproate overdose.
    a) The reported peak valproate concentration was 18,900 micromoles/liter 6 hours after admission (Connacher et al, 1987).
    b) A 37-year-old woman ingested a massive amount of valproic acid resulting in a serum concentration of 1579 mcg/mL and profound coma. Aggressive naloxone therapy (total dose = 30 milligrams) was ineffective (Palatnick et al, 1989).
    D) LEVOCARNITINE
    1) Carnitine administration has been shown to lower serum ammonia concentrations and improve outcome in patients with valproate-induced hepatotoxicity and encephalopathy after chronic use (Ohtani et al, 1982).
    2) Carnitine supplementation should be considered in patients with elevated valproic acid concentrations and hyperammonemic encephalopathy.
    3) DOSE
    a) The Pediatric Neurology Advisory Committee recommends carnitine replacement for valproate-induced hepatotoxicity and overdose. For carnitine replacement, oral doses of 100 mg/kg/day or 2 grams/day (divided into 3 or 4 doses) should be given. In cases of overdose or valproate-induced hepatotoxicity, rescue therapy with intravenous carnitine 150 to 500 mg/kg/day (up to maximum of 3 grams/day) should be administered (Lheureux & Hantson, 2009; Raskind & El-Chaar, 2000).
    b) Based on a review of the literature, optimal dosing of L-carnitine for acute toxicity has not been established. Suggested guidelines for the use of L-carnitine following acute valproate toxicity are as follows (Russell, 2007):
    1) Acute overdose with NO evidence of hepatotoxicity: Prophylactic dose of 100 mg/kg/day IV in divided doses every 6 hours (maximum 3 g)
    2) Acute overdose and symptomatic hepatotoxicity or hyperammonemia:
    a) Loading dose: 100 mg/kg IV over 30 minutes (maximum of 6 g)
    b) Maintenance dose: 15 mg/kg every 4 hours administered over 10 to 30 minutes
    4) EFFICACY
    a) SUMMARY: Hyperammonemia may develop after acute overdose, or acute-on-therapeutic overdose, or following chronic valproic acid therapy (Lheureux & Hantson, 2009).
    b) CHRONIC: In a series of 92 patients with severe valproate induced hepatotoxicity from chronic therapy, 48% of the 42 patients treated with L-carnitine survived, compared with only 10% of those treated with aggressive supportive care (Bohan et al, 2001). Patients treated with intravenous L-carnitine and those treated early in the course of illness appeared to do best. Most of these patients appeared to be malnourished. In a review, it was noted that levocarnitine supplementation improved hyperammonemia associated with chronic valproic acid therapy; however, ammonia concentrations did not necessarily correlate with clinical symptoms (Lheureux et al, 2005).
    c) ACUTE: Carnitine has also been used in cases of valproic acid overdose (Papaseit et al, 2012; Sztajnkrycer et al, 2001; Thole et al, 2001). There are no studies demonstrating improved outcome in patients treated with L-carnitine after valproic acid overdose (Sztajnkrycer, 2002).
    1) INDICATIONS: Despite a lack of controlled studies or limited evidence supporting the use of L-carnitine (Ghannoum et al, 2015), it is suggested that L-carnitine be used to treat acute valproic acid-induced toxicity under the following circumstances (Papaseit et al, 2012; Jung et al, 2008; Lheureux & Hantson, 2009; Russell, 2007):
    1) Hepatotoxicity
    2) Hyperammonemia
    3) High risk patients; especially children or those who ingested a large amount of valproic acid
    4) Valproic acid concentration greater than or equal to 450 mg/L
    2) VALPROATE-INDUCED HYPERAMMONEMIC ENCEPHALOPATHY OCCURRING AT THERAPEUTIC LEVELS: In a small number of cases, valproate-induced hyperammonemic encephalopathy has been reported at therapeutic valproate dosages and has been successfully treated with levocarnitine. In one case, a 31-year-old man was started on valproate after developing a generalized tonic-clonic seizure and was later diagnosed with a glioma. Six days after surgery he became lethargic and his ammonia level was 204 micromol/L (normal, less than 32); valproate level was 95 ng/mL (normal, 50 to 100). He received levocarnitine at 100 mg/kg over 20 minutes, followed by 50 mg/kg every 8 hours for 24 hours. Within 24 hours, his neurologic status had improved along with a decrease in his ammonia levels (Rigamonti et al, 2014).
    3) PEDIATRIC
    a) CASE REPORT: L-carnitine was administered to a 16-month-old child who ingested approximately 4000 mg of valproate (Ishidura et al, 1996). L-carnitine administration was associated with increased urinary elimination of beta oxidation metabolites of valproate and decreased urinary elimination of omega and omega-1-oxidation metabolites and 4-en-valproate, a potential hepatotoxin. The child recovered without sequelae.
    4) ADULT
    a) CASE REPORT: L-carnitine (1 g 3 times per day) IV was administered for 24 hours in a 30-year-old man who intentionally ingested an estimated 35 g of extended-release valproate (VPA) 15 hours prior to admission. An initial VPA plasma concentration was 391 mcg/mL with clinical evidence of hyperammonemic encephalopathy (ie, lethargy, ammonia concentration: 256 micromoles/L) and no signs of liver failure. His level of consciousness improved with decreasing concentrations of both VPA (to therapeutic range) and ammonia concentrations (124 to 128 micromoles/L; normal range: 10 to 47) approximately 13 hours later (28 hours postingestion) (Papaseit et al, 2012).
    b) CASE REPORT: Levocarnitine (100 mg/kg) was given IV to a 51-year-old woman who presented 10 days after initiation of valproate (10 mg/kg/day) therapy with severe hyperammonemic encephalopathy. Blood arterial ammonia concentration on presentation was 234 s/L, which decreased over a 10 hour period following intravenous L-carnitine to 35 mmol/L. Her neurologic condition improved within 18 hours (Borbath et al, 2000).
    c) CASE REPORT: Following the ingestion of an unknown quantity of valproic acid, a 24-year-old man was treated for an elevated serum ammonia concentration (176 mEq/dL) with L-carnitine 25 mg/kg IV every 6 hours for about 30 hours, when his ammonia concentration dropped to 44 mEq/dL. At 46 hours postingestion the patient was asymptomatic with normal vital signs (Thole et al, 2001).
    d) CASE REPORT: A single bolus dose of L-carnitine (20 mg/kg IV) was given postdialysis to a 41-year-old man, after reportedly ingesting 50,000 mg of valproic acid, for prophylaxis against hepatotoxicity and hyperammonemia (Guillaume et al, 2004).
    5) SAFETY
    a) A retrospective review of patients treated with L-carnitine after acute valproic acid (VPA) overdose and elevated ammonia concentrations was conducted. Fifty-five doses of L-carnitine were administered to 19 patients who had isolated VPA ingestions, and 196 doses of L-carnitine were administered to patients with mixed overdoses that included VPA. Of the 251 doses administered, there were no adverse events (defined as an allergic reaction or hypotension) reported (LoVecchio et al, 2005).

