MOBILE VIEW  | 

PHENYTOIN

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Phenytoin is commonly used anticonvulsant, with toxicity occurring during acute (or long-term) oral ingestions and intravenous administration. The toxic cardiovascular effects of phenytoin injection are primarily related to the diluent, propylene glycol.
    B) Ethotoin is a hydantoin anticonvulsant which is chemically related to phenytoin.
    C) For further information on fosphenytoin see the FOSPHENYTOIN management.

Specific Substances

    A) PHENYTOIN
    1) DPH
    2) Diphenylhydantoin
    3) 5,5-Diphenylhydantoin
    4) 5,5-Diphenylimidazolidine-2,4-dione
    5) Hydantoin, 5,5-diphenyl-
    6) Molecular formula: C15-H12-N2-O2
    7) CAS 57-41-0; 125-59-7
    ETHOTOIN
    1) 3-Ethyl-5-phenylhydantoin
    2) Hydantoin, 3-ethyl-5-phenyl-
    3) Molecular Formula: C11-H12-N2-O2
    4) CAS 86-35-1

    1.2.1) MOLECULAR FORMULA
    1) C15-H12-N2-O2.Na

Available Forms Sources

    A) FORMS
    1) PHENYTOIN
    a) Phenytoin 50 mg chewable tablet (Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    b) Phenytoin oral suspension in concentrations of 125 mg/5 mL (Prod Info Dilantin-125(R) oral suspension, 2011).
    c) Phenytoin sodium extended-release capsules 30 mg and 100 mg (Prod Info DILANTIN(R) oral extended release capsules, 2011; Prod Info DILANTIN(R) extended capsule, oral, 2009).
    d) Phenytoin sodium parenteral solution 50 mg/mL (Prod Info Dilantin(R) intravenous injection solution, 2011).
    2) ETHOTOIN
    a) Ethotoin is available as 250 mg scored tablets (Prod Info PEGANONE(R) oral tablets, 2009).
    B) SOURCES
    1) Phenytoin sodium covers various anticonvulsant preparations, most notably Dilantin(R). Generic phenytoin preparations often exhibit erratic kinetics. Calculations based on kinetic parameters for Dilantin Kapseals(R) will frequently be wrong with overdoses of generic phenytoin.
    C) USES
    1) Phenytoin and ethotoin are used for the control of generalized tonic-clonic (grand mal) and complex partial (psychomotor, temporal lobe) seizures. Phenytoin is also used for the prevention and treatment of seizures which may occur during or following neurosurgery (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info Dilantin(R) intravenous injection solution, 2011; Prod Info PEGANONE(R) oral tablets, 2009).
    2) RECREATIONAL USE: There have been several reports in the literature of phenytoin being used as a recreational drug. Phenytoin was combined with crack cocaine and smoked (Jessen, 2004). In one case, a man ingested several crack cocaine rocks adulterated with phenytoin (Roldan, 2014). In one case, a young adult "sipped syrup" (phenytoin), smoked marijuana and drank alcohol to elicit a "high" and developed symptoms associated with phenytoin toxicity (Jessen, 2004).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) This management includes phenytoin and ethotoin, both in the hydantoin class of anticonvulsants; however, phenytoin will be the primary drug discussed. Fosphenytoin is covered in a separate management.
    B) USES: Phenytoin is primarily used as an anticonvulsant, both for status epilepticus and for seizure prevention.
    C) PHARMACOLOGY: Phenytoin stabilizes neuronal membranes and decreases seizure activity by increasing efflux or decreasing influx of sodium ions across cell membranes in the motor cortex during nerve impulse generation.
    D) TOXICOLOGY: Phenytoin prolongs effective refractory period and suppresses ventricular pacemaker automaticity, thus shortening the action potential in the heart. The intravenous form of phenytoin is dissolved in 40% propylene glycol and 10% ethanol at a pH of 12. This formulation can have its own toxicity secondary to cardiac toxicity of propylene glycol (mechanism unknown) and tissue necrosis from infiltration (secondary to the alkaline nature of the formulation).