Enhanced Elimination

    A) SUMMARY
    1) Hemodialysis, hemoperfusion and hemofiltration enhance the clearance of valproic acid after severe overdose, because protein binding of the drug is saturated; however, it is not clear if these modalities affect outcome after overdose. They should be considered for patients with severe intoxication and rising concentrations despite supportive care. Because hemodialysis is most readily available in most practice settings, it is the method used most often for sever valproate poisoning.
    2) In addition, clinical improvement has been noted to occur faster in patients that receive extracorporeal treatment within the first 24 hours of admission (Ghannoum et al, 2015).
    B) HEMODIALYSIS
    1) SUMMARY
    a) THERAPEUTIC CONCENTRATIONS: Hemodialysis is NOT very effective for removal of therapeutic concentrations of valproic acid (VPA) since VPA is highly protein bound, but it is effective for VPA removal following overdose because the fraction of drug not bound to protein is high in this setting and hemodialysis (or tandem hemodialysis and hemoperfusion) may result in significant drug removal (Prod Info DEPAKENE(R) oral capsules, oral solution, 2007; Meek et al, 2004; Kay et al, 2003; Kroll & Nand, 2002; Hicks & McFarlane, 2001). Hemodialysis has been reported as effective in removing valproic acid (VPA) from uremic patients (Marbury et al, 1980).
    b) OVERDOSE: In overdose, saturated protein binding results in an increased fraction of unbound valproic acid, resulting in more effective removal by hemodialysis (Ghannoum et al, 2015; Al Aly et al, 2005; Guillaume et al, 2004).
    c) Most patients can be managed with supportive care alone. Hemodialysis should be considered in patients with hemodynamic instability. Some authors suggest that markedly elevated valproate (greater than 950 mcg/mL) may be an indication for hemodialysis or charcoal hemoperfusion. However, in a retrospective review of 50 cases of overdose, there were 4 patients who had documented peak valproate concentrations greater than 950 mcg/mL and did not have persistent hemodynamic instability or metabolic acidosis (Graeme et al, 1999). None received hemodialysis or hemoperfusion, and all recovered without sequelae.
    d) The percent of total serum valproate that is not protein bound increases substantially in severe overdose, from 11% free drug at a level of 79 mcg/mL to 71% free drug at a concentration of 451 mcg/mL in one case report (Farrar et al, 1993). This suggests that hemodialysis might clear a significant amount of drug in severe overdose. Calculated VPA half-life was reported to be 2.48 hours during hemodialysis and 16.64 hours after dialysis in a VPA overdose patient. In the same patient, serum VPA concentration dropped from 752.7 mcg/mL to 269 mcg/mL after 3.5 hours of hemodialysis (Ruskosky et al, 1999).
    e) High efficiency hemodialysis, which can be performed at the bedside and requires lower blood flow rates, may be used for severe valproic acid intoxications when high-flux hemodialysis is not readily available (Sharma et al, 2001).
    2) SYSTEMATIC REVIEW AND RECOMMENDATIONS
    a) In a systematic review of the literature that included 79 articles, including 1 observational study, 1 uncontrolled cohort with aggregate results, 70 case reports or case series and 7 pharmacokinetic studies, the Extracorporeal Treatments in Poisoning (EXTRIP) workgroup (included international experts from the US, Canada, Europe, Australia, Brazil and China) concluded that valproic acid is moderately dialyzable (moderate level of evidence (grade B)). Of note, most reported poisonings involved regular-release formulations of valproic acid. Patient history alone was considered an unreliable indicator by the workgroup to begin ECTR considering the potential risks of treatment. Close monitoring was indicated to assess the patient and to determine if ECTR was needed. Despite a low quality of evidence for all recommendations, extracorporeal treatment was recommended in the setting of severe valproic acid poisoning. Intermittent hemodialysis is the preferred method. If unavailable, intermittent hemoperfusion or continuous renal replacement therapy are appropriate alternatives (Ghannoum et al, 2015).
    b) ECTR is recommended if ANY of the following are present (Ghannoum et al, 2015):
    1) Valproic acid concentration of greater than 1300 mg/L (9000 micromol/L)
    2) Valproic acid poisoning-associated cerebral edema or shock
    c) ECTR is suggested if ANY of the following are present (Ghannoum et al, 2015):
    1) Valproic acid concentration of greater than 900 mg/L (6250 micromol/L)
    2) Coma or respiratory depression requiring mechanical ventilation
    3) Acute hyperammonemia
    4) pH less than or equal to 7.10
    d) DISCONTINUATION OF THERAPY: Hemodialysis should be discontinued when the patient is clinically improving (ie correction of severe manifestations including coma, acidemia, respiratory depression and hemodynamic instability) or the serum valproic acid concentration is between 50 and 100 mg/L (therapeutic concentration). In the presence of a mixed ingestion that may not be removed by ECTR, further evaluation and assessment of the patient may be indicated (Ghannoum et al, 2015).
    e) REBOUND: Valproic acid concentrations may rebound after a session of ECTR and it is likely due to a redistribution from deeper compartments in the plasma. Although this event has been reported, it did not produce significant changes (ie, clinical deterioration) in most patients. If necessary, a second session of ECTR may be indicated to correct the effects of rebound (Ghannoum et al, 2015).
    3) CASE SERIES
    a) CASE SERIES: In a series of 32 patients with valproic acid overdose and peak serum concentrations greater than 100 mcg/mL reported to a single poison center, 6 were treated with hemodialysis. Valproic acid elimination was increased about ten fold during hemodialysis. Half-life during hemodialysis averaged 1.9 hours in the three patients in whom there was sufficient data to calculate, compared with 29.6 hours after hemodialysis. In the one patient treated with combined hemodialysis and hemoperfusion half life during the procedure was 4.3 hours. Complications included a seizure in a patient with a seizure disorder, and thrombocytopenia in the patient treated with hemoperfusion (Singh et al, 2004).
    b) CASE SERIES: In a series of 6 patients with valproic acid intoxication, the half-life of valproic acid during hemodialysis was 2.5 to 5.2 hours, compared with half lives of 10.2 to 60.4 hours prior to dialysis and 10.3 to 62.7 hours after dialysis (Eyer et al, 2005).
    4) CASE REPORTS
    a) CASE REPORT: A 48-year-old unconscious female with a peak serum valproate concentration of 1402 mg/L (dose ingested 48 g) was initially started on continuous veno-venous hemodiafiltration (CVVHDF) with a high-flux membrane for 8 hours with no change in neuro status, and minimal change in valproate clearance. Estimated elimination half-life during hemodialysis was 2.2 hours, compared to 16.1 hours during CVVHDR and 28.5 hours without treatment. Plasma clearance was 90 mL/min during the first hour of dialysis, and 56 mL/min during the fourth hour. She had a complete recovery (Kay et al, 2003).
    b) CASE REPORT: Hemodialysis was used to treat a 44-year-old woman with severe valproic acid and trazodone overdose complicated by seizures, myoclonic jerking, coma and hypotension (Williams & Clark, 1995). Initial valproate concentration was 995 mcg/mL falling to 949 mcg/mL 3.5 hours later. Within 30 minutes of initiating hemodialysis she was able to follow commands and after 4 hours of hemodialysis valproic acid concentration was 113 mcg/mL. Dialysis clearance of valproic acid was not determined.
    c) CASE REPORT: Hemodialysis was used successfully for treatment of valproic acid (VPA) overdose (approximately 19 grams) in a 43-year-old woman (Johnson et al, 1999a). Renal and hepatic functions were normal. Four hours after hospital presentation hemodialysis was begun. Serum VPA concentration decreased from 940 mcg/mL to 164 mcg/mL after 6 hours of hemodialysis. Prior to hemodialysis VPA half-life was 7.2 hr and after hemodialysis VPA half-life was 2.4 hr. Valproate clearance was 76 to 100 milliliters/minute during dialysis.
    d) CASE REPORT: High-flux hemodialysis without charcoal hemoperfusion was successfully used in a case of valproic acid overdose in a 25-year-old female. The amount ingested was unknown, and resulted in coma, hypotension and lactic acidosis, with serum a concentration greater than 1200 mcg/mL. During 4-hour high-flux hemodialysis, valproic acid half-life was calculated as 2.7 hours, compared to 23.4 hours post dialysis (Kane et al, 2000).
    e) CASE REPORT: High-flux hemodialysis was used in a patient with acute-on-chronic renal failure (previous history of nephrosclerosis, hypertension, SLE and thrombocytopenia) and severe valproic acid poisoning (coma, hypotension, valproic acid concentration 650 mcg/mL). Serum valproate and ammonia concentrations declined rapidly. Dialyzer clearance was 121 mL/min during the first dialysis and 56 mL/min during the second. His course was complicated by thrombocytopenia and aspiration pneumonia but he recovered (Kielstein et al, 2003).
    f) CASE REPORT: A 41-year-old man was treated with hemodialysis after reportedly ingesting 50,000 mg of valproic acid (100 500-mg extended release tablets) and subsequently developing lethargy and tachycardia. His plasma valproic acid (VPA) concentration on admission was 789 mg/L (therapeutic range 50 to 100 mg/L). Two hours later, his plasma VPA concentration increased to 1150 mg/L. Approximately 6 hours post-admission, hemodialysis was initiated, with a mean blood flow rate of 200 mL/min and a dialysate flow rate of 500 mL/min. Postdialysis, the patient was given a single bolus dose of L-carnitine (20 mg/kg) intravenously for prophylaxis against hepatotoxicity and hyperammonemia. The patient completely recovered and was discharged 48 hours later (Guillaume et al, 2004).
    1) The calculated plasma half-life of VPA was approximately 4 hours during hemodialysis and 15.8 hours postdialysis. Prior to hemodialysis, the concentration of unbound VPA increased from 400 mg/L to 781 mg/L, resulting in a protein binding of approximately 35% (normal is greater than 80%). Twenty-eight hours postdialysis, the protein binding normalized (Guillaume et al, 2004).
    C) SUSTAINED LOW-EFFICIENCY DIALYSIS WITH FILTRATION (SLEDD-f)
    1) CASE REPORT: A 39-year-old woman became rapidly comatose with metabolic acidosis and refractory hypotension after ingesting 100 (1.2 g/kg) enteric coated sodium valproate tablets. Approximately, 18 hours after ingestion a peak valproate concentration of 12,488 micromol/L was observed. The patient was treated with slow low-efficient daily diafiltration using a Fresenius 408S ArRT-Plus online hemodiafiltration system with a Fresenius AV600S standard hollow-fiber polysulfone membrane. The plasma valproate concentration fell rapidly over 16 hours of therapy, along with clinical and laboratory improvement. No rebound phenomenon occurred. The patient was discharged 3 days after dialysis with no permanent sequelae (Khan et al, 2008). The authors suggest that this form of hybrid dialysis provides the benefit of continuous renal replacement therapy and intermittent hemodialysis.
    D) HEMOPERFUSION
    1) SUMMARY: Intermittent hemoperfusion can be useful to enhance elimination after a severe valproate overdose, however it is often less available than hemodialysis in most healthcare settings and can produce more complications that can be life threatening. It can also produce hypocalcemia or thrombocytopenia (Ghannoum et al, 2015).
    2) RECOMMENDATIONS: Intermittent hemoperfusion is recommended as an alternative therapy when intermittent hemodialysis is not available (Ghannoum et al, 2015).
    3) MECHANISM: Hemoperfusion may be effective in removing valproic acid (VPA) following overdose. VPA has a relatively low volume of distribution and small molecular weight (144 daltons), and hemoperfusion with charcoal is effective in removing protein bound drugs circulation (Jung et al, 2008).
    4) CASE REPORT: A 23-year-old woman intentionally ingested approximately 24 g of valproic acid (VPA) and was found stuporous. A serum VPA concentration of 1159.4 mcg/mL (range: 50-100 mcg/mL) was reported on admission. The patient underwent three courses of hemoperfusion with activated charcoal for 6 hours (serum concentration decreased to 628.9 mcg/mL after the first session of hemoperfusion) with a normal serum level by day 3 and improving neurologic function. The patient was also treated with L-carnitine for hyperammonemia and metabolic acidosis. No permanent sequelae was reported (Jung et al, 2008).
    5) CASE REPORT: Activated charcoal hemoperfusion was used to remove sodium valproate from a 38-year-old man with a documented sodium valproate blood concentration of 1080 mcg/mL. Activated charcoal hemoperfusion was performed for 8 hours initially and again later for 5 hours at 150 to 200 mL/min (18 hours after admission). When the patient awoke, his hematologic, liver enzymes and kidney function tests were all within normal limits (Van der Merwe et al, 1985) .
    6) CASE REPORT/MIXED INGESTION: A 16-year-old girl intentionally ingested an estimated 16.4 g of carbamazepine (CBZ) (extended release tablets) and 14.5 g of valproic acid (extended release tablets) approximately 3 hours prior to admission. She was found unconscious by her parents. Five hours after ingestion, her CBZ level was 43.1 mcg/mL and her valproic acid level was 111.5 mcg/mL (peak, 283 mcg/mL). Activated charcoal was given every 6 hours for the first 24 hours along with enteral polyethylene glycol (Macrogol) to accelerate bowel elimination. However, 15 hours after ingestion, no neurologic improvement was observed and she was started on continuous charcoal hemoperfusion for 4 hours with significant laboratory improvement in drug levels, but a decrease in her platelet count (167,000/mcL to 45,000/mcL) occurred. Continuous venovenous hemodiafiltration (CVVHDF) was then performed for 48 hours. At the end of the treatment period, CBZ level was 17.8 mcg/l and valproic acid was 119 mcg/L and both were continuing to decrease. Ninety hours after ingestion, the patient became more alert and was successfully extubated at 120 hours. She continued to do well with no neurologic deficits and was discharged to home on day 8 with ongoing mental health support (Moinho et al, 2014).
    7) CASE REPORT: Hemoperfusion was used to treat a 32-year-old woman with severe valproic acid and chlorpheniramine overdose, complicated by obtundation and hepatic injury. Peak valproic acid level was 1,380 mcg/mL 17 hours post ingestion. The initial extraction ratio of the hemoperfusion cartridge was 0.54 with a plasma clearance of 55 mL/min. Half-life during 3 hours of hemoperfusion was 3 hours compared with 4.8 hours after hemoperfusion with multiple dose activated charcoal (Graudins & Aaron, 1996).
    8) CASE REPORT: Serum valproate levels of 1066 mg/L (2.2 hours postingestion) and 1562 mg/L (5 hours postingestion) were reported following an overdose of 22.5 g in a 21-year-old woman. Following 2 hemoperfusion sessions, resulting valproic acid levels of 835 and 413 mg/L after 4 and 8 hours, respectively, were reported (von Bardeleben et al, 1998).
    9) CASE REPORT: A 50-year-old woman had a valproic acid level of 491 mcg/mL 5 to 6 hours after an ingestion of an unknown quantity. Hemoperfusion was initiated at a flow rate of 400 mL/min (standard for hemodialysis) instead of a typical rate of 150 to 250 mL/min. Within 2 hours the patient suffered severe intravascular hemolysis and renal failure. The patient required a total of 23 days of dialysis before recovery of renal function (Rahman et al, 2006).
    E) COMBINED HEMOPERFUSION/HEMODIALYSIS
    1) CASE REPORT: Simultaneous "in series" hemoperfusion and hemodialysis were used in a case of acute valproate toxicity (peak level 1,262 mcg/mL) (Tank & Palmer, 1993). Plasma valproate clearance was 91 mL/minute 20 minutes into the procedure and fell to 34 mL/minute at 4 hours. Half-life during the procedure was 1.7 hours compared with a half life of 8 hours prior to the procedure.
    2) CASE REPORT: A 20-year-old woman presented with coma and a peak serum valproate level of 2120 mcg/mL at 8.5 hours postingestion. Hemodialysis was started 10.5 hours postingestion on a CDAK 1.3 square meter artificial kidney (blood flow was 150 to 200 mL/min; dialysate flow was 600 mL). Dialysis continued for 8 hours followed by 4 hours of activated charcoal hemoperfusion. The patient woke up gradually over the next 3 days (Mortensen et al, 1983).
    3) CASE REPORT: Serial hemodialysis and hemoperfusion was used in a 27-year-old man following a massive overdose (Franssen et al, 1999). Reported VPA clearances during the treatments were 80 mL/min (hemodialysis); 40 mL/min (hemoperfusion by charcoal); and 80 mL/min (hemoperfusion by resin only in first hour). Because saturated protein binding resulted in an increased fraction of unbound valproic acid, hemodialysis was more effective than hemoperfusion in this case.
    4) CASE REPORT: An "in series" treatment of hemodialysis and hemoperfusion, followed by continuous venovenous hemodiafiltration (CVVHDF), were performed on a 35-year-old man who reportedly ingested 30 g (120 capsules) of valproic acid (VPA). His peak VPA level, at the initiation of hemodialysis and hemoperfusion, was 573 mcg/mL. During the "in series" treatment, the blood flow was maintained at 240 to 330 mL/min and the dialysate flow was maintained at 500 mL/min; whole blood valproate clearance ranged from 87 to 160 ml/min and plasma clearance ranged from 42 to 88 mL/min during treatment. At the end of the 4-hour treatment, the VPA level had decreased to 268 mcg/mL, but 3 hours after the hemodialysis and hemoperfusion treatment, the VPA level increased to 343 mcg/mL. Due to the post-dialytic rebound of his VPA level, CVVHDF was initiated. The dialysate flow rate was maintained at 1000 mL/hour and the filtration rate was 1000 mL/hour. At the end of the treatment, 18 hours later, the patient's VPA level was 118 mcg/mL (Al Aly et al, 2005).
    F) CONTINUOUS RENAL REPLACEMENT THERAPY
    1) RECOMMENDATIONS: Intermittent continuous renal replacement therapy (CRRT) is recommended as an alternative therapy when intermittent hemodialysis is not available (Ghannoum et al, 2015). It was found to be inferior to the high-efficiency of intermittent dialysis; however, it may be considered when technical or logistic reasons do not allow for the use of hemodialysis. CRRT has lower clearance rates and clinical improvement has been shown to be slower compared to hemodialysis or hemoperfusion (Ghannoum et al, 2015).
    G) HEMOFILTRATION
    1) Several cases have been reported concerning the use of combinations of multiple-dose activated charcoal, forced diuresis and continuous arteriovenous hemofiltration (CAVH) following severe valproic acid intoxications. CAVH is recommended when imminent hemodynamic instability is present. It has been hypothesized that efficient clearance of valproic acid metabolites rather than direct clearance of the drug occurs (van Keulen et al, 2001).
    2) Continuous arteriovenous hemofiltration (CAVH) was used to treat a comatose 15-year-old boy after an ingestion of 30 g of slow release valproic acid (VPA) (van Keulen et al, 2001). During CAVH, plasma VPA concentration decreased from 350 mg/L to 68 mg/L over 17 hours. VPA half-life during CAVH was calculated as 7.3 hours, which increased to >300 hours after CAVH. The authors concluded that CAVH was superior to endogenous clearance, but less effective than clearance via hemoperfusion, as reported in the literature.
    3) Single, conventional hemodiafiltration (HDF) without hemoperfusion was successfully used to treat a severe acute valproic acid ingestion (160 grams). Following 3 hours of HDF, valproic serum levels decreased from 2493 to 821 mg/L, with a rebound of 16% after one hour. Valproic acid clearance before HDF was 6.2 mL/min, during HDF was 93 mL/min, and after HDF was 6.9 mL/min (Minari et al, 2002).
    4) CASE REPORT/MIXED INGESTION: A 16-year-old girl intentionally ingested an estimated 16.4 g of carbamazepine (CBZ) (extended release tablets) and 14.5 g of valproic acid (extended release tablets) approximately 3 hours prior to admission. She was found unconscious by her parents. Five hours after ingestion, her CBZ level was 43.1 mcg/mL and her valproic acid level was 111.5 mcg/mL (peak, 283 mcg/mL). Activated charcoal was given every 6 hours for the first 24 hours along with enteral polyethylene glycol (Macrogol) to accelerate bowel elimination. However, 15 hours after ingestion, no neurologic improvement was observed and she was started on continuous charcoal hemoperfusion for 4 hours with significant laboratory improvement in drug levels, but a decrease in her platelet count (167,000/mcL to 45,000/mcL) occurred. Continuous venovenous hemodiafiltration (CVVHDF) was then performed for 48 hours. At the end of the treatment period, CBZ level was 17.8 mcg/l and valproic acid was 119 mcg/L and both were continuing to decrease. Ninety hours after ingestion, the patient became more alert and was successfully extubated at 120 hours. She continued to do well with no neurologic deficits and was discharged to home on day 8 with ongoing mental health support (Moinho et al, 2014).
    5) LACK OF BENEFIT: A 20-year-old man with a past history of head injury, epilepsy, and depression became obtunded following the ingestion of 40 g of sodium valproate. Serum valproic acid (VPA) reached 1136 mg/L within 24 hours of exposure. Continuous veno-venous hemodialfiltration (CVVHDF) was started, and within 4 hours the VPA was less than 500 mg/L. However, the patient remained unconscious and a head CT revealed cerebral edema (CT obtained approximately 36 hours earlier had been normal) . Mental status slowly improved and by day 7 a repeat head CT showed a gradual reduction in cerebral edema. By day 17, the patient was transferred from ICU with no evidence of neurologic deficits. The authors concluded that cerebral edema was not directly related to serum VPA levels and in this setting CVVHDF did not provide any clinical benefit (Field & Daly, 2002).
    H) MULTIPLE DOSE ACTIVATED CHARCOAL
    1) SUMMARY
    a) GASTROINTESTINAL DECONTAMINATION RECOMMENDATIONS: In a systematic review of the literature that included 79 articles, including 1 observational study, 1 uncontrolled cohort with aggregate results, 70 case reports or case series and 7 pharmacokinetic studies, the Extracorporeal Treatments in Poisoning (EXTRIP) workgroup (includes international experts from the US, Canada, Europe, Australia, Brazil and China) concluded that patients may benefit from a single dose of activated charcoal following a recent ingestion. Currently, the use of multiple dose activated charcoal is NOT recommended (Ghannoum et al, 2015).
    2) CASE REPORTS
    a) Two case reports suggest that multiple dose activated charcoal may enhance valproate elimination in patients with large overdoses. There are no studies to determine if MDAC improves outcome or shortens the duration of toxicity after valproate overdose. MDAC has also been associated with adverse events including aspiration and bowel obstruction (Sztajnkrycer, 2002). Routine use is not recommended.
    b) CASE REPORT: A 15-year-old boy ingested approximately 115 250-mg valproate tablets, and was treated with multiple dose activated charcoal (MDAC) (Su et al, 2002). Activated charcoal 60 g via a nasogastric tube was administered after the patient's neurological status deteriorated (i.e., delirium and pinpoint pupils). Initial serum VPA was 915 mcg/mL, and peaked two hours later at 1183 mcg/mL. The patient was given three additional doses of AC (30 g) every 6 hours. A calculated serum half-life of 6.45 hours was reported while receiving MDAC, as compared to 24.75 hours after MDAC was discontinued.
    c) CASE REPORT: A 26-month-old who ingested at least 4.5 g (375 mg/kg) of valproate was treated with continuous nasogastric infusion of activated charcoal 3 grams/hour. Calculated elimination half-life was 4.8 hours compared with and expected elimination half-life of 10 to 16 hours in children taking therapeutic doses and 21 to 23 hours after overdose (Farrar et al, 1993).
    3) LACK OF EFFECT
    a) In healthy volunteers, multiple dose activated charcoal had NO effect on the elimination rate after therapeutic doses of valproic acid (Al-Shareef et al, 1997).
    b) ANIMAL DATA: In a porcine model, multiple dose activated charcoal did not increase the clearance of intravenously administered valproic acid (Chuyka et al, 1995).

Case Reports

    A) ADULT
    1) CEREBRAL INFARCT: A 29-year-old man with bipolar affective disorder sustained permanent injury when he developed cerebral edema, herniation, and infarct after intentionally ingesting 4 g of valproic acid (VPA) and 75 mg of diazepam (10 hours prior to ingesting VPA). He presented to the emergency department 5 hours after ingestion of VPA with agitation, an unsteady gait, and vomiting. At that time, his vital signs and neurologic examination were normal. Initial lab analyses showed a VPA level of 5469 micromol/L (therapeutic: 350 to 700 micromol/L), serum ammonia of 348 micromol/L (normal: 0 to 50 micromol/L), and a pH of 7.34. CT scan confirmed diffuse cerebral edema with early herniation. Nine hours after ingestion, his Glasgow coma scale score deteriorated to 8/15. He was intubated, gastric contents were aspirated with a tube, and he was treated with a single 50 g dose of activated charcoal followed by 4 doses of L-carnitine (900 mg each, 1 hour apart), and mannitol. Thirty-eight hours after ingestion, VPA levels were therapeutic. A dilated right pupil, absent right papillary reflex, reduced visual acuity with skew deviation of the right eye, and right-sided weakness were observed on day 5. At that point, MRI findings confirmed cerebral infarct and ECG findings were consistent with encephalopathy. Ammonia levels normalized on day 6 and on day 8 he was extubated. After 150 days of hospitalization including extensive rehabilitation he was discharged to an assisted living facility with residual right-sided weakness and a modified Rankin scale score of 3 (moderate disability; requiring some help, but able to walk without assistance) (Rupasinghe & Jasinarachchi, 2011; van Swieten et al, 1988).
    2) A 22-year-old man presented somnolent with depressed motor and respiratory activity following an ingestion of an unknown substance. Gag reflex was absent, breath sounds were diminished, and pupils were pinpoint and nonreactive. An IV of normal saline was started and naloxone 2 mg was administered by IV push. Approximately 1 minute after naloxone, before administration of any other medication, the patient became alert, responsive, and talking. He admitted taking only valproic acid 250 mg, about 20 tablets six hours prior to presentation. His initial valproic acid concentration was 180.4 mcg/mL (Alberto et al, 1989).
    B) PEDIATRIC
    1) FATALITY: A 20-month-old boy died from valproic acid overdose. He ingested 15 grams (12 grams reportedly absorbed). The boy was comatose and developed bronchopneumonia with high fever; death occurred from cardiorespiratory failure 46 hours following ingestion. Peak valproic acid concentrations were 1061 mcg/mL 3 hours postingestion; 15 minutes prior to death, concentrations were 187 mcg/mL with a normal half-life (16 hours). The most prominent findings on autopsy were acute hypoxic damage of the myocardium, kidneys and vulnerable cerebral neurons, as well as substantial cerebral edema (Schnabel et al, 1984).
    2) A 2-month-old girl with epilepsy presented with generalized seizures, progressive coma, and anuria associated with a serum sodium valproate concentration of 1299 mcg/mL. Both parents had epilepsy and apparently accidentally overdosed this child. Two days prior to admission the toddler had a serum sodium valproate concentration of 53 mcg/mL. She was treated with hemoperfusion, serially connected with hemodialysis during 3 hours. On hospital day 2, peritoneal dialysis was started for persistent anuria and electrolyte disturbances and was maintained for 5 days. On the 5th day of hospitalization, a urine sample was obtained and found to contain myoglobin. Phenobarbital was given as an antiepileptic drug, beginning the 5th day of hospitalization (Roodhooft et al, 1990).
    3) Eeg-Olofsson & Lindskog (1982) reported a 5-year-old severely mentally retarded boy who received valproate monotherapy in a daily dose of 40 mg/kg for treatment of minor motor seizures. Normal plasma concentrations fluctuated around 350 mcmol/L (50 mcg/mL). Presentation on admission revealed a somnolent boy with generalized myoclonic seizures which were treated with clonazepam. Very high plasma concentrations of valproate were noted (8130 mcmol/L) (1172 mcg/mL) on day one. In addition, elevated concentrations of urine metabolites of valproate, high serum concentrations of sodium, acidosis, and hypocalcemia were noted. Within 24 hours, the patient gradually awoke and was in his usual state. By day three, valproate concentrations had dropped to 437 mcmol/L (63 mcg/mL).
    4) TEENAGER: Only minor CNS toxicity developed following overdose of 30 grams valproic acid in a 16-year-old girl. The patient had been taking the drug for 1.5 years. Five hours after ingestion the patient was admitted stuporous; physical examination was otherwise normal. The patients was fully awake 12 hours after ingestion and remained in the hospital for 3 days without complications. Serum concentration was 4925 micromoles/L (710 mcg/mL) on admission (6 hours postingestion). Slight liver function abnormalities were reported, but were not considered clinically significant (Karlsen et al, 1983).

Summary

    A) TOXICITY: MILD: Greater than 200 mg/kg risk of CNS depression; MODERATE: Greater than 400 mg/kg risk of multiogran system toxicities; and SEVERE: Greater than 750 mg/kg potentially lethal.
    B) THERAPEUTIC DOSE: ADULTS and CHILDREN 10 years of age and older: Initial dose: 10 to 15 mg/kg/day, titrate as needed; maximum dose: 60 mg/kg/day.

Therapeutic Dose

    7.2.1) ADULT
    A) COMPLEX PARTIAL SEIZURES
    1) MONOTHERAPY: INITIAL DOSE: 10 to 15 mg/kg/day, increasing by 5 to 10 mg/kg/week until optimal clinical response or obtain serum concentrations to determine if therapeutic levels (therapeutic range, 50 to 100 mcg/mL) have been reached. MAX: 60 mg/kg; if dose exceeds 250 mg, give in divided doses (Prod Info DEPAKENE oral capsules, oral solution, 2014; Prod Info STAVZOR(R) oral delayed release capsules, 2014).
    2) ADJUNCTIVE THERAPY: May be added to a patient's regimen starting at 10 to 15 mg/kg/day with the dosage increased by 5 to 10 mg/kg/week to achieve an optimal response. MAX: 60 mg/kg. If the total daily dose exceeds 250 mg, it should be given in divided doses (Prod Info DEPAKENE oral capsules, oral solution, 2014; Prod Info STAVZOR(R) oral delayed release capsules, 2014)
    B) MANIA
    1) 750 mg daily in divided doses; MAX: 60 mg/kg/day (Prod Info STAVZOR(R) oral delayed release capsules, 2014).
    C) MIGRAINE
    1) INITIAL DOSE: 250 mg twice daily; MAX: 1000 mg/day (Prod Info STAVZOR(R) oral delayed release capsules, 2014).
    7.2.2) PEDIATRIC
    A) INTRAVENOUS
    1) STATUS EPILEPTICUS, REFRACTORY
    a) LOADING DOSE: 20 to 30 mg/kg IV bolus over 5 to 10 minutes (Agarwal et al, 2007; Mehta et al, 2007; Misra et al, 2006; Yu et al, 2003); a second bolus of 10 mg/kg has been given 10 to 15 minutes after the first bolus in patients with continued seizure activity (Mehta et al, 2007; Uberall et al, 2000).
    b) Rapid IV infusions of 2 to 6 mg/kg/min have been given safely in children (Morton et al, 2007; Yu et al, 2003; Ramsay et al, 2003; Venkataraman & Wheless, 1999).
    c) MAINTENANCE INFUSION: Maintenance infusions of 1 to 6 mg/kg/hour have been used to sustain a seizure-free period (Mehta et al, 2007; Uberall et al, 2000) or for continued seizure control (Hovinga et al, 1999). Following continuous infusion, conversion to equivalent scheduled IV or oral maintenance doses can be initiated (Mehta et al, 2007; Hovinga et al, 1999).
    2) EPILEPSY (IF UNABLE TO TAKE ORAL)
    a) Initial, 10 to 15 mg/kg/day IV divided every 6 hours, increasing at 1-week intervals by 5 to 10 mg/kg/day until optimal control achieved. Maximum recommended dose is 60 mg/kg/day (Prod Info DEPACON(R) IV injection, 2006).
    B) ORAL
    1) COMPLEX PARTIAL SEIZURES
    a) CHILDREN: 10 YEARS OF AGE AND OLDER
    1) MONOTHERAPY: INITIAL DOSE: 10 to 15 mg/kg/day, increasing by 5 to 10 mg/kg/week until optimal clinical response or obtain serum concentrations to determine if therapeutic levels (therapeutic range, 50 to 100 mcg/mL) have been reached. MAX: 60 mg/kg; if dose exceeds 250 mg, give in divided doses (Prod Info DEPAKENE oral capsules, oral solution, 2014; Prod Info STAVZOR(R) oral delayed release capsules, 2014).
    2) ADJUNCTIVE THERAPY: May be added to a patient's regimen starting at 10 to 15 mg/kg/day with the dosage increased by 5 to 10 mg/kg/week to achieve an optimal response. MAX: 60 mg/kg. If the total daily dose exceeds 250 mg, it should be given in divided doses (Prod Info DEPAKENE oral capsules, oral solution, 2014; Prod Info STAVZOR(R) oral delayed release capsules, 2014).
    2) BIPOLAR DISORDER
    a) 5 YEARS AND OLDER: INITIAL: 15 to 20 mg/kg/day (maximum 759 mg) orally in 2 to 3 divided doses, titrated to serum trough level of 50 to 125 mcg/mL based on clinical effect and tolerability (Azorin & Findling, 2007; DelBello et al, 2006; Pavuluri et al, 2005; Findling et al, 2005; Danielyan & Kowatch, 2005; Wagner et al, 2002).
    b) In a randomized, placebo-controlled, double-blind, clinical trial (n=144), extended-release divalproex was not significantly better than placebo in improving Young Mania Rating Scale (YMRS) scores from baseline (Wagner et al, 2009).
    3) MIGRAINE PROPHYLAXIS
    a) 3 TO 17 YEARS OF AGE: 10 to 30 mg/kg/day orally in 2 divided doses (Bidabadi & Mashouf, 2010; Ashrafi et al, 2005). One clinical trial initiated dosing at 30 mg/kg/day and decreased the dose to 15 mg/kg/day after the first month of therapy (Bidabadi & Mashouf, 2010). Another clinical trial initiated dosing at 10 mg/kg/day and slowly increased up to 40 mg/kg/day based on tolerance and efficacy (Ashrafi et al, 2005). Maximum doses were not reported.

Minimum Lethal Exposure

    A) CASE REPORTS
    1) ADULT
    a) A 23-year-old man died following initiation of valproic acid therapy at a dose of 750 mg by mouth three times a day. The maximum daily dose recommended by the manufacturer for this patient should have been 960 mg/day (15 mg/kg/day). He received 2250 mg/day for 5 days (11.25 g over 5 days) (Tift, 1980).
    b) A 24-year-old woman died approximately 48 hours after ingesting 350 g of Depakote(R). Upon arrival to ED, 3 hours after ingestions, the patient received gastric lavage and activated charcoal and was drowsy for 6 hours. Eleven hours postingestion she became unresponsive, requiring mechanical ventilation and dopamine for hypotension. Despite aggressive care the patient died (Christianson et al, 2001).
    c) MIXED INGESTION: After ingesting 20 g (333 mg/kg) of valproic acid extended release, 60 mg of risperidone, and 3 g of venlafaxine extended release, a 19-year-old man developed hyperammonemia (peak ammonia concentration 1,191 mcg/dL at 65 hours postingestion). The valproic acid concentration peaked at 305.4 mcg/dL. Despite treatment with activated charcoal with sorbitol, and lactulose, the patient's condition deteriorated. CT scan revealed cerebral edema and possible tentorial herniation. At approximately 120 hours after ingestion, cerebral blood flow studies showed an absence of cerebral perfusion, and the patient was pronounced brain dead (Camilleri et al, 2005).
    2) INFANT
    a) A 26-day-old full term infant (4500 g) was inadvertently given 300 mg valproate (intended dose 30 mg/kg twice daily); symptoms began within 12 hours. Within 36 hours, the pupils were fixed and dilated. A CT scan of the head showed cerebral edema, and a serum ammonia of 279 mcg/dL (normal 15 to 56 mcg/dL) was also present. Despite aggressive care, the patient died of cardiorespiratory arrest secondary to severe brain edema 42 hours after ingestion (Unal et al, 2007).
    b) Death due to cardiorespiratory failure occurred in a 20-month-old boy following an ingestion of 15 g (750 mg/kg) (Janssen et al, 1985).

Maximum Tolerated Exposure

    A) SUMMARY
    1) CASE SERIES: Based on a small case series of 8 of 15 valproate only overdoses, greater than 200 mg/kg (greater than 14 g) was ingested with no severe toxicity reported (Ibister et al, 2003). Drowsiness was reported in 2 patients ingesting 20 g and 25 g, respectively. Based on these findings, the authors concluded that doses less than 400 mg/kg (valproate only) were unlikely to cause severe toxicity.
    B) CASE REPORTS
    1) ADULT
    a) A 29-year-old man with bipolar affective disorder sustained permanent injury when he developed cerebral edema, herniation, and infarct after intentionally ingesting 4 g of valproic acid (VPA) and 75 mg of diazepam (10 hours prior to ingesting VPA). The resulting serum VPA level was 5469 micromol/L (therapeutic; 350 to 700 micromol/L). After 150 days of hospitalization including extensive rehabilitation he was discharged to an assisted living facility with residual right-sided weakness and a modified Rankin scale score of 3 (moderate disability; requiring some help, but able to walk without assistance) (Rupasinghe & Jasinarachchi, 2011; van Swieten et al, 1988).
    b) CASE REPORT: A 26-year-old man, with a history of bipolar disease, intentionally ingested an unknown amount of valproic acid and was admitted a few hours later with sopor without focal neurologic deficits. There were no signs of trauma and a CT of the brain was negative. Laboratory studies were normal. A valproic acid serum level was 2896 micromol/L (therapeutic range: 350 to 690 micromol/L). His cognitive function gradually improved over several days. The patient reported weakness in his right arm on day 3 and he had evidence of weakness of flexion and abduction of the right arm and loss of sensation in the skin over the lateral upper right arm. Multiple diagnostic studies (ie, radiography, MR) were negative. Paresis of the right axillary nerve was detected on physical exam and electrodiagnostic studies. A physical therapy program resulted in gradual improvement (Marusic et al, 2014).
    c) Garnier et al (1982) reported 516 cases of acute valproate overdoses in which patients were unconscious only following ingestions greater than 200 mg/kg, and only 5 had seizures following the ingestion of 100 to 700 mg/kg (Garnier et al, 1982).
    d) Karlsen et al (1983) reported a case where 30 g of sodium valproate was ingested (resulting in a serum concentration of 4925 micromol/L (710 mcg/mL) in 6 hours) but only mild CNS depression occurred for 12 hours (Karlsen et al, 1983).
    e) Lee et al (1998) reported a case of a 45 g ingestion in an 18-year-old man, which resulted in coma, seizures, hyperammonemia, metabolic acidosis, elevated hepatic enzyme concentrations, hypernatremia, and hypocalcemia, with a serum valproate concentration of 575 mcg/mL (Lee et al, 1998).
    f) Following the ingestion of approximately 19 g, a 43-year-old woman developed coma, severe hypotension refractory to fluid replacement and vasopressor therapy, metabolic acidosis, and thrombocytopenia, with a serum valproate concentration of 1,380 mcg/mL (Johnson et al, 1999).
    g) A 41-year-old man developed lethargy and tachycardia approximately 2 hours after reportedly ingesting 50 g of valproate extended release tablets. The patient recovered following hemodialysis (Guillaume et al, 2004).
    2) PEDIATRIC
    a) CASE REPORT: A 9-year-old, 50 kg, girl intentionally ingested 196 mg/kg of valproic acid and upon admission 4 hours after ingestion she was stuporous and required immediate intubation and ventilation. Serum valproic acid level was 599.2 mcg/mL (reference range: 50 to 100 mcg/mL). Hyperammonemia (serum ammonia: 111 mcg/dL) was observed and treated with L-carnitine. Hemoperfusion was started within 8 hours of exposure that was repeated on day 2 for 3 hours; serum valproic level rapidly improved with therapy. The patient was also treated with fresh frozen plasma for an increased prothrombin and activated partial thromboplastin time. The child gradually improved and was extubated on day 4. She was discharged to home on day 9 with no permanent sequelae (Colak et al, 2011).
    3) CHRONIC
    a) Elevated blood ammonia concentrations of 80 (1360 mcg/mL) and 158 (2212 mcg/mL) micromol/L have been reported following chronic administration of 52 and 60 mg/kg/day of valproic acid, respectively (Coulter & Allen, 1981; Rawat et al, 1981).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) The therapeutic serum concentration of valproic acid ranges from 40 to 100 mg/L (Frankfort et al, 2004; Guillaume et al, 2004).
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) ACUTE
    a) Toxicity was associated with serum concentrations exceeding 100 mcg/mL (Turnbull et al, 1983). Delayed peak serum concentrations may occur after overdose due to a delay in the absorption of valproic acid (Spiller & Krenzelok, 2001).
    b) ADVERSE EVENTS: In a retrospective study (n=108) comparing serum drug concentrations with the incidence of adverse outcomes, a peak drug concentration of greater than 450 mcg/mL was associated with the vast majority of adverse outcomes (Beauchamp & Olson, 1999).
    c) CASE SERIES: In a case series (n=133), patients with peak valproic acid concentrations greater than 450 mcg/mL were more likely to have more significant clinical adverse effects and have longer hospital stays (Spiller et al, 2000). Patients with peak concentrations greater than 850 mcg/mL were more likely to develop coma, respiratory depression, aspiration or metabolic acidosis.
    d) SUSTAINED RELEASE PRODUCT: Delayed toxicity has been reported following ingestion of enteric coated and sustained release divalproex sodium. In one adult case a peak valproic acid serum concentration of 7,450 micromol/L (1075 mg/L) was reported 14 hours after ingestion of an unknown quantity of enteric-coated divalproex sodium (Brubacher et al, 1999).
    e) Markedly elevated valproate (greater than 950 mg/L) have been an indication for hemodialysis or charcoal hemoperfusion. However, in one retrospective series of 50 cases of overdose, 4 had documented peak valproate concentrations greater than 950 mg/L (Graeme et al, 1999). None of these had persistent hemodynamic instability or metabolic acidosis and none received hemodialysis or hemoperfusion. All recovered without sequelae.
    2) CASE REPORTS
    a) ADULT
    1) SURVIVAL: A 29-year-old man with bipolar affective disorder intentionally ingested 4 g of valproic acid (VPA) and 75 mg of diazepam and sustained permanent injury after developing cerebral edema, herniation, and infarct. The resulting serum VPA level was 5469 micromol/L (therapeutic: 350 to 700 micromol/L) along with hyperammonemia (348 micromol/L (normal: 0 to 50 micromol/L)) (Rupasinghe & Jasinarachchi, 2011).
    2) SURVIVAL: Peak serum valproate concentration of 1,380 mcg/mL was reported in a 43-year-old woman following an approximate 19 g ingestion (Johnson et al, 1999).
    3) SURVIVAL: A peak serum concentration of 2120 mcg/mL was reported in a 20-year-old, 8.5 hours following a 75 g valproic acid ingestion (Mortensen et al, 1983).
    4) FATALITY: After ingesting 20 g (333 mg/kg) of valproic acid extended release, 60 mg of risperidone, and 3 g of venlafaxine extended release, a 19-year-old man developed hyperammonemia (peak ammonia concentration 1,191 mcg/dL at 65 hours postingestion). The valproic acid concentration peaked at 305.4 mcg/dL. Despite treatment with activated charcoal with sorbitol, and lactulose, the patient's condition deteriorated. The brain CT scan revealed cerebral edema and possible tentorial herniation. At approximately 120 hours after ingestion, cerebral blood flow studies showed an absence of cerebral perfusion and the patient was pronounced brain dead (Camilleri et al, 2005).
    5) FATALITY: Peak serum valproic acid concentration of 2204 mg/L, at 14 hours postingestion, was reported after an overdose of 350 g Depakote(R) in a 24-year-old woman. She died approximately 48 hours postingestion (Christianson et al, 2001).
    6) FATALITY: The peak valproate concentration was 18,900 micromol/L (2725.7 mg/L) at 6 hours postadmission in a 40-year-old epileptic woman who ingested a fatal overdose of an unknown amount of sodium valproate (Connacher et al, 1987).
    7) POSTMORTEM: Postmortem blood concentrations of VPA following a 56 g suicidal ingestion were 720 mg/L. The liver concentration was 800 mg/kg (Lokan & Dinan, 1988).
    8) POSTMORTEM: A 23-year-old man died following initiation of valproic acid therapy. He received a total of 11.25 g over a 5 day period. Serum concentration reported on autopsy was 52 mcg/mL (therapeutic: 50 to 100 mcg/mL). Concentrations were probably higher at the time the valproic acid was discontinued prior to death (Tift, 1980).
    b) INFANT
    1) FATALITY: Peak serum valproate concentration of 268.6 mcg/mL, approximately 12 hours after ingestion, was observed in a 26-day-old infant after being given 300 mg of valproate. By 36 hours, the serum concentration was 60.7 mcg/mL, but severe cerebral edema was present and the infant died 42 hours after exposure (Unal et al, 2007).
    2) FATALITY: A peak serum concentration of 1061 mcg/mL was reported within 3 hours of a 15 g ingestion in a 20-month-old boy (Janssen et al, 1985).
    c) PEDIATRIC
    1) SURVIVAL: A serum concentration of 185 mcg/mL has been associated with coma in a child (Steiman et al, 1979). Serum concentrations of 113 mcg/mL and 120 mcg/mL were associated with mild CNS depression in two adults.