    E) EPIDEMIOLOGY: There are thousands of exposures reported to poison centers every year, as it is a widely used anticonvulsant. However, deaths are extremely rare and severe manifestations occur in only a minority of cases.
    F) WITH THERAPEUTIC USE
    1) Significant adverse effects seen from intravenous use of phenytoin include venous irritation and pain, and thrombophlebitis. Extravasation can cause tissue necrosis. Adverse effects unrelated to plasma phenytoin levels include hypertrichosis, gingival hypertrophy, thickening of facial features, carbohydrate intolerance, folic acid deficiency, peripheral neuropathy, vitamin D deficiency, osteomalacia, and systemic lupus erythematosus (SLE). Some rarely seen but important adverse reactions include blood dyscrasias, dyskinesias, hepatitis, lymphadenopathy, lymphoma, pseudolymphoma, SLE-like syndrome, Steven-Johnson Syndrome, and toxic epidermal necrolysis.
    2) Phenytoin is a known teratogen. Fetal hydantoin syndrome is characterized by microcephaly, mental retardation, craniofacial abnormalities, and digital hypoplasia. It occurs in 10% to 30% of exposed pregnancies.
    G) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Nausea and vomiting may develop early after overdose. Nystagmus, ataxia, and mild CNS depression are common.
    2) SEVERE TOXICITY: Large oral ingestions can cause more severe CNS depression, coma, and, rarely, respiratory depression. Rapid infusion of the parenteral formulation (faster than 50 mg/min) can cause hypotension, bradycardia, AV conduction delays, and ventricular dysrhythmias which may be fatal. It is felt that the cardiotoxicity of the intravenous formulation of phenytoin is secondary to the diluent, propylene glycol, and not the phenytoin itself.
    0.2.20) REPRODUCTIVE
    A) Ethotoin, phenytoin, and phenytoin sodium have been classified as FDA pregnancy category D. Fetal hydantoin syndrome occurs in 10% to 30% of exposed pregnancies. It may result in multisystem anomalies including CNS dysfunction, craniofacial anomalies, major malformations, and nail/digital hypoplasia. If used during pregnancy or if patient becomes pregnant, weigh the risks and benefits of use during pregnancy, inform the patient of the potential harm to the fetus, and measure phenytoin plasma concentrations periodically. Prevent potentially life-threatening bleeding disorders in the infant by administering vitamin K to the mother before delivery and then to the neonate after birth. Breastfeeding while taking phenytoin is not recommended. However, phenytoin is considered to be compatible with breastfeeding by the American Academy of Pediatrics and the World Health Organization. Monitor infant for side effects.
    0.2.21) CARCINOGENICITY
    A) Phenytoin is a suspect human carcinogen. Elevated risks for Hodgkin's disease, lymphosarcomas, and reticulum-cell sarcoma have been seen in patients receiving phenytoin therapy (HSDB , 1995). Patients treated with anticonvulsants have shown an elevated risk for cancers of the brain and CNS, but clustering within 10 years of hospitalization suggests that pre-existing brain tumors may have been the cause of the seizure disorders for which the phenytoin was prescribed (Olsen et al, 1989).
    B) There is no evidence that phenytoin can cause liver cancer in humans from the 60 years of experience in its use; therefore the induction of liver tumors in mice is deemed not to be relevant to the human situation (Dethloff et al, 1996).