Workplace Standards

    A) ACGIH TLV Values for CAS99-66-1 (American Conference of Governmental Industrial Hygienists, 2010):
    1) Not Listed

    B) NIOSH REL and IDLH Values for CAS99-66-1 (National Institute for Occupational Safety and Health, 2007):
    1) Not Listed

    C) Carcinogenicity Ratings for CAS99-66-1 :
    1) ACGIH (American Conference of Governmental Industrial Hygienists, 2010): Not Listed
    2) EPA (U.S. Environmental Protection Agency, 2011): Not Listed
    3) IARC (International Agency for Research on Cancer (IARC), 2016; International Agency for Research on Cancer, 2015; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2010; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2010a; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2008; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2007; IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2006; IARC, 2004): Not Listed
    4) NIOSH (National Institute for Occupational Safety and Health, 2007): Not Listed
    5) MAK (DFG, 2002): Not Listed
    6) NTP (U.S. Department of Health and Human Services, Public Health Service, National Toxicology Project ): Not Listed

    D) OSHA PEL Values for CAS99-66-1 (U.S. Occupational Safety, and Health Administration (OSHA), 2010):
    1) Not Listed

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) LD50- (INTRAPERITONEAL)MOUSE:
    1) 470 mg/kg ((RTECS, 2002))
    B) LD50- (ORAL)MOUSE:
    1) 1098 mg/kg ((RTECS, 2002))
    C) LD50- (SUBCUTANEOUS)MOUSE:
    1) 860 mg/kg ((RTECS, 2002))
    D) LD50- (ORAL)RAT:
    1) 670 mg/kg ((RTECS, 2002))

Pharmacologic Mechanism

    A) Valproic acid (sometimes formulated as sodium valproate) is structurally unrelated to other anticonvulsant agents.
    1) The mechanism of action is currently unknown, however, it is postulated to interact with the brain gamma-aminobutyric acid (GABA), possibly increasing brain concentrations of this transmitter (Beckner, 1979).
    a) Valproic acid may also inhibit the reuptake of GABA into the glia and nerve endings. It has been shown to be effective in grand mal, petit mal, absence seizures, myoclonic seizures and temporal lobe seizures.
    b) Valproic acid may be effective in seizures resistant to other anticonvulsants.

Toxicologic Mechanism

    A) NEUROTOXICITY: It has been suggested that the 2-EN-valproic acid metabolite may be at least partially responsible for neurologic toxicity (Dupuis et al, 1990).
    B) CEREBRAL EDEMA: The etiology of cerebral edema is unclear, but may be due to a direct toxic effect of VPA or its metabolites and hyperammonemia, increasing intracellular osmolarity, promoting an influx of water, cerebral edema, and increased intracranial pressure (Camilleri et al, 2005; Thabet et al, 2000). Other suspected mechanisms of action include activation of neuronal N-methyl-D-aspartate (NMDA) receptors accompanied by increases in intracellular glutamine acting as an osmolyte and activation of neuronal nitrogen oxide (NO) synthase resulting in vasodilatation (Eyer et al, 2005).
    C) CARDIOTOXICITY: A retrospective analysis of overdoses found that QT prolongation was a common adverse outcome (Kupferschmidt et al, 1999). Atrial tumor myocytes (AT-1 cells, n=4) Ikr was recorded using whole-cell configuration of the patch clamp technique. Valproic acid (50 micromoles/L) completely abolished Ikr in vitro, suggesting that valproic acid-related QT prolongation could be mediated by diminished potassium currents in cardiomyocytes.
    D) CARNITINE DEFICIENCY: It has been hypothesized that carnitine deficiency may occur in some pediatric patients following valproic acid therapy resulting in nonspecific symptoms of lethargy, weakness or hypotonia, hepatotoxicity, and hyperammonemia. Carnitine, an essential amino acid, is necessary for B-oxidation of fatty acids and energy production in cellular mitochondria. In the liver, disturbance of mitochondrial function may result in an inhibition of the hepatic urea cycle and ammonia accumulation (Eyer et al, 2005; Raskind & El-Chaar, 2000).
    E) PANCREATITIS: Postulated mechanisms of action of valproic acid-induced pancreatitis include impairment of the antioxidant defense mechanisms and an excess of free radicals leading to tissue damage (Eyer et al, 2005).

Physical Characteristics

    A) Valproic acid is a white, odorless, crystalline, deliquescent, powder with a characteristic salty taste (JE Reynolds , 2000).

Molecular Weight

    A) VALPROIC ACID: 144.21 (Budavari, 1996)
    B) SODIUM VALPROATE: 166.2 (Budavari, 1996).

General Bibliography

    1) 40 CFR 372.28: Environmental Protection Agency - Toxic Chemical Release Reporting, Community Right-To-Know, Lower thresholds for chemicals of special concern. National Archives and Records Administration (NARA) and the Government Printing Office (GPO). Washington, DC. Final rules current as of Apr 3, 2006.
    2) 40 CFR 372.65: Environmental Protection Agency - Toxic Chemical Release Reporting, Community Right-To-Know, Chemicals and Chemical Categories to which this part applies. National Archives and Records Association (NARA) and the Government Printing Office (GPO), Washington, DC. Final rules current as of Apr 3, 2006.
    3) 49 CFR 172.101 - App. B: Department of Transportation - Table of Hazardous Materials, Appendix B: List of Marine Pollutants. National Archives and Records Administration (NARA) and the Government Printing Office (GPO), Washington, DC. Final rules current as of Aug 29, 2005.
    4) 62 FR 58840: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 1997.
    5) 65 FR 14186: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2000.
    6) 65 FR 39264: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2000.
    7) 65 FR 77866: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2000.
    8) 66 FR 21940: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2001.
    9) 67 FR 7164: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2002.
    10) 68 FR 42710: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2003.
    11) 69 FR 54144: Notice of the National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances - Proposed AEGL Values, Environmental Protection Agency, NAC/AEGL Committee. National Archives and Records Administration (NARA) and the Government Publishing Office (GPO), Washington, DC, 2004.
    12) AAP: The transfer of drugs and other chemicals into human milk. American Academy of Pediatrics, Committee on Drugs. AAP: Pediatrics 1994; 93:137-150.
    13) AIHA: 2006 Emergency Response Planning Guidelines and Workplace Environmental Exposure Level Guides Handbook, American Industrial Hygiene Association, Fairfax, VA, 2006.
    14) Abbot FS, Kassam J, & Orr JM: The effect of aspirin on valproic acid matabolism. Clin Pharmacol Ther 1986; 40:94-100.
    15) Acharya S & Bussel JB: Hematologic toxicity of sodium valproate. J Pediatr Hematol Oncol 2000; 22(1):62-65.
    16) Addison GM & Gordon NS: Sodium valproate and acute hepatic failure. Dev Med Child Neurol 1980; 22:248-249.
    17) Agarwal P, Kumar N, Chandra R, et al: Randomized study of intravenous valproate and phenytoin in status epilepticus. Seizure 2007; 16(6):527-532.
    18) Al Aly Z, Yalamanchili P, & Gonzalez E: Extracorporeal management of valproic acid toxicity: a case report and review of the literature. Semin Dial 2005; 18(1):62-66.
    19) Al-Shareef A, Buss DC, & Shetty HGM: The effect of repeated dose activated charcoal on the pharmacokinetics of sodium valproate in healthy volunteers. Br J Clin Pharmacol 1997; 43:109-111.
    20) Alaspaa AO, Kuisma MJ, Hoppu K, et al: Out-of-hospital administration of activated charcoal by emergency medical services. Ann Emerg Med 2005; 45:207-12.
    21) Alberto G, Erickson T, & Popiel R: Central nervous system manifestations of a valproic acid overdose responsive to naloxone. Ann Emerg Med 1989; 18:889-891.
    22) American Conference of Governmental Industrial Hygienists : ACGIH 2010 Threshold Limit Values (TLVs(R)) for Chemical Substances and Physical Agents and Biological Exposure Indices (BEIs(R)), American Conference of Governmental Industrial Hygienists, Cincinnati, OH, 2010.
    23) Andersen GO & Ritland S: Life threatening intoxication with sodium valproate. Clin Toxicol 1995; 33:279-284.
    24) Anon: Dear Health Care Professional (letter). U.S. Food and Drug Administration. Rockville, MD, USA. 2000. Available from URL: http://www.fda.gov/medwatch/safety/2000/depako.pdf. As accessed Accessed 2000 Aug 02.
    25) Ardinger HH, Atkin JF, & Blackston RD: Verification of the fetalvalproate syndrome phenotype. Am J Med Genetics 1988; 29:171-185.
    26) Armon C, Shin C, & Miller P: Reversible parkinsonism and cognitive impairment with chronic valproate use. Neurology 1996; 47:626-635.
    27) Ashrafi MR, Shabanian R, Zamani GR, et al: Sodium Valproate versus Propranolol in paediatric migraine prophylaxis. Eur J Paediatr Neurol 2005; 9(5):333-338.
    28) Azorin JM & Findling RL: Valproate use in children and adolescents with bipolar disorder. CNS Drugs 2007; 21(12):1019-1033.
    29) Barnes SE & Bower BD: Sodium valproate in the treatment of intractible childhood epilepsy. Develop Med Child Neurol 1975; 17:175.
    30) Barr RD, Copeland SA, & Stockwell ML: Valproic acid and immune thrombocytopenia. Arch Dis Child 1982; 57:681-684.
    31) Barrueto F & Hack JB: Hyperammonemia and coma without hepatic dysfunction induced by valproate therapy. Acad Emerg Med 2001; 8:999-1001.
    32) Baselt RC: Disposition of Toxic Drugs and Chemicals in Man. 2nd ed, Biomedical Publications, Davis, CA, 1982.
    33) Bavoux F, Fournier-Perhilou AI, & Wood C: Neonatal fibrinogen depletion caused by sodium valproate (letter). Ann Pharmacother 1994; 28:1307.
    34) Beauchamp J & Olson K: Valproic acid overdoses: a retrospective study comparing serum drug levels and the incidence of adverse outcomes (abstract). J Toxicol-Clin Toxicol 1999; 37:637-638.
    35) Beckner RR: Valproic acid. Drug Intell Clin Pharm 1979; 13:18-21.
    36) Bidabadi E & Mashouf M: A randomized trial of propranolol versus sodium valproate for the prophylaxis of migraine in pediatric patients. Paediatr Drugs 2010; 12(4):269-275.
    37) Bigler D: Neurologic sequelae after intoxication with sodium valproate. Acta Neurol Scand 1985; 72:351-352.
    38) Binkerd PE, Rowland JM, & Neu H: Evaluation of valproic acid (VPA) developmental toxicity and pharmacokinetics in Sprague-Dawley rats. Fundam Appl Toxicol 1988; 11:485-493.
    39) Blackburn SCF, Oliart AD, & Rodriguez LAG: Antiepileptics and blood dyscrasias: a cohort study. Pharmacother 1998; 18:1277-1283.
    40) Blaheta RA & Cinatl J: Anti-tumor mechanisims of valproate: A novel role for an old drug. Med Res Rev 2002; 22:492-511.
    41) Bohan TP, Helton E, MacDonald I, et al: Effect of L-carnitine treatment for valproate-induced hepatotoxicity. Neurology 2001; 56:1405-09.
    42) Borbath I, Raftopoulos CH, & de Barsy TH: Severe hyperammonemic encephalopathy shortly after valproic acid prescription and rapid awakening following levocarnitine administration (abstract). XX International Congress, EAPCC, European Association of Poisons Centres and Clinical Toxicologists, Amsterdam, The Netherlands, 2000.
    43) Branten AJW, Wetzels JFM, & Weber AM: Hyponatremia due to sodium valproate. Ann Neurol 1998; 43:265-267.
    44) Briggs GG, Freeman RK, & Yaffe SJ: Drugs in Pregnancy and Lactation. 5th ed, Williams and Wilkins, Baltimore, MD, 1998.
    45) Brubacher JR, Dahghani P, & McKnight D: Delayed toxicity following ingestion of enteric-coated divalproex sodium (Epival). J Emerg Med 1999; 17:463-467.
    46) Bruni J & Wilder BJ: Valproic acid. Review of a new antiepileptic drug. Arch Neurol 1979; 36:393-398.
    47) Bryant AE & Dreifuss FE: Valproic acid hepatic fatalities .3. US experience since 1986. Neurology 1996; 46:465-469.
    48) Bryson SM, Verma N, & Scott PJW: Pharmacokinetics in young and elderly subjects. Br J Clin Pharmacol 1983; 16:104-105.
    49) Budavari S: The Merck Index, 12th ed, Merck & Co, Inc, Whitehouse Station, NJ, 1996, pp 1691.
    50) Camfield PR: Pancreatitis due to valproic acid. Lancet 1979; 1:1198.
    51) Camilleri C, Albertson T, & Offerman S: Fatal cerebral edema after moderate valproic acid overdose. Ann Emerg Med 2005; 45(3):337-338.
    52) Caravati EM, Knight HH, & Linscott MS: Esophageal laceration and charcoal mediastinum complicating gastric lavage. J Emerg Med 2001; 20:273-276.
    53) Chadwick DW, Cumming WJ, & Livingstone I: Acute intoxication with sodium valproate. Ann Neurol 1979; 6:552-553.
    54) Chan YC, Tse ML, & Lau FL: Two cases of valproic acid poisoning treated with L-carnitine. Hum Exp Toxicol 2007; 26(12):967-969.
    55) Chapman SA, Wacksman P, & Patterson BD: Pancreatitis associated with valproic acid: a review of the literature. Pharmacotherapy 2001; 21(12):1549-1560.
    56) Chappell KA, Markowitz JS, & Jackson CW: Is valproate pharmacotherapy associated with polycystic ovaries?. Ann Pharmacother 1999; 33:1211-1216.
    57) Chen WT, Yen DJ, & Yu HY: Valproate-induced encephalopathy. Zhonghua Yi Xue Za Zhi (Taipei) 2001; 64:474-478.
    58) Christensen J, Gronborg TK, Sorensen MJ, et al: Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA 2013; 309(16):1696-1703.
    59) Christianson GS, Mowry JB, & Furbee RB: Death associated with massive valproic acid ingestion. (abstract). J Toxicol-Clin Toxicol 2001; 39:498.
    60) Chun AH, Hofman DJ, & Friedfmann N: Bioavailability of valproic acid under fasting/non fasting regimens. J Clin Pharmacol 1980; 20:30-36.
    61) Chuyka PA, Holley JE, & Mandrell TD: Correlation of drug pharmacokinetics and effectiveness of multiple-dose activated charcoal therapy. Ann Emerg Med 1995; 25:356-362.
    62) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    63) Cloyd JC & Kriel RL: Bioavailability of rectally administered valproic acid syrup. Neurology 1981; 31:1348-1352.
    64) Coban D, Kurtoglu S, Akin MA, et al: Neonatal episodic hypoglycemia: a finding of valproic acid withdrawal. J Clin Res Pediatr Endocrinol 2010; 2(2):92-94.
    65) Colak A, Memis D, Guzel A, et al: Valproic acid intoxication with suicide attempt in a pediatric patient. Pediatr Int 2011; 53(5):781-783.
    66) Conley EL, Coley KC, & Pollock BG: Prevalence and risk of thrombocytopenia with valproic acid: experience at a psychiatric teaching hospital. Pharmacotherapy 2001; 21(11):1325-1330.
    67) Connacher AA, Macnab JP, & Moody RT: Fatality due to massive overdose of sodium valproate. Scott Med J 1987; 32:85-86.
    68) Coulter DL & Allen RJ: Hyperammonemia with valproic acid therapy. J Pediatr 1981; 99:317-319.
    69) Coulter DL, Wu H, & Allen RJ: Valproic acid therapy in childhood epilepsy. JAMA 1980; 244:784-788.
    70) DFG: List of MAK and BAT Values 2002, Report No. 38, Deutsche Forschungsgemeinschaft, Commission for the Investigation of Health Hazards of Chemical Compounds in the Work Area, Wiley-VCH, Weinheim, Federal Republic of Germany, 2002.
    71) Dagnone D, Matsui D, & Rieder MJ: Assessment of the palatability of vehicles for activated charcoal in pediatric volunteers. Pediatr Emerg Care 2002; 18:19-21.
    72) Dalens B, Raynaud EJ, & Gaulme J: Teratogenicity of valproic acid. J Pediatr 1980; 97:332-333.
    73) Danielyan A & Kowatch RA: Management options for bipolar disorder in children and adolescents. Paediatr Drugs 2005; 7(5):277-294.
    74) DeToledo JC, Haddad H, & Ramsay RE: Status epilepticus associated with the combination of valproic acid and clomipramine. Therapeutic Drug Monitor 1997; 19:71-73.
    75) DelBello MP, Kowatch RA, Adler CM, et al: A double-blind randomized pilot study comparing quetiapine and divalproex for adolescent mania. J Am Acad Child Adolesc Psychiatry 2006; 45(3):305-313.
    76) DiLiberti JH, Farndon PA, Dennis NR, et al: The fetal valproate syndrome. Am J Med Genet 1984; 19:473-481.
    77) Dickinson RG, Harland RC, & Lynn RK: Transmission of valproic acid (Depakene) across the placenta: half-life of the drug in mother and baby. J Pediatr 1979; 94:832.
    78) Dreifuss FE, Santilli N, & Langer DH: Valproic acid hepatic fatalities: a retrospective review. Neurology 1987; 37:379-385.
    79) Dupuis RE, Lichtman SN, & Pollack GM: Acute valproic acid overdose: clinical course and pharmacokinetic disposition of valproic acid and metabolites. Drug Safety 1990; 5:65-71.
    80) EPA: Search results for Toxic Substances Control Act (TSCA) Inventory Chemicals. US Environmental Protection Agency, Substance Registry System, U.S. EPA's Office of Pollution Prevention and Toxics. Washington, DC. 2005. Available from URL: http://www.epa.gov/srs/.
    81) Ebbesen F, Joergensen A, & Hoseth E: Neonatal hypoglycaemia and withdrawal symptoms after exposure in utero to valproate. Arch Dis Child Fetal Neonatal Ed 2000a; 83:F124-F129.
    82) Ebbesen F, Joergensen A, Hoseth E, et al: Neonatal hypoglycemia and withdrawal symptoms after exposure in utero to valproate. Arch Dis Child Fetal Neonatal Ed 2000; 83:F124-F129.
    83) Eeg-Olofsson O & Linkskog U: Acute intoxication with valproate. Lancet 1982; 1:1306.
    84) Egger J & Brett EM: Effects of sodium valproate in 100 children with special reference to weight. Br Med J 1981; 283:577-581.
    85) Ellaway CJ, Bennetts B, & Tuck RR: Clumsiness, confusion, coma, and valproate. Lancet 1999; 353:1408.
    86) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    87) Emmanouil-Nikoloussi EN, Foroglou NG, Kerameos-Foroglou CH, et al: Effect of valproic acid on fetal and maternal organs in the mouse: a morphological study. Morphologie 2004; 88:41-45.
    88) Eriksson K, Viinikainen K, Monkkonen A, et al: Children exposed to valproate in utero--population based evaluation of risks and confounding factors for long-term neurocognitive development. Epilepsy Res 2005; 65(3):189-200.
    89) Eubanks AL, Aguirre B, & Bourgeois JA: Severe acute hyperammonemia after brief exposure to valproate. Psychosomatics 2008; 49(1):82-83.
    90) Evans RJ, Miranda RN, & Jordan J: Fatal acute pancreatitis caused by valproic acid. Am J Forensic Med Path 1995; 16:62-65.
    91) Evans WE, Schentag JJ, & Jusko WJ: Applied Pharmacokinetics. Principles of Therapeutic Drug Monitoring. 2nd ed, Applied Therapeutics, Inc, Spokane, WA, 1986, pp 543-546.
    92) Eyer F, Felgenhauer N, Gempel K, et al: Acute valproate poisoning: pharmacokinetics, alteration in fatty acid metabolism, and changes during therapy. J Clin Psychopharmacol 2005; 25(4):376-380.
    93) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    94) Falker G & Krause W: Obligoasthenoteratzoo-spermia in valproic acid therapy. Zetschrift Fur Haukrankeiten 1988; 63:142.
    95) Farrar HC, Herold DA, & Reed MD: Acute valproic acid intoxication: enhanced drug clearance with oral-activated charcoal. Crit Care Med 1993; 21:299-301.
    96) Farrell K, Orr JM, & Abbott FS: The effect of acetylsalicylic acid on serum free valproate concentrations and valproate clearance in children. J Pediatr 1982; 101:142-144.
    97) Felgenhauer N, Clarmann MV, & Zilker TH: Severe hyperammonaemia in a case of fetal valproate overdose. (#260), Scientific Meeting, European Association of Poisons Centres and Clinical Toxicologists, Birmingham, UK, 1993.
    98) Field J & Daly FS: Continuous veno-venous haemodiafiltration in sodium valproate overdose complicated by cerebral oedema: a case report. Crit Care Resusc 2002; 4(3):173-176.
    99) Findling RL, McNamara NK, Youngstrom EA, et al: Double-blind 18-month trial of lithium versus divalproex maintenance treatment in pediatric bipolar disorder. J Am Acad Child Adolesc Psychiatry 2005; 44(5):409-417.
    100) Finnell RH, Bennett GD, & Karras SB: Common hierarchies of susceptibility to the induction of neural tube defects in mouse embryos by valproic acid and its 4-propyl-4-pentenoic acid metabolite. Teratology 1988; 38:313-320.
    101) Finsterer J, Pelzl G, & Hess B: Severe, isolated thrombocytopenia under polytherapy with carbamazepine and valproate. Psychiatry Clin Neurosci 2001; 55:423-426.
    102) Fischman MA, Hull D, & Bartus SA: Valproate for epilepsy in renal trnsplant recipients receiving cyclosporine (letter). Transplantation 1989; 48:542.
    103) Forster D: Anticonvulsant therapy with dipropyl acetate in children. Muchen Med Wochenschr 1972; 114:399.
    104) Frankfort SV, Tulner LR, Knol W, et al: Prescription error resulting in valproic acid intoxication. J Am Geriatr Soc 2004; 52(12):2142-2143.
    105) Franssen EJF, van Essen GG, & Portman AT: Valproic acid toxicokinetics: serial hemodialysis and hemoperfusion. Ther Drug Monitor 1999; 21:289-292.
    106) Fuller JP, Girardel M, & Saunder P: Intoxication aigue par le valproate de sodium. Nouv Presse Med 1981; 10:3323.
    107) Garnier R & Fournier E: Intoxication aigue par le valproate de sodium. Nouv Presse Med 1982; 11:678.
    108) Garnier R, Boudignat O, & Fournier PE: Valproate poisoning. Lancet 1982; 2:97.
    109) Gerber N, Dickinson RG, & Harland RC: Reye-like syndrome associated with valproic acid therapy. J Pediatr 1979; 95:142-144.
    110) Gerstenbrand F: Augmented indication field for convulex therapy. Wein Klin Wochenschr 1976; 88:760.
    111) Ghannoum M, Laliberte M, Nolin TD, et al: Extracorporeal treatment for valproic acid poisoning: systematic review and recommendations from the EXTRIP workgroup. Clin Toxicol (Phila) 2015; 53(5):454-465.
    112) Gidal B, Spencer N, & Maly M: Valproate-mediated disturbances of hemostasis: relationship to dose and plasma concentration. Neurology 1994; 44:1418-1422.
    113) Gigli GL, Scalise A, & Pauri F: Valproate-induced systemic lupus erythematosus in a patient with partial trisomy of chromosome 9 and epilepsy. Epilepsia 1996; 37:587-588.
    114) Gillman MA & Sandyk R: Nicotinic acid deficiency induced by sodium valproate (letter). S Afr Med J 1984; 65:986.
    115) Glover SJ, Quinn AG, & Barter P: Ophthalmic findings in fetal anti-convulsant syndrome(s). Ophthalmology 2002; 109:942-947.
    116) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    117) Goulden KJ, Dooley JM, & Camfield PR: Clinical valproate toxicity induced by acetylsalicylic acid. Neurology 1987; 37:1392-1394.
    118) Gourru JL: Intoxication aigue massive par le valproate de sodium: a propos d'une observation d'intoxication voluntaire mortelle, Medical thesis, Univ Lyon I, Lyons, France, 1981.
    119) Graeme K, Higgins T, & Curry S: Markedly elevated valproate levels do not serve as an indication for hemodialysis or hemoperfusion (abstract). J Toxicol-Clin Toxicol 1999; 37:636.
    120) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    121) Graudins A & Aaron CK: Delayed peak serum valproic acid level in massive divalproex overdose - treatment with charcoal hemoperfusion. Clin Toxicol 1996; 34:335-341.
    122) Guenther Skokan E, Junkins EP, & Corneli HM: Taste test: children rate flavoring agents used with activated charcoal. Arch Pediatr Adolesc Med 2001; 155:683-686.
    123) Gugler R, Schell A, & Eichelbaum M: Disposition of valproic acid in man. Eur J Clin Pharmacol 1977; 12:125-132.
    124) Guillaume CP, Stolk L, Dejagere TF, et al: Successful use of hemodialysis in acute valproic acid intoxication. J Toxicol Clin Toxicol 2004; 42(3):335-336.
    125) Halaby A, Haddad R, & Naja WJ: Hyperammonemia induced by interaction of valproate and quetiapine. Curr Drug Saf 2013; 8(4):284-286.
    126) Hamer HM, Knake S, & Schomburg U: Valproate-induced hyperammonemic encephalopathy in the presence of topiramate. Neurology 2000; 54:230-232.
    127) Hamer HM, Knake S, Schomburg U, et al: Valproate-induced hyperammonemic encephalopathy in the presence of topiramate. Neurology 2000a; 54:230-232.
    128) Hardy MA & Nardacci D: Does valproate pose a threat to human immunodeficiency virus-infected patients? (letter). J Clin Psychopharmacol 1999; 19:189-190.
    129) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    130) Herranz JL: Epilepsia 1982; 23:203.
    131) Hicks LK & McFarlane PA: Valproic acid overdose and haemodialysis. Nephrol Dialysis Transplant 2001; 16:1483-1486.