Laboratory Monitoring

    A) Obtain a basic metabolic panel, aspirin concentration, and acetaminophen concentration after deliberate overdose.
    B) Monitor phenytoin concentrations every 4 hours until clearly declining.
    C) Monitor serial neurologic examinations.
    D) Obtain an ECG and institute continuous cardiac monitoring during and after rapid intravenous infusion or intravenous overdose.
    E) Obtain serum albumin concentration as phenytoin toxicity may occur in the setting of normal total serum phenytoin concentrations, but elevated free phenytoin concentrations.
    F) Some clinical effects reported in elevated phenytoin concentrations include:
    1) Greater than 20 mcg/mL: far lateral nystagmus
    2) Greater than 30 mcg/mL: 45 degrees lateral gaze nystagmus and ataxia
    3) Greater than 40 mcg/mL: decreased mentation
    4) Greater than 100 mcg/mL: death

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) For mild and moderate toxicity, treat with supportive care, as the patient eventually metabolizes the phenytoin. If the patient is awake and alert, administer a dose of activated charcoal. Protect the patient from self-injury secondary to ataxia.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) For large phenytoin overdoses, treat with supportive care, which may include intubation for comatose patients. If seizures do occur, treat with benzodiazepines and evaluate for other causes of seizures.
    C) DECONTAMINATION
    1) PREHOSPITAL: Prehospital activated charcoal might be considered if the patient is awake, alert, and cooperative, and the exposure was relatively recent (within the last hour).
    2) HOSPITAL: Activated charcoal could be considered if the patient is awake, alert, and cooperative, and the ingestions is relatively recent. Gastric lavage should be avoided in most phenytoin overdoses as it is not life-threatening.
    D) AIRWAY MANAGEMENT
    1) Perform endotracheal intubation in patients with significant CNS depression.
    E) ANTIDOTE
    1) None
    F) EXTRAVASATION INJURY
    1) If extravasation occurs, stop the infusion. Disconnect the IV tubing, but leave the cannula or needle in place. Attempt to aspirate the extravasated drug from the needle or cannula. If possible, withdraw 3 to 5 mL of blood and/or fluids through the needle/cannula. Administer hyaluronidase. Elevate the affected area. Apply a warm or cold compress as indicated. One study recommended applying a dry warm compress. However, another source recommended ice packs for 15 to 20 minutes at least 4 times daily. Administer analgesics for severe pain. If pain persists, there is concern for compartment syndrome, or injury is apparent, an early surgical consult should be considered. Close observation of the extravasated area is suggested. If tissue sloughing, necrosis or blistering occurs, treat as a chemical burn (ie, antiseptic dressings, silver sulfadiazine, antibiotics when applicable). Surgical or enzymatic debridement may be required. Risk of infection is increased in chemotherapy patients with reduced neutrophil count following extravasation. Consider culturing any open wounds. Monitor the site for the development of cellulitis, which may require antibiotic therapy.
    G) ENHANCED ELIMINATION
    1) There is some evidence that repeated doses of charcoal may enhance phenytoin elimination, but the evidence is weak that actually improves outcomes, and it should not be given in drowsy patients secondary to the risk of aspiration pneumonitis, so it is rarely indicated. In a systematic review of literature that included 51 studies, including 30 case reports/case series (31 patients), 17 pharmacokinetic studies (54 patients), 1 animal experiment, and 3 in vitro studies, the Extracorporeal Treatments in Poisoning (EXTRIP) workgroup (included international experts) concluded that phenytoin is moderately dialyzable, despite its high protein binding, and recommended the use of ECTR in selected patients with severe phenytoin toxicity. Despite a low quality of evidence for all recommendations, the following guideline was developed: ECTR is suggested if prolonged coma is present or expected. ECTR is reasonable if prolonged incapacitating ataxia is present or expected. ECTR is NOT recommended solely based on suspected ingested dose of phenytoin or based on serum phenytoin concentration. Discontinuation of ECTR is recommended in patients with apparent clinical improvement. The preferred ECTR is intermittent hemodialysis, with an acceptable alternative being intermittent hemoperfusion, if hemodialysis is not available.
    H) PATIENT DISPOSITION
    1) HOME CRITERIA: For unintentional ingestions where the patient is asymptomatic and the dose is less than 20 mg/kg, the patient can be observed at home.