    132) Hintze G, Klein HH, & Prange H: A case of valproate intoxication with excessive brain edema. Klin Wochenschr 1987; 65:424-427.
    133) Hovinga CA, Chicella MF, Rose DF, et al: Use of intravenous valproate in three pediatric patients with nonconvulsive or convulsive status epilepticus. Ann Pharmacother 1999; 33(5):579-584.
    134) Hu LJ, Lu XF, & Lu BQ: The effect of valproic acid on SCE and chromosome aberrations in epileptic children. Mutat Res 1990; 243:63-66.
    135) Huot C, Gauthier M, & Lebel M: Congenital malformations associated with maternal use of valproic acid. Can J Neurol Sci 1987; 14:290-293.
    136) Hurd RW, Van Rinsvelt HA, & Wilder BJ: Selenium, zinc, and copperchanges with valproic acid: possible relation to drug side effects. Neurology 1984; 34:1393-1395.
    137) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: 1,3-Butadiene, Ethylene Oxide and Vinyl Halides (Vinyl Fluoride, Vinyl Chloride and Vinyl Bromide), 97, International Agency for Research on Cancer, Lyon, France, 2008.
    138) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Formaldehyde, 2-Butoxyethanol and 1-tert-Butoxypropan-2-ol, 88, International Agency for Research on Cancer, Lyon, France, 2006.
    139) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Household Use of Solid Fuels and High-temperature Frying, 95, International Agency for Research on Cancer, Lyon, France, 2010a.
    140) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Smokeless Tobacco and Some Tobacco-specific N-Nitrosamines, 89, International Agency for Research on Cancer, Lyon, France, 2007.
    141) IARC Working Group on the Evaluation of Carcinogenic Risks to Humans : IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Some Non-heterocyclic Polycyclic Aromatic Hydrocarbons and Some Related Exposures, 92, International Agency for Research on Cancer, Lyon, France, 2010.
    142) IARC: List of all agents, mixtures and exposures evaluated to date - IARC Monographs: Overall Evaluations of Carcinogenicity to Humans, Volumes 1-88, 1972-PRESENT. World Health Organization, International Agency for Research on Cancer. Lyon, FranceAvailable from URL: http://monographs.iarc.fr/monoeval/crthall.html. As accessed Oct 07, 2004.
    143) Ibister GK, Balit CR, Whyte IM, et al: Valproate overdose: A comparative cohort study of self poisonings. Br J Clin Pharmacol 2003; 55:398-404.
    144) International Agency for Research on Cancer (IARC): IARC monographs on the evaluation of carcinogenic risks to humans: list of classifications, volumes 1-116. International Agency for Research on Cancer (IARC). Lyon, France. 2016. Available from URL: http://monographs.iarc.fr/ENG/Classification/latest_classif.php. As accessed 2016-08-24.
    145) International Agency for Research on Cancer: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. World Health Organization. Geneva, Switzerland. 2015. Available from URL: http://monographs.iarc.fr/ENG/Classification/. As accessed 2015-08-06.
    146) Iqbal MM, Sohhan T, & Mahmud SZ: The effects of lithium, valproic acid, and carbamazepine during pregnancy and lactation. Clin Toxicol 2001; 39:381-392.
    147) Ishidura H, Matsu N, & Matsubara I: Valproic acid overdose and L-carnitine therapy. J Anal Toxicol 1996; 20:55-58.
    148) Isojarvi JIT, Tauboll E, & Pakarinen AJ: Altered ovarian function and cardiovascular risk factors in valproate-treated women. Am J Med 2001; 111:290-296.
    149) JE Reynolds : Martindale: The Extra Pharmacopoeia (Internet Version). The Pharmaceutical Press. London, UK (Internet Version). Edition expires 2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    150) Jaeken J, Caesar P, & Corbeel L: Valproate hyperammonaemia and hyperglycinaemia. Lancet 1980; 2:260.
    151) Janssen F, Rambeck B, & Schnabel R: Acute valproate intoxication with fatal outcome in an infant. Neuropediatr 1985; 16:235-238.
    152) Jeavons PM: Non-dose-related side effects of valproate. Epilepsia 1984; 25(Suppl 1):S50-S55.
    153) Jeavons PM: Sodium valproate and neural tube defects. Lancet 1982; 2:1282-1283.
    154) Jentink J, Loane MA, Dolk H, et al: Valproic Acid Monotherapy in Pregnancy and Major Congenital Malformations. N Engl J Med 2010; 362(23):2185-2193.
    155) Johannessen FL: Preliminary observations on valproic acid kinetics in patients with epilepsy. Arzneim Forsch 1977; 27:1083-1085.
    156) Johnson LZ, Martinez I, & Fernandez MC: Successful treatment of valproic acid overdose with hemodialysis. Amer J Kidney Dis 1999; 33:786-789.
    157) Johnson LZ, Martinez L, Fernandez MC, et al: Successful treatment of valproic acid overdose with hemodialysis. Am J Kidney Dis 1999a; 33(4):786-789.
    158) Jonville AP, Gauchez AS, & Autret E: Interaction between isoniazid and valproate: a case of valproate overdosage. Eur J Clin Pharmacol 1991; 40:197-198.
    159) Jung J, Eo E, & Ahn KO: A case of hemoperfusion and L-carnitine management in valproic acid overdose. Am J Emerg Med 2008; 26(3):388e3-388e4.
    160) Kane SL, Constantiner M, & Staubus AE: High-flux hemodialysis without hemoperfusion is effective in acute valproic acid overdose. Ann Pharmacother 2000; 34:1146-1151.
    161) Karlsen RL, Kett K, & Henriksen O: Intoxication with sodium valproate. Acta Med Scand 1983; 213:405-406.
    162) Kay TD, Playford HR, & Johnson DW: Hemodialysis versus continuous veno-venous hemodiafiltration in the management of severe valproate overdose. Clin Nephrol 2003; 59:56-58.
    163) Kearney TE: Valproic acid, in: Olson KR (Ed), Poisoning and Drug Overdose (2nd ed), Appleton & Lange, Norwalk, CT, 1994, pp 302-305.
    164) Khan E, Huggan P, Celi L, et al: Sustained low-efficiency dialysis with filtration (SLEDD-f) in the management of acute sodium valproate intoxication. Hemodial Int 2008; 12(2):211-214.
    165) Kielstein JT, Woywodt A, Schumann G, et al: Efficiency of high-flux hemodialysis in the treatment of valproic acid intoxication. J Toxicol Clin Toxicol 2003; 41:873-876.
    166) Kingsley E, Tweedale R, & Gray P: The role of toxic metabolites in the hepatotoxicity of valproic acid. Gastroenterol 1980; 79:511.
    167) 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.
    168) 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.
    169) Klotz U: Pharmacokinetics studies with valproic acid in man. Arzneim Forsch 1977; 27:1085.
    170) Koelliker P, Koelliker D, & Toerne T: Bullous skin lesions associated with severe valproic acid overdose in a four year-old child (abstract). J Toxicol-Clin Toxicol 1999; 37:638.
    171) Kroll P & Nand C: Hemodialysis in the treatment of valproic acid overdose (letter). J Clin Psychiatry 2002; 63:78-79.
    172) Kugler J: The treatment of therapy-resistant epilepsy with dipropyl actate. Munchen Med Wochenschr 1973; 115:1103.
    173) Kuperschmidt H, Seger D, & Dawling S: Multiple life-threatening metabolic complications in severe valproic acid overdose (abstract). J Toxicol-Clin Toxicol 1998; 36:471.
    174) Kupferschmidt HHT, Kevorkian JP, & Yang T: Cardiotoxicity in valproic acid poisoning (abstract). J Toxicol-Clin Toxicol 1999; 37:385.
    175) Lautin A: Br Med J 1979; 2:1035.
    176) Lee WL, Yang CC, & Deng JF: A case of severe hyperammonemia and unconsciousness following sodium valproate intoxication. Vet Human Toxicol 1998; 40:346-348.
    177) Lewis DP, Van Dyke DC, & Stumbo PJ: Drug and environmental factors associated with adverse pregnancy outcomes. PartI: Antiepileptic drugs, contraceptives, smoking, and folate. Ann Pharmacother 1998; 32:802-817.
    178) Lewis JR: Valproic acid (Depakene): a new anticonvulsant agent. JAMA 1978; 240:2190-2192.
    179) Lheureux PE & Hantson P: Carnitine in the treatment of valproic acid-induced toxicity. Clin Toxicol (Phila) 2009; 47(2):101-111.
    180) Lheureux PE, Penaloza A, Zahir S, et al: Science review: carnitine in the treatment of valproic acid-induced toxicity - what is the evidence?. Crit Care 2005; 9(5):431-440.
    181) Lin WC, Tsao JH, & Tsai SJ: Delayed thrombocytopenia after valproic acid overdose. Gen Hosp Psychiatry 2008; 30(5):489-490.
    182) LoVecchio F, Shriki J, & Samaddar R: L-carnitine was safely administered in the setting of valproate toxicity. Am J Emerg Med 2005; 23(3):321-322.
    183) Loiseau P, Brachet A, & Henry P: Concentration of dipropyl acetate in plasma. Epilepsia 1975; 16:609.
    184) Loiseau P: Treatment of concomitant illneses in patients receiving anticonvulsants. Drug interactions of clinical significance. Drug Safety 1998; 19:495-510.
    185) Lokan RJ & Dinan AC: An apparent fatal valproic acid poisoning. J Analyt Tox 1988; 12:35-37.
    186) Lynch A & Tobias JD: Acute valproate ingestion induces symptomatic methemoglobinemia. Ped Emerg Care 1998; 14:205-207.
    187) MacDougall LG: JAMA 1982; 247:53.
    188) Manoguerra AS, Erdman AR, Woolf AD, et al: Valproic acid poisoning: an evidence-based consensus guideline for out-of-hospital management. Clin Toxicol (Phila) 2008; 46(7):661-676.
    189) Marbury TC, Lee CS, & Bruni J: Hemodialysis of valproic acid in uremic patients. Dialysis and Transplantation 1980; 9:961.
    190) Marescaux C, Warter JM, & Laroge M: Sodium valproate; a hyperammonemiadrug-study in epileptics and normal subjects. J Neurol Sci 1983; 58:195-209.
    191) Marusic S, Obreli Neto PR, Vuletic V, et al: Peripheral mononeuropathy associated with valproic acid poisoning in an adult patient. Int J Clin Pharmacol Ther 2014; 52(9):802-804.
    192) Mauz PS, Morike K, Kaiserling E, et al: Valproic acid-associated sialadenosis of the parotid and submandibular glands: diagnostic and therapeutic aspects. Acta Otolaryngol 2005; 125(4):386-391.
    193) May CA, Garnett WR, & Small RE: Effects of three antacids on the bioavailability of valproic acid. Clin Pharm 1982; 1:244-247.
    194) McLaughlin DB, Eadie MJ, & Parker-Scott SL: Valproate metabolism during valproate-associated hepatotoxicity in a surviving adult patient. Epilep Res 2000; 41:259-268.
    195) McMahon CL & Braddock SR: Septo-optic dysplasia as a manifestation of valproic acid embryopathy. Teratology 2001; 64:83-86.
    196) Meador KJ, Baker GA, Browning N, et al: Fetal antiepileptic drug exposure and cognitive outcomes at age 6 years (NEAD study): a prospective observational study. Lancet Neurol 2013; 12(3):244-252.
    197) Meek MF, Broekroelofs J, Yska JP, et al: Valproic acid intoxication: sense and non-sense of haemodialysis. Neth J Med 2004; 62(9):333-336.
    198) Mehta V, Singhi P, & Singhi S: Intravenous sodium valproate versus diazepam infusion for the control of refractory status epilepticus in children: a randomized controlled trial. J Child Neurol 2007; 22(10):1191-1197.
    199) Menegola E, Broccia ML, & Nau H: Teratogenic effects of sodium valproate in mice and rats at midgestation and at term. Teratogen Carcinogen Mutagen 1996; 16:97-108.
    200) Meyer S, Kuhlmann MK, Peters FT, et al: Severe valproic acid intoxication is associated with atrial tachycardia: secondary detoxication by hemoperfusion. Klin Padiatr 2005; 217(2):82-85.
    201) Mileusnic D, Donoghue ER, & Lifschultz BD: Pathological case of the month: Sudden death in a child as a result of pancreatitis during valproic acid therapy. Ped Pathol Molecular Med 2002; 21:477-484.
    202) Minari M, Maggiore U, & Tagliavini D: Severe acute valproic acid intoxication successfully treated with hemodiafiltration without hemoperfusion (letter). Ann Emerg Med 2002; 39:204-205.
    203) Misra UK, Kalita J, & Patel R: Sodium valproate vs phenytoin in status epilepticus: a pilot study. Neurology 2006; 67(2):340-342.
    204) Mo CN & Ladusans EJ: Anomalous right pulmonary artery origins in association with the fetal valproate syndrome. J Med Genet 1999; 36:83-84.
    205) Moffa AM, White JA, & MacKay EG: Valproic acid, zinc and open neural tubes in the 9 day-old hamster embryos. Teratology 1984; 29:47A.
    206) Moinho R, Dias A, Estanqueiro P, et al: Overdose with antiepileptic drugs: the efficacy of extracorporeal removal techniques. BMJ Case Rep 2014; 2014:1.
    207) Montero FJ: Naloxone in the reversal of coma induced by sodium valproate (letter). Ann Emerg Med 1999; 33:357-358.
    208) Morikawa N, Mori T, & Kawashima H: Change of valproic acid concentrations during cerebrospinal fluid perfusion chemotherapy (letter). Ann Pharmacother 2000; 34:536-537.
    209) Morrow J, Russell A, Guthrie E, et al: Malformation risks of antiepileptic drugs in pregnancy: a prospective study from the UK Epilepsy and Pregnancy Register. J Neurol Neurosurg Psychiatry 2006; 77(2):193-198.
    210) Morrow JI & Routledge PA: Poisoning by anticonvulsants. Adverse Drug React Acute Poison Rev 1989; 2:97-109.
    211) Mortensen PB, Hansen HE, & Pedersen B: Acute valproate intoxication: biochemical investigations and hemodialysis treatment. Int J Clin Pharmacol Ther Toxicol 1983; 21:64-68.
    212) Morton LD, O'Hara KA, Coots BP, et al: Safety of rapid intravenous valproate infusion in pediatric patients. Pediatr Neurol 2007; 36(2):81-83.
    213) NFPA: Fire Protection Guide to Hazardous Materials, 13th ed., National Fire Protection Association, Quincy, MA, 2002.
    214) NRC: Acute Exposure Guideline Levels for Selected Airborne Chemicals - Volume 1, Subcommittee on Acute Exposure Guideline Levels, Committee on Toxicology, Board on Environmental Studies and Toxicology, Commission of Life Sciences, National Research Council. National Academy Press, Washington, DC, 2001.
    215) NRC: Acute Exposure Guideline Levels for Selected Airborne Chemicals - Volume 2, Subcommittee on Acute Exposure Guideline Levels, Committee on Toxicology, Board on Environmental Studies and Toxicology, Commission of Life Sciences, National Research Council. National Academy Press, Washington, DC, 2002.
    216) NRC: Acute Exposure Guideline Levels for Selected Airborne Chemicals - Volume 3, Subcommittee on Acute Exposure Guideline Levels, Committee on Toxicology, Board on Environmental Studies and Toxicology, Commission of Life Sciences, National Research Council. National Academy Press, Washington, DC, 2003.
    217) NRC: Acute Exposure Guideline Levels for Selected Airborne Chemicals - Volume 4, Subcommittee on Acute Exposure Guideline Levels, Committee on Toxicology, Board on Environmental Studies and Toxicology, Commission of Life Sciences, National Research Council. National Academy Press, Washington, DC, 2004.
    218) Nagarajan L, Johnston K, & Williams S: Hypothermia and thermoregulatory derangements induced by valproic acid (letter). Neurology 2001; 56:139.
    219) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,2,3-Trimethylbenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2006k. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d68a&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    220) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,2,4-Trimethylbenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2006m. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d68a&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    221) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,2-Butylene Oxide (Proposed). United States Environmental Protection Agency. Washington, DC. 2008d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648083cdbb&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    222) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,2-Dibromoethane (Proposed). United States Environmental Protection Agency. Washington, DC. 2007g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064802796db&disposition=attachment&contentType=pdf. As accessed 2010-08-18.
    223) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 1,3,5-Trimethylbenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2006l. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d68a&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    224) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for 2-Ethylhexyl Chloroformate (Proposed). United States Environmental Protection Agency. Washington, DC. 2007b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648037904e&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    225) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Acrylonitrile (Proposed). United States Environmental Protection Agency. Washington, DC. 2007c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648028e6a3&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    226) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Adamsite (Proposed). United States Environmental Protection Agency. Washington, DC. 2007h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    227) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Agent BZ (3-quinuclidinyl benzilate) (Proposed). United States Environmental Protection Agency. Washington, DC. 2007f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064803ad507&disposition=attachment&contentType=pdf. As accessed 2010-08-18.