    2) OBSERVATION CRITERIA: Any intentional overdose or inadvertent overdoses above 20 mg/kg, or if symptomatic, the patient should be sent to a healthcare facility for observation. Patients should be observed for at least 4 to 6 hours, and should not be sent home until symptoms are clearly improving, or they are asymptomatic and serum concentrations are clearly declining.
    3) ADMISSION CRITERIA: Patients who have worsening symptoms after 4 to 6 hours or with significant ataxia or CNS depression, and those with rising serum concentrations, should be admitted to the hospital. If the CNS depression is severe enough that there is respiratory depression or concern of airway protection, the patient should be admitted to an intensive care setting. Otherwise, most patients can be safely admitted to a hospital ward bed. Criteria for discharge from the hospital should include clinical symptomatic improvement, which should be reflected by declining phenytoin serum concentrations.
    4) CONSULT CRITERIA: In patients with cardiac toxicity secondary to the intravenous formulation of phenytoin, it might be reasonable to consult a cardiologist. Neurologists are often very familiar with both the acute and chronic toxicity of phenytoin. Consult a medical toxicologist or poison center for patients with severe toxicity.
    I) PITFALLS
    1) Intravenous phenytoin should NEVER be infused rapidly (greater than 40 to 50 mg/min or more than 0.5 to 1 mg/kg/min) as this may cause myocardial depression and cardiac arrest. Since phenytoin is protein bound, patients with decreased protein binding have greater concentrations of free phenytoin and therefore greater toxicity at a given serum concentration. Two types of conditions cause decreased phenytoin protein binding: disease states that cause a decrease in serum albumin concentrations (ie, burns, hepatic cirrhosis, nephrotic syndrome, pregnancy, cystic fibrosis) and diseases that results in apparent decrease in affinity of phenytoin for serum albumin (ie, renal failure, severe jaundice, other medications, hyperbilirubinemia (total bilirubin greater than 15 mg/dL), and uremia (creatinine clearance less than 25 mL/minute with the unbound fraction increased two- to three-fold in uremia)). Small changes in dose can cause rapidly increasing serum concentrations and toxicity secondary to saturable metabolism.
    J) PHARMACOKINETICS
    1) Onset of action from intravenous dosing is immediate, while oral absorption is quite variable and slow. The volume of distribution following single IV doses in children (9.4 to 21.3 mg/kg) was 0.95 L/kg and declined with age, with a range of 1 to 1.5 L/kg below the age of 5 years to 0.6 to 0.8 L/kg above the age of 8 years. In adults, the volume of distribution ranges from 0.6 to 0.7 L/kg. Protein binding in adults is greater than 90%. Metabolism is hepatic and is dose-dependent and capacity-limited (Michaelis-Menten) pharmacokinetics. Major metabolites undergo enterohepatic recirculation. Less than 5% is excreted unchanged in the urine. Time to peak concentrations is 2 to 3 hours after immediate-release and 4 to 12 hours after extended-release formulations. Half-life of elimination ranges widely, from 7 to 42 hours, with an average of 22 hours.
    K) TOXICOKINETICS
    1) Phenytoin metabolism is saturable at high concentrations, leading to zero order elimination and very prolonged half-life. In large overdoses, patients may be symptomatic for days.
    L) PREDISPOSING CONDITIONS
    1) Patients with low albumin levels will have an increase in the free fraction of serum phenytoin concentrations and hence will have an increased pharmacologic response at the same total phenytoin concentration. Phenytoin is metabolized by several p450 enzymes and thus drugs that interact with p450 enzymes may cause changes in phenytoin concentrations. In addition, drugs that are highly protein-bound, such as warfarin or thyroid hormone, may compete with phenytoin for albumin binding and thus increase the concentration of free phenytoin.
    M) DIFFERENTIAL DIAGNOSIS
    1) The differential diagnosis for this ingestion includes other sedating medications, such as other antiepileptics or benzodiazepines, barbiturates, or opioids.
    0.4.6) PARENTERAL EXPOSURE
    A) In patients with parenteral overdoses, monitor mental status and intubate patients with significant CNS depression. Treat hypotension with intravenous fluids, add vasopressors if hypotension persists. Treat dysrhythmias with standard ACLS protocols. Thrombophlebitis and infiltrations should be treated supportively with pain medications such as NSAIDs and warm compresses.

Range Of Toxicity

    A) TOXICITY: The minimum acute toxic oral overdose is 20 mg/kg. However, since phenytoin has dose-dependent kinetics, inadvertent intoxication from drug interactions or small dose adjustments can easily occur in patients taking the medication chronically. Fatalities have most often been associated with the intravenous formulation being given too rapidly, and is thought to be secondary to the propylene glycol diluent rather than the phenytoin itself.
    B) THERAPEUTIC DOSE: SEIZURE DISORDER: ADULT: Usual maintenance dose is 100 mg orally 3 to 4 times daily, up to a maximum of 600 mg/day. PEDIATRIC: Children less than 6 years of age: 5 mg/kg/day divided into 2 or 3 doses up to a maximum of 300 mg/day; usual maintenance dose is 4 to 8 mg/kg/day. Children older than 6 years of age: May require the minimum adult dose of 300 mg/day. STATUS EPILEPTICUS: ADULT: Loading dose is between 10 to 15 mg/kg IV with a maximum rate of 50 mg/minute. PEDIATRIC: The recommended loading dose is 15 to 20 mg/kg IV at a rate not exceeding 1 to 3 mg/kg/min or 50 mg/min (whichever is slower).

Summary Of Exposure

    A) This management includes phenytoin and ethotoin, both in the hydantoin class of anticonvulsants; however, phenytoin will be the primary drug discussed. Fosphenytoin is covered in a separate management.
    B) USES: Phenytoin is primarily used as an anticonvulsant, both for status epilepticus and for seizure prevention.
    C) PHARMACOLOGY: Phenytoin stabilizes neuronal membranes and decreases seizure activity by increasing efflux or decreasing influx of sodium ions across cell membranes in the motor cortex during nerve impulse generation.
    D) TOXICOLOGY: Phenytoin prolongs effective refractory period and suppresses ventricular pacemaker automaticity, thus shortening the action potential in the heart. The intravenous form of phenytoin is dissolved in 40% propylene glycol and 10% ethanol at a pH of 12. This formulation can have its own toxicity secondary to cardiac toxicity of propylene glycol (mechanism unknown) and tissue necrosis from infiltration (secondary to the alkaline nature of the formulation).
    E) EPIDEMIOLOGY: There are thousands of exposures reported to poison centers every year, as it is a widely used anticonvulsant. However, deaths are extremely rare and severe manifestations occur in only a minority of cases.
    F) WITH THERAPEUTIC USE
    1) Significant adverse effects seen from intravenous use of phenytoin include venous irritation and pain, and thrombophlebitis. Extravasation can cause tissue necrosis. Adverse effects unrelated to plasma phenytoin levels include hypertrichosis, gingival hypertrophy, thickening of facial features, carbohydrate intolerance, folic acid deficiency, peripheral neuropathy, vitamin D deficiency, osteomalacia, and systemic lupus erythematosus (SLE). Some rarely seen but important adverse reactions include blood dyscrasias, dyskinesias, hepatitis, lymphadenopathy, lymphoma, pseudolymphoma, SLE-like syndrome, Steven-Johnson Syndrome, and toxic epidermal necrolysis.
    2) Phenytoin is a known teratogen. Fetal hydantoin syndrome is characterized by microcephaly, mental retardation, craniofacial abnormalities, and digital hypoplasia. It occurs in 10% to 30% of exposed pregnancies.
    G) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Nausea and vomiting may develop early after overdose. Nystagmus, ataxia, and mild CNS depression are common.
    2) SEVERE TOXICITY: Large oral ingestions can cause more severe CNS depression, coma, and, rarely, respiratory depression. Rapid infusion of the parenteral formulation (faster than 50 mg/min) can cause hypotension, bradycardia, AV conduction delays, and ventricular dysrhythmias which may be fatal. It is felt that the cardiotoxicity of the intravenous formulation of phenytoin is secondary to the diluent, propylene glycol, and not the phenytoin itself.

Vital Signs

    3.3.3) TEMPERATURE
    A) WITH POISONING/EXPOSURE
    1) HYPOTHERMIA: A case of phenytoin toxicity manifesting solely in the form of hypothermia has been reported (Alhaj & Alhaj, 2001). Hypothermia resolved following discontinuation of phenytoin and use of warming blankets. Lowry et al (2005) reported a one-month-old infant with hypothermia (92 degrees F rectally) and pink mottled skin following an unintentional overdose of oral phenytoin (Lowry et al, 2005).