    228) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Allyl Chloride (Proposed). United States Environmental Protection Agency. Washington, DC. 2008. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648039d9ee&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    229) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Aluminum Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    230) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Arsenic Trioxide (Proposed). United States Environmental Protection Agency. Washington, DC. 2007m. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480220305&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    231) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Automotive Gasoline Unleaded (Proposed). United States Environmental Protection Agency. Washington, DC. 2009a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7cc17&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    232) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Biphenyl (Proposed). United States Environmental Protection Agency. Washington, DC. 2005j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064801ea1b7&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    233) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Bis-Chloromethyl Ether (BCME) (Proposed). United States Environmental Protection Agency. Washington, DC. 2006n. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648022db11&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    234) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Boron Tribromide (Proposed). United States Environmental Protection Agency. Washington, DC. 2008a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064803ae1d3&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    235) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Bromine Chloride (Proposed). United States Environmental Protection Agency. Washington, DC. 2007d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648039732a&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    236) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Bromoacetone (Proposed). United States Environmental Protection Agency. Washington, DC. 2008e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064809187bf&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    237) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Calcium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    238) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Carbonyl Fluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2008b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064803ae328&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    239) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Carbonyl Sulfide (Proposed). United States Environmental Protection Agency. Washington, DC. 2007e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648037ff26&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    240) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Chlorobenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2008c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064803a52bb&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    241) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Cyanogen (Proposed). United States Environmental Protection Agency. Washington, DC. 2008f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064809187fe&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    242) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Dimethyl Phosphite (Proposed). United States Environmental Protection Agency. Washington, DC. 2009. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7cbf3&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    243) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Diphenylchloroarsine (Proposed). United States Environmental Protection Agency. Washington, DC. 2007l. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    244) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ethyl Isocyanate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648091884e&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    245) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ethyl Phosphorodichloridate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480920347&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    246) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ethylbenzene (Proposed). United States Environmental Protection Agency. Washington, DC. 2008g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064809203e7&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    247) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ethyldichloroarsine (Proposed). United States Environmental Protection Agency. Washington, DC. 2007j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    248) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Germane (Proposed). United States Environmental Protection Agency. Washington, DC. 2008j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963906&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    249) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Hexafluoropropylene (Proposed). United States Environmental Protection Agency. Washington, DC. 2006. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064801ea1f5&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    250) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Ketene (Proposed). United States Environmental Protection Agency. Washington, DC. 2007. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020ee7c&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    251) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Magnesium Aluminum Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    252) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Magnesium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    253) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Malathion (Proposed). United States Environmental Protection Agency. Washington, DC. 2009k. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064809639df&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    254) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Mercury Vapor (Proposed). United States Environmental Protection Agency. Washington, DC. 2009b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a8a087&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    255) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyl Isothiocyanate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008k. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963a03&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    256) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyl Parathion (Proposed). United States Environmental Protection Agency. Washington, DC. 2008l. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963a57&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    257) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyl tertiary-butyl ether (Proposed). United States Environmental Protection Agency. Washington, DC. 2007a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064802a4985&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    258) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methylchlorosilane (Proposed). United States Environmental Protection Agency. Washington, DC. 2005. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5f4&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    259) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyldichloroarsine (Proposed). United States Environmental Protection Agency. Washington, DC. 2007i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    260) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Methyldichlorosilane (Proposed). United States Environmental Protection Agency. Washington, DC. 2005a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c646&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    261) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Mustard (HN1 CAS Reg. No. 538-07-8) (Proposed). United States Environmental Protection Agency. Washington, DC. 2006a. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d6cb&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    262) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Mustard (HN2 CAS Reg. No. 51-75-2) (Proposed). United States Environmental Protection Agency. Washington, DC. 2006b. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d6cb&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    263) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Mustard (HN3 CAS Reg. No. 555-77-1) (Proposed). United States Environmental Protection Agency. Washington, DC. 2006c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d6cb&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    264) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Tetroxide (Proposed). United States Environmental Protection Agency. Washington, DC. 2008n. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648091855b&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    265) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Nitrogen Trifluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2009l. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963e0c&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    266) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Parathion (Proposed). United States Environmental Protection Agency. Washington, DC. 2008o. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480963e32&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    267) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Perchloryl Fluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2009c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7e268&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    268) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Perfluoroisobutylene (Proposed). United States Environmental Protection Agency. Washington, DC. 2009d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7e26a&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    269) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phenyl Isocyanate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008p. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096dd58&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    270) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phenyl Mercaptan (Proposed). United States Environmental Protection Agency. Washington, DC. 2006d. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020cc0c&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    271) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phenyldichloroarsine (Proposed). United States Environmental Protection Agency. Washington, DC. 2007k. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020fd29&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    272) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phorate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008q. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096dcc8&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    273) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phosgene (Draft-Revised). United States Environmental Protection Agency. Washington, DC. 2009e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a8a08a&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    274) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Phosgene Oxime (Proposed). United States Environmental Protection Agency. Washington, DC. 2009f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7e26d&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    275) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Potassium Cyanide (Proposed). United States Environmental Protection Agency. Washington, DC. 2009g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7cbb9&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    276) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Potassium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005c. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    277) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Propargyl Alcohol (Proposed). United States Environmental Protection Agency. Washington, DC. 2006e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020ec91&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    278) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Selenium Hexafluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2006f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020ec55&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    279) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Silane (Proposed). United States Environmental Protection Agency. Washington, DC. 2006g. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d523&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    280) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Sodium Cyanide (Proposed). United States Environmental Protection Agency. Washington, DC. 2009h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7cbb9&disposition=attachment&contentType=pdf. As accessed 2010-08-15.
    281) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Sodium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    282) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Strontium Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005f. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    283) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Sulfuryl Chloride (Proposed). United States Environmental Protection Agency. Washington, DC. 2006h. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020ec7a&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    284) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Tear Gas (Proposed). United States Environmental Protection Agency. Washington, DC. 2008s. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096e551&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    285) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Tellurium Hexafluoride (Proposed). United States Environmental Protection Agency. Washington, DC. 2009i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7e2a1&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    286) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Tert-Octyl Mercaptan (Proposed). United States Environmental Protection Agency. Washington, DC. 2008r. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096e5c7&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    287) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Tetramethoxysilane (Proposed). United States Environmental Protection Agency. Washington, DC. 2006j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d632&disposition=attachment&contentType=pdf. As accessed 2010-08-17.
    288) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Trimethoxysilane (Proposed). United States Environmental Protection Agency. Washington, DC. 2006i. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d632&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    289) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Trimethyl Phosphite (Proposed). United States Environmental Protection Agency. Washington, DC. 2009j. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=0900006480a7d608&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    290) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Trimethylacetyl Chloride (Proposed). United States Environmental Protection Agency. Washington, DC. 2008t. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648096e5cc&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    291) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for Zinc Phosphide (Proposed). United States Environmental Protection Agency. Washington, DC. 2005e. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020c5ed&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    292) National Advisory Committee for Acute Exposure Guideline Levels for Hazardous Substances: Acute Exposure Guideline Levels (AEGLs) for n-Butyl Isocyanate (Proposed). United States Environmental Protection Agency. Washington, DC. 2008m. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=09000064808f9591&disposition=attachment&contentType=pdf. As accessed 2010-08-12.
    293) National Institute for Occupational Safety and Health: NIOSH Pocket Guide to Chemical Hazards, U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, Cincinnati, OH, 2007.
    294) National Research Council : Acute exposure guideline levels for selected airborne chemicals, 5, National Academies Press, Washington, DC, 2007.
    295) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 6, National Academies Press, Washington, DC, 2008.
    296) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 7, National Academies Press, Washington, DC, 2009.
    297) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 8, National Academies Press, Washington, DC, 2010.
    298) Nau H & Loscher W: Pharmacologic evaluation of various metabolites andanalogs of valproic acid: Teratogenic potencies in mice. Fundam Appl Toxicol 1986; 6:669-676.
    299) Nau H & Scott WJ: Weak acids may act as teratogens by accumulating in the basic milieu of the early mammalian embryo. Nature 1986; 323:276-278.
    300) Neuvonen PJ, Kannisto H, & Hirvisalo EL: Effect of activated charcoal on absorption of tolbutamide and valproate in man. Eur J Clin Pharmacol 1983; 24(2):243-246.
    301) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    302) Ohtani Y, Endo F, & Matsuda I: Carnitine deficiency and hyperammonemia associated with valproic acid therapy. J Pediatr 1982; 101:782-785.
    303) Orr JM, Abbott FS, & Farrell K: Interaction between valproic acid and aspirin in epileptic children: serum protein binding and metabolic effects. Clin Pharmacol Ther 1982; 31:642-649.
    304) Ozkan H, Cetinkaya M, Koksal N, et al: Severe fetal valproate syndrome: combination of complex cardiac defect, multicystic dysplastic kidney, and trigonocephaly. J Matern Fetal Neonatal Med 2011; 24(3):521-524.
    305) Palatnick W, Honcharik N, & Roberts D: Coma, anion gap, and metabolic derangements associated with massive valproic acid (abstract 155). Vet Hum Toxicol 1989; 31:368.
    306) Panikkar GP & Gilman SM: Valproate-induced hyperammonemia in the psychiatric setting: 2 cases (letter). J Clin Psychiatry 1999; 60:557-559.
    307) Papaseit E, Farre M, Lopez MJ, et al: A case of acute valproic acid poisoning treated successfully with L-carnitine. Euro J Emerg Med 2012; 19(1):57-58.
    308) Pavuluri MN, Henry DB, Carbray JA, et al: Divalproex sodium for pediatric mixed mania: a 6-month prospective trial. Bipolar Disord 2005; 7(3):266-273.
    309) Payen C, Frantz P, Martin O, et al: Delayed toxicity following acute ingestion of valpromide. Human Experimental Toxicol 2004; 23:145-148.
    310) 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.
    311) Pedersen B & Juul-Jensen P: Electroencephalographic alterations during intoxication with sodium valproate: a case report. Epilepsia 1984; 25:121-124.
    312) Pedersen B: Intoxication after large overdose of valproate. In: Valproate in the Treatment of Seizures. Danish Epilepsy Society, 1981.
    313) Perucca E, Gatti G, & Frigo GM: Disposition of sodium valproate in epileptic patients. Br J Clin Pharmacol 1978; 5:495-499.
    314) Picart N, Periole B, & Mazereeuw J: Drug hypersensitivity syndrome to valproic acid, [Article in French]. Presse Med 2000; 29:648-650.
    315) Pinder RM, Borgden RN, & Speight TM: Sodium valproate: a review of its pharmacological properties in therapeutic efficacy in epilepsy. Drugs 1977; 13:81.
    316) Pinkston R & Walker LA: Multiorgan system failure caused by valproic acid toxicity. Am J Emerg Med 1997; 15:504-506.
    317) Piontek CM, Baab S, & Peindl KS: Serum valproate levels in 6 breastfeeding mother-infant pairs. J Clin Psychiatry 2000; 61(3):170-172.
    318) Plaza MM, Goyanes GR, & Cuina L: On the toxicity of valproic-acid. Clin Nephrol 1999; 51:187-189.
    319) Poklis A, Poklis JL, & Trautman D: Disposition of valproic acid in a case of fatal intoxication. J Anal Toxicol 1998; 22:537-540.
    320) Polifka JE & Friedman JM: Medical genetics: 1.Clinical teratology in the age of genomics. CMAJ 2002; 167:265-273.
    321) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    322) Popiel R, Erickson T, & Leikin JB: Valproic acid overdose: response to naloxone (abstract 156). Vet Hum Toxicol 1989; 31:368.
    323) Product Information: DEPACON(R) IV injection, valproate sodium IV injection. Abbott Laboratories, North Chicago, IL, 2006.
    324) Product Information: DEPAKENE oral capsules, oral solution, valproic acid oral capsules, oral solution. AbbVie Inc. (per FDA), North Chicago, IL, 2014.
    325) Product Information: DEPAKENE oral capsules, oral solution, valproic acid oral capsules, oral solution. AbbVie Inc. (per FDA), North Chicago, IL, 2015.
    326) Product Information: DEPAKENE(R) oral capsules, oral solution, valproic acid oral capsules, oral solution. Abbott Laboratories, North Chicago, IL, 2007.
    327) Product Information: DEPAKOTE(R) Sprinkle Capsules oral delayed release capsules, divalproex sodium oral delayed release capsules. AbbVie Inc. (per FDA), North Chicago, IL, 2015.
    328) Product Information: Depacon intravenous injection, valproate sodium intravenous injection . AbbVie Inc. (per Manufacturer), North Chicago, IL, 2013.
    329) Product Information: Depacon intravenous injection, valproate sodium intravenous injection. AbbVie Inc. (per FDA), North Chicago, IL, 2015.
    330) Product Information: Depakene(R), valproic acid. Abbott Laboratories, North Chicago, IL, 1990.
    331) Product Information: Depakote ER oral extended release tablets, divalproex sodium oral extended release tablets. AbbVie Inc. (per Manufacturer), North Chicago, IL, 2013.
    332) Product Information: Depakote ER oral extended-release tablets, divalproex sodium oral extended-release tablets. AbbVie Inc. (per FDA), North Chicago, IL, 2015.
    333) Product Information: Depakote oral tablets, divalproex sodium oral tablets. AbbVie Inc. (per FDA), North Chicago, IL, 2015.
    334) Product Information: Depakote oral tablets, divalproex sodium oral tablets. AbbVie Inc. (per FDA), North Chicago, IL, 2015a.
    335) Product Information: Depakote oral tablets, divalproex sodium oral tablets. AbbVie Inc. (per Manufacturer), North Chicago, IL, 2013.
    336) Product Information: Depakote(R), divalproex. Abbott Laboratories, North Chicago, IL, 1990.
    337) Product Information: STAVZOR(R) delayed release oral capsules, valproic acid delayed release oral capsules. Noven Therapeutics, LLC, Miami, FL, 2008.
    338) Product Information: STAVZOR(R) oral delayed release capsules, valproic acid oral delayed release capsules. Noven Therapeutics, LLC (per FDA), Miami, FL, 2014.
    339) Product Information: Stavzor(R) oral delayed release capsules, valproic acid oral delayed release capsules. Noven Therapeutics, LLC (per FDA), Miami, FL, 2013.
    340) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    341) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    342) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2002; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    343) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 7/31/1996; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    344) Rahman MH, Haqqie SS, & McGoldrick MD: Acute hemolysis with acute renal failure in a patient with valproic acid poisoning treated with charcoal hemoperfusion. Hemodial Int 2006; 10(3):256-259.
    345) Rajantie J, Kajosaari M, & Ylitalo V: Fatal pancytopenia during high-dose valproate monotherapy (letter). Eur J Pediatr 1992; 151:619.
    346) Ramsay RE, Cantrell D, Collins SD, et al: Safety and tolerance of rapidly infused Depacon. A randomized trial in subjects with epilepsy. Epilepsy Res 2003; 52(3):189-201.
    347) Raskind JY & El-Chaar GM: The role of carnitine supplementation during valproic acid therapy. Ann Pharmacother 2000; 34:630-638.
    348) Rattya J, Turkka J, & Pakarinen AJ: Reproductive effects of valproate, carbamazepine, and oxcarbazepine in men with epilepsy. Neurology 2001; 56:31-36.
    349) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    350) Rawat S, Borkowski WJ Jr, & Swick HM: Valproic acid and secondary hyperammonemia. Neurol 1981; 31:1173-1174.
    351) Raworth RE & Birchall G: Sodium valproate and platelet count. Lancet 1978; 1:670-671.
    352) Redington K, Wells C, & Petito F: Erythromycin and valproate interaction (letter). Ann Intern Med 1992; 116:877-878.
    353) Rigamonti A, Lauria G, Grimod G, et al: Valproate induced hyperammonemic encephalopathy successfully treated with levocarnitine. J Clin Neurosci 2014; 21(4):690-691.
    354) Rimmer EM & Richens A: An update on sodium valproate. Pharmacotherapy 1985; 5:171-184.
    355) Ritter EJ, Scott WJ, & Randall JL: Teratogenicity of Di(2-ethylhexyl)phthalate, 2-ethylhexanol, 2-ethylhexanoic acid, and valproic acid, andpotentiation by caffeine. Teratology 1987; 35:41-46.
    356) Roberge RJ & Francis EH: Use of naloxone in valproic acid overdose: case report and review. J Emerg Med 2002; 22:67-70.
    357) Robert E, Robert JM, & Lapras C: L'acide valproique est-il teratogene?. Rev Neurol (Paris) 1983; 139:445-447.
    358) Robert E: Valproic acid as a human teratogen. Cong Anom 1988; 28:571-580.
    359) Robinson P & Abbott C: Severe hypothermia in association with sodium valproate overdose. N Z Med J 2005; 118(1223):U1681-.
    360) Roodhooft AM, Van Dam K, & Haentjens D: Acute sodium valproate intoxication: occurrence of renal failure and treatment with haemoperfusion-haemodialysis. Eur J Pediatr 1990; 149:363-364.
    361) Roste LS, Tauboll E, & Berner A: Valproate, but not lamotrigine, induces ovarian morphological changes in Wistar rats. Exp Toxicol Pathol 2001; 52:545-552.
    362) Rupasinghe J & Jasinarachchi M: Progressive encephalopathy with cerebral oedema and infarctions associated with valproate and diazepam overdose. J Clin Neurosci 2011; 18(5):710-711.
    363) Ruskosky D, Holmes C, & Schauben J: Utilization of hemodialysis to enhance valproic acid (VPA) elimination (abstract). J Toxicol-Clin Toxicol 1999; 37:636-637.
    364) Russell S: Carnitine as an antidote for acute valproate toxicity in children. Curr Opin Pediatr 2007; 19(2):206-210.
    365) Rzany B, Correia O, & Kelly JP: Risk of Stevens-Johnson syndrome and toxic epidermal necrolysis during first weeks of antiepileptic therapy: a case control study. Lancet 1999; 353:2190-2194.
    366) Sandler RM, Ememrson C, & Roberts GE: IgM platelet antibody due to sodium valproate. Br Med J 1978; 2:1683-1684.
    367) SantosdeOliveira R, Lajeunie E, Arnaud E, et al: Fetal exposure to sodium valproate associated with Baller-Gerold syndrome: case report and review of the literature. Childs Nerv Syst 2006; 22(1):90-94.
    368) Sato M, Shirota M, & Nagao T: Pantothenic acid decreases valproic acid-induced neural tube defects in mice .1. Teratology 1995; 52:143-148.
    369) Schnabel R, Rambeck B, & Janssen F: Fatal intoxication with sodium valproate. Lancet 1984; 1:221-222.
    370) Serdaroglu G, Tutuncuoglu S, & Kavakh K: Coagulation abnormalities and acquired von Willebrand's disease type 1 in children receiving valproic acid. J Child Neurol 2002; 17(1):41-43.
    371) Sharma AN, Ilamathi E, & Nelson LS: High-efficiency hemodialysis is effective in removing valproic acid (abstract). J Toxicol-Clin Toxicol 2001; 39:497-498.
    372) Shen MR & Chen SS: The inhibitory effect of anticonvulsants on human sperm motility: measured with a trans-membrane migration method. Kao-Hsiung I Hsueh Ko Hseueh Tsa Chih 1990; 6:295-301.
    373) Shill HA & Fife TD: Valproic acid toxicity mimicking multiple system atrophy. Neurology 2000; 55:1936-1937.
    374) Sikma MA, Mier JC, & Meulenbelt J: Massive valproic acid overdose, a misleading case. Am J Emerg Med 2008; 26(1):110-116.
    375) Simon D & Penry JK: Sodium di-n-propylacetate (DPA) in the treatment of epilepsy. Epilepsia 1975; 16:549-573.
    376) Singh SM, McCormick BB, Musata S, et al: Extracorporeal management of valproic acid overdose: a large regional experience. J Nephrol 2004; 17:43-49.
    377) Sleiman C, Raffy O, & Roue C: Fatal pulmonary hemorrhage during high-dose valproate monotherapy (letter). Chest 2000; 117:613.
    378) Smith SW, Ling LJ, & Halstenson CE: Whole-bowel irrigation as a treatment for acute lithium overdose. Ann Emerg Med 1991; 20:536-539.
    379) Spiller HA & Krenzelok EP: Are there any unique valproic acid ingestions? (letter). Clin Toxicol 2001; 39:421-422.
    380) Spiller HA & Rogers GC: Evaluation of administration of activated charcoal in the home. Pediatrics 2002; 108:E100.
    381) Spiller HA, Krenzelok EP, & Klein-Schwartz W: Multicenter case series of valproic acid ingestion: serum concentrations and toxicity. Clin Toxicol 2000; 38:755-760.
    382) Stahl MMS, Neiderud J, & Vinge E: Thrombocytopenic purpura and anemia in a breast-fed infant whose mother was treated with valproic acid. J Pediatr 1997; 130:1001-1003.
    383) Stecker MM & Kita M: Paradoxical response to valproic acid in a patient with a hypothalamic hamartoma. Ann Pharmacother 1998; 32:1168-1172.
    384) Steiman GS, Woerpel RW, & Sherard ES: Treatment of accidental sodium valproate overdose with an opiate antagonist. Ann Neurol 1979; 6:274.
    385) Su M, Fong J, Howland MA, et al: Multiple-dose activated charcoal used to treat valproic acid overdose (abstract). J Toxicol Clin Toxicol 2002; 40(3):381-382.
    386) Suchy FJ, Balistreri WF, & Buchino JJ: Acute hepatic failure associated with the use of sodium valproate. Report of two fatal cases. N Engl J Med 1979; 300:962-966.
    387) Susanto F & Reinauer H: Candidate reference method - determination of valproic acid in serum by isotope-dilution mass spectrometry. Chromatographia 1995; 41:407-413.
    388) Sveberg Roste L, Tauboll E, & Berner A: Morphological changes in the testis after long-term valproate treatment in male Wistar rats. Seizure 2001; 10:559-565.
    389) Symon DNK & Russell G: Arch Dis Child 1983; 58:235.
    390) Sztajnkrycer MD, Scaglione JM, & Bond GR: Valproate-induced hyperammonemia: preliminary evaluation of ammonia elimination with carnitine administration (abstract). J Toxicol-Clin Toxicol 2001; 39:497.
    391) Sztajnkrycer MD: Valproic acid toxicity: overview and management. J Toxicol Clin Toxicol 2002; 40(6):789-801.
    392) Taneja N, Kucheria K, & Jain S: Sister chromatid exchanges are increased in epileptics but not by sodium valproate. Mutation Res 1992; 283:233-235.
    393) Tank JE & Palmer BF: Simultaneous "in series" hemodialysis and hemoperfusion in the management of valproic acid overdose. Am J Kid DIs 1993; 22:341-344.
    394) Tenenbein M, Cohen S, & Sitar DS: Whole bowel irrigation as a decontamination procedure after acute drug overdose. Arch Int Med 1987; 147:905-907.
    395) Thabet H, Brahmi N, & Amamou M: Hyperlactatemia and hyperammonemia as secondary effects of valproic acid poisoning (letter). Am J Emerg Med 2000; 18:508.
    396) Thakore S & Murphy N: The potential role of prehospital administration of activated charcoal. Emerg Med J 2002; 19:63-65.
    397) Thanacoody HK: Chronic valproic acid intoxication: reversal by naloxone. Emerg Med J 2007; 24(9):677-678.
    398) Thole D, Bagnasco T, & Lovecchio F: Delayed absorption of valproic acid resulting in coma (abstract). J Toxicol-Clin Toxicol 2001; 39:497.
    399) Thruston JH, Hauhart RE, & Schulz DW: Chronic valproate administration produces hepatic dysfunction and may delay brain maturation in infant mice. Neurology 1981; 31:1063-1069.
    400) Tift JP: Valproic acid (letter). N Engl J Med 1980; 303:394.
    401) Tohen M, Castillo J, & Baldessarini RJ: Blood dyscrasias with carbamazepine and valproate: a pharmacoepidemiological study of 2,228 patients at risk. Am J Psychiatry 1995; 152:413-418.
    402) Turnbull DM, Rawlins MD, & Weightman D: Plasma concentrations of sodium valproate: their clinical value. Ann Neurol 1983; 14:38-42.
    403) U.S. Department of Energy, Office of Emergency Management: Protective Action Criteria (PAC) with AEGLs, ERPGs, & TEELs: Rev. 26 for chemicals of concern. U.S. Department of Energy, Office of Emergency Management. Washington, DC. 2010. Available from URL: http://www.hss.doe.gov/HealthSafety/WSHP/Chem_Safety/teel.html. As accessed 2011-06-27.
    404) U.S. Department of Health and Human Services, Public Health Service, National Toxicology Project : 11th Report on Carcinogens. U.S. Department of Health and Human Services, Public Health Service, National Toxicology Program. Washington, DC. 2005. Available from URL: http://ntp.niehs.nih.gov/INDEXA5E1.HTM?objectid=32BA9724-F1F6-975E-7FCE50709CB4C932. As accessed 2011-06-27.
    405) U.S. Environmental Protection Agency: Discarded commercial chemical products, off-specification species, container residues, and spill residues thereof. Environmental Protection Agency's (EPA) Resource Conservation and Recovery Act (RCRA); List of hazardous substances and reportable quantities 2010b; 40CFR(261.33, e-f):77-.
    406) U.S. Environmental Protection Agency: Integrated Risk Information System (IRIS). U.S. Environmental Protection Agency. Washington, DC. 2011. Available from URL: http://cfpub.epa.gov/ncea/iris/index.cfm?fuseaction=iris.showSubstanceList&list_type=date. As accessed 2011-06-21.
    407) U.S. Environmental Protection Agency: List of Radionuclides. U.S. Environmental Protection Agency. Washington, DC. 2010a. Available from URL: http://www.gpo.gov/fdsys/pkg/CFR-2010-title40-vol27/pdf/CFR-2010-title40-vol27-sec302-4.pdf. As accessed 2011-06-17.
    408) U.S. Environmental Protection Agency: List of hazardous substances and reportable quantities. U.S. Environmental Protection Agency. Washington, DC. 2010. Available from URL: http://www.gpo.gov/fdsys/pkg/CFR-2010-title40-vol27/pdf/CFR-2010-title40-vol27-sec302-4.pdf. As accessed 2011-06-17.
    409) U.S. Environmental Protection Agency: The list of extremely hazardous substances and their threshold planning quantities (CAS Number Order). U.S. Environmental Protection Agency. Washington, DC. 2010c. Available from URL: http://www.gpo.gov/fdsys/pkg/CFR-2010-title40-vol27/pdf/CFR-2010-title40-vol27-part355.pdf. As accessed 2011-06-17.
    410) U.S. Food and Drug Administration (FDA): FDA Drug Safety Communication: Children born to mothers who took Valproate products while pregnant may have impaired cognitive development. U.S. Food and Drug Administration (FDA). Rockvillle, MD. 2011. Available from URL: http://www.fda.gov/Drugs/DrugSafety/ucm261543.htm. As accessed 2011-06-30.
    411) U.S. Occupational Safety and Health Administration: Part 1910 - Occupational safety and health standards (continued) Occupational Safety, and Health Administration's (OSHA) list of highly hazardous chemicals, toxics and reactives. Subpart Z - toxic and hazardous substances. CFR 2010 2010; Vol6(SEC1910):7-.
    412) U.S. Occupational Safety, and Health Administration (OSHA): Process safety management of highly hazardous chemicals. 29 CFR 2010 2010; 29(1910.119):348-.
    413) US Food and Drug Administration: FDA Drug Safety Communication: Valproate Anti-seizure Products Contraindicated for Migraine Prevention in Pregnant Women due to Decreased IQ Scores in Exposed Children. US Food and Drug Administration. Silver Springs, MD. 2013. Available from URL: http://www.fda.gov/downloads/Drugs/DrugSafety/UCM350748.pdf. As accessed 05/06/2013.
    414) US Food and Drug Administration: Information for Healthcare Professionals: Risk of Neural Tube Birth Defects following prenatal exposure to Valproate. US Food and Drug Administration. Silver Spring, MD. 2009. Available from URL: http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyInformationforHeathcareProfessionals/ucm192649.htm. As accessed 2009-12-03.
    415) Uberall MA, Trollmann R, Wunsiedler U, et al: Intravenous valproate in pediatric epilepsy patients with refractory status epilepticus. Neurology 2000; 54(11):2188-2189.
    416) Unal E, Kaya U, & Aydin K: Fatal valproate overdose in a newborn baby. Hum Exp Toxicol 2007; 26(5):453-456.
    417) United States Environmental Protection Agency Office of Pollution Prevention and Toxics: Acute Exposure Guideline Levels (AEGLs) for Vinyl Acetate (Proposed). United States Environmental Protection Agency. Washington, DC. 2006. Available from URL: http://www.regulations.gov/search/Regs/contentStreamer?objectId=090000648020d6af&disposition=attachment&contentType=pdf. As accessed 2010-08-16.
    418) Vale JA, Kulig K, American Academy of Clinical Toxicology, et al: Position paper: Gastric lavage. J Toxicol Clin Toxicol 2004; 42:933-943.
    419) 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.
    420) Van der Merwe AC, Albrecht CF, & Brink MS: Sodium valproate poisoning. S Afr Med J 1985; 67:735.
    421) Venkataraman V & Wheless JW: Safety of rapid intravenous infusion of valproate loading doses in epilepsy patients. Epilepsy Res 1999; 35(2):147-153.
    422) Verloes A, Frikiche A, & Fremillet C: Proximal phocomelia and radial ray aplasia in fetal valproic syndrome. European J Ped 1990; 149:266-267.
    423) Volzke E & Doose H: Diapropyl acetate, (Depakine, Ergenyl) in the treatment of epilepsy. Epilepsia 1973; 14:185.
    424) Von Uhruh GE, Froescher W, Hoffmann F, et al: Valproic acid in breast milk: How much is really there?. Ther Drug Monitor 1984; 6:272-276.
    425) Von Voss H: Sodium valproate and platelet function. Br Med J 1976; 2:179.
    426) Vorhees CV: Teratogenicity and developmental toxicity of valproic acid in rats. Teratology 1987; 35:195-202.
    427) Wagner KD, Redden L, Kowatch RA, et al: A double-blind, randomized, placebo-controlled trial of divalproex extended-release in the treatment of bipolar disorder in children and adolescents. J Am Acad Child Adolesc Psychiatry 2009; 48(5):519-532.
    428) Wagner KD, Weller EB, Carlson GA, et al: An open-label trial of divalproex in children and adolescents with bipolar disorder. J Am Acad Child Adolesc Psychiatry 2002; 41(10):1224-1230.
    429) Wasserman GS, Aldridge SC, & Kelly JC: A unique valproic acid ingestion (letter). Clin Toxicol 2001; 39:419-420.
    430) Watkins JR, Gough AW, & McGuire EJ: Calcium valproate - induced on SCE and chromosome aberrations in epileptic children. Mutation Res 1990; 243:63-66. Watkins JR, Gough AW, McGuire EJ et al: Calcium valproate - induced uterine adenocarcinomas in Wistar rats. Toxicology 1992; 71:35-47.
    431) Weiner D, Nir V, Klein-Kremer A, et al: Chronic valproic acid intoxication. Pediatr Emerg Care 2013; 29(6):756-757.
    432) Wilimowska J, Florek E, & Piekoszewski W: Disposition of valproic acid in self-poisoned adults. Basic Clin Pharmacol Toxicol 2006; 99(1):22-26.
    433) Williams SR & Clark RF: Hemodialysis of a valproic acid poisoning (abstract). J Toxicol Clin Toxicol 1995; 33:491.
    434) Willmore LJ: Neurology 1978; 28:961.
    435) Wisner KL & Perel JM: Serum levels of valproate and carbamazepine in breastfeeding mother-infant pairs. J Clin Psychopharmacol 1998; 18:167-169.
    436) Wu S, Legido A, & DeLuca F: Effects of valproic acid on longitudinal bone growth. J Child Neurol 2004; 19(1):26-30.
    437) Wyllie E, Wyllie R, & Cruse RP: Pancreatitis associated with valproic acid therapy. Am J Dis Child 1984; 138:912-914.
    438) Wyllie E, Wyllie R, & Rothner AD: Valproate-induced hyperammonaemia in asymptomatic children. Cleve Clin Q 1983; 50:275-277.
    439) Wyllie E: Am J Dis Child 1984; 138:912.
    440) Xiong GL , Ferranti J , & Leamon MH : Toxic interaction between valproate and oxcarbazepine: a case detected by the free valproate level. J Clin Psychopharmacol 2008; 28(4):472-473.
    441) Yazdani K, Lippmann M, & Gala I: Fatal pancreatitis associated with valproic acid: review of the literature. Medicine 2002; 81:305-310.
    442) Yehya N, Saldarini CT, Koski ME, et al: Valproate-induced hyperammonemic encephalopathy. J Am Acad Child Adolesc Psychiatry 2004; 43(8):926-927.
    443) Yoshikawa H, Watanabe T, & Abe T: Tubulo-interstitial nephritis caused by sodium valproate. Brain Dev 2002; 24:102-105.
    444) Yu KT, Mills S, Thompson N, et al: Safety and efficacy of intravenous valproate in pediatric status epilepticus and acute repetitive seizures. Epilepsia 2003; 44(5):724-746.
    445) Ziyeh S, Thiel T, Spreer J, et al: Valproate-induced encephalopathy: Assessment with MR imaging and H MR spectroscopy. Epilepsia 2002; 43(9):1101-1105.
    446) de Laat SA, Hillegers MH, Jansen FE, et al: Hallucinations after withdrawal of valproic acid. Pediatrics 2012; 130(1):e236-e238.
    447) de Leon J, Kiesel JL, Fleming MW, et al: Valproic acid toxicity associated with low dose of aspirin and low total valproic acid levels: a case report. J Clin Psychopharmacol 2009; 29(5):509-511.
    448) van Keulen JG, van Wijk JAE, & Touw DJ: Effectiveness of haemofiltration in valproic acid intoxication (letter). Acta Paediatr 2001; 90:958-959.
    449) van Swieten JC, Koudstaal PJ, Visser MC, et al: Interobserver agreement for the assessment of handicap in stroke patients. Stroke 1988; 19(5):604-607.
    450) von Bardeleben RS, Sturer A, & Weilemann LS: Valproic acid intoxications in 96 documented cases 1995-1998: The role of hemoperfusion in therapy (abstract). J Toxicol-Clin Toxicol 1998; 36:471.