Heent

    3.4.3) EYES
    A) WITH POISONING/EXPOSURE
    1) CATARACT
    a) CASE REPORT: Subcapsular cataract formation was associated with ingestion of 400 mg/day of phenytoin over several weeks producing a toxic phenytoin concentration of 17.4 mcg/mL in a 26-year-old man (Mathers et al, 1987).
    2) NYSTAGMUS
    a) Nystagmus is commonly seen with toxic concentrations of phenytoin (Roldan, 2014; Kumar et al, 2012; Jessen, 2004; Kozer et al, 2002; Turkdogan et al, 2002; Brandolese et al, 2001; Lau et al, 2000; Grant & Schuman, 1993a). Absent nystagmus is a significant negative finding.
    b) CASE REPORT: Both vertical and horizontal nystagmus were present 9 hours following an ingestion of 100 capsules of phenytoin 100 mg in a 21-year-old woman (Griffiths et al, 1987).
    c) CASE REPORT: Periodic alternating nystagmus was reported in a 47-year-old chronic alcoholic receiving phenytoin 300 mg/day after an extra 1200 mg within 24 hours (Campbell, 1980).
    d) CASE REPORT: Dysconjugate gaze and absent doll's eye movements were reported in a one-month-old infant following an unintentional overdose of oral phenytoin (40 mg 3 times/day for 10 days) (Lowry et al, 2005).
    3) COLOR VISION DEFICIENCY
    a) CASE REPORT: Xanthopsia and blurred vision were reported in a 19-year-old man with a history of seizures 2 days after receiving a phenytoin loading dose of 1000 mg over 20 minutes followed by a maintenance dose of 300 mg/day. Neurologic function remained normal. Visual symptoms gradually resolved with no permanent effects following drug withdrawal (Thakral et al, 2003).
    b) Bayer et al (1997) report blue-yellow color vision deficiencies in patients with signs of phenytoin-induced neurotoxicity, with no correlation to phenytoin serum concentrations(Bayer et al, 1997).
    4) OPHTHALMOPLEGIA
    a) Nearly complete external ophthalmoplegia was reported with a rapid onset following overdose and a duration of one day (Grant & Schuman, 1993a).
    5) OCULAR MOTOR ABNORMALITIES
    a) CASE REPORT: A 55-year-old man, who was taking phenytoin sodium (300 mg/day) for generalized tonic clonic seizures, developed ocular motor disturbances, simulating bilateral internuclear ophthalmoplegia (INO), after ingesting about 2.5 g of phenytoin sodium. The cranial nerves evaluation showed mid-positioned eyes with bilateral 4 mm sized round pupils with preserved direct and consensual light reflexes. He had forwardly fixed gaze, could not follow moving objects, or move his eyes on command. Laboratory result revealed a serum phenytoin concentration of 77.9 mcg/mL (therapeutic range 20 to 40 mcg/mL). His symptoms gradually improved as his serum phenytoin levels decreased. On day 11, his eye movements were normal and his serum phenytoin concentration was 23.4 mcg/mL. He was discharged 14 days after presentation (Praveen-kumar & Desai, 2014).
    b) OSCILLOPSIA, a very fine vertical or horizontal periodic dancing of the eyes, has been noted (Grant & Schuman, 1993a).
    6) DIPLOPIA
    a) Diplopia may occur (Roldan, 2014; Grillone & Myssiorek, 1992).
    3.4.6) THROAT
    A) WITH THERAPEUTIC USE
    1) GINGIVAL HYPERPLASIA
    a) Gingival hyperplasia has been reported with phenytoin therapy (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011), occurring in 21% of children receiving folic acid to 88% of children with no folic-acid supplementation (Arya et al, 2011). It occurs within 2 to 3 months, peak severity is at 9 to 12 months (Grillone & Myssiorek, 1992a), and is often severe in young children. The degree of severity of drug-induced gingival hyperplasia has been correlated to increased dose and phenytoin serum levels, decreased age and weight of the patients, and poor oral hygiene (Addy et al, 1983). Folic acid supplementation is associated with prevention of gingival overgrowth in children taking phenytoin monotherapy (Arya et al, 2011).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) BRADYCARDIA
    1) WITH THERAPEUTIC USE
    a) Phenytoin administered intravenously at rates faster than 50 mg/minute may produce significant bradycardia and hypotension which can lead to atrial and then ventricular asystole (due to the diluent, propylene glycol) (Prod Info Dilantin(R) intravenous injection solution, 2011).
    b) While most cases are reported in older patients with known cardiac disease, bradycardia, hypotension, and dysrhythmias have been observed in young healthy volunteers at acceptable infusion rates (50 mg/minute or less) (Barron, 1976; Earnest et al, 1983).
    c) Complications will reportedly be minimized if IV phenytoin is administered in concentrations of 6.7 mg/mL or less and at rates of 40 mg/minute or less (Earnest et al, 1983).
    B) CARDIOTOXICITY
    1) WITH THERAPEUTIC USE
    a) CHRONIC THERAPY: A 35-year-old man with a history of traumatic brain injury, epilepsy and pulmonary tuberculosis in a vegetative state, developed severe cardiotoxicity (i.e., junctional bradycardia, BP 87/75 mmHg) after receiving phenytoin 200 mg twice daily for approximately 5 months. Initial serum phenytoin was 91 mcg/mL, serum sodium was 121 mEq/L and serum potassium was 5.4 mEq/L. The patient was hemodynamically stabilized with temporary transvenous pacing, fluid resuscitation and inotropes. Electrolytes were corrected. ECG returned to normal sinus rhythm by day 3 with a phenytoin concentration of 67.3 mcg/mL. The patient gradually improved to baseline with intensive supportive care and the pacemaker was removed on day 13; phenytoin concentration was 2.65 mcg/mL (Su et al, 2009). The authors suggested that underlying metabolic derangements and a possible drug interaction with an anti-TB medication likely contributed to the toxicity observed in this patient.
    C) HYPOTENSIVE EPISODE
    1) WITH THERAPEUTIC USE
    a) Hypotension appears to be common following intravenous phenytoin, and is concentration- and dose-related. Binder et al (1996) reported a 4.9% incidence of hypotension in a retrospective study of 164 cases of intravenous phenytoin toxicity in the emergency department setting (Binder et al, 1996). No apnea or dysrhythmias were reported in any cases.
    D) CONDUCTION DISORDER OF THE HEART
    1) WITH THERAPEUTIC USE
    a) ORAL
    1) In a retrospective review of 94 cases of phenytoin toxicity, ECG records were available in 71 patients. No abnormalities or hemodynamic instability were seen in any of the cases. Serum phenytoin concentrations were reported to range from 21.4 to 90 mcg/mL (Curtis et al, 1989).
    2) No incidents of hypotension or dysrhythmia requiring treatment were revealed in 36 patients monitored by continuous single-lead ECG for a mean duration of 26.5 +/- 21.6 hours. Peak phenytoin concentration in these patients ranged from 40 to 76 mcg/mL (Wyte & Berk, 1989).
    b) INTRAVENOUS
    1) Atrial and ventricular conduction depression and ventricular fibrillation have been reported following high-dose infusions of phenytoin. These effects are more typically reported in elderly or gravely ill patients (Prod Info Dilantin(R) intravenous injection solution, 2011).
    2) WITH POISONING/EXPOSURE
    a) ORAL
    1) Several cases of massive oral ingestion (10 g phenytoin or larger in an adult) with no development of hypotension or dysrhythmias have been described (Laubscher, 1966; Tichner & Enselberg, 1951; Griffiths et al, 1987; Weichbrodt & Elliott, 1987; Theil et al, 1961; Nauth-Misir, 1948; Mellick et al, 1989).
    2) In a series of 38 phenytoin oral overdoses (36 chronic, 2 acute) no tachycardia, bradycardia, or cardiac dysrhythmias were noted (Wagner & Leikin, 1986).
    3) In a retrospective review of 94 cases of phenytoin toxicity, ECG records were available in 71 patients. No abnormalities or hemodynamic instability were seen in any of the cases. Serum phenytoin concentrations were reported to range from 21.4 to 90 mcg/mL (Curtis et al, 1989).
    4) No incidents of hypotension or dysrhythmia requiring treatment were revealed in 36 patients monitored by continuous single-lead ECG for a mean duration of 26.5 +/- 21.6 hours. Peak phenytoin concentration in these patients ranged from 40 to 76 mcg/mL (Wyte & Berk, 1989).
    5) A retrospective study of 57 patients with acute phenytoin concentrations greater than 40 mcg/mL after oral overdose revealed no clinically significant cardiac abnormalities in 91% of cases. No deaths or circulatory complications were reported (Wyte & Berk, 1991).
    b) INTRAVENOUS
    1) Atrial and ventricular conduction depression and ventricular fibrillation have been reported following high-dose infusions of phenytoin. These effects are more typically reported in elderly or gravely ill patients (Prod Info Dilantin(R) intravenous injection solution, 2011).
    E) CARDIAC ARREST
    1) WITH THERAPEUTIC USE
    a) Cardiotoxicity with hypotension, dysrhythmias, and cardiac arrest is a common complication of high dose phenytoin administered intravenously at a rate greater than 50 mg/minute. The vehicle for phenytoin, propylene glycol, is believed to be responsible for this effect (Prod Info Dilantin(R) intravenous injection solution, 2011).
    b) CASE REPORT: Intravenous phenytoin administration was related to 8 deaths in elderly patients. Digoxin had been administered in 7 patients and 4 of these had evidence of digoxin toxicity.
    1) Three patients were given rapid injections (1.7 to 2.5 mL/minute), while the remainder received 1 mL/minute or less.
    2) Prior to death, sinus dysrhythmias, ventricular fibrillation, and asystole were noted (Gellerman & Martinez, 1967; Goldschlager & Karliner, 1967; Russell & Bousvaros, 1968; Unger & Sklaroff, 1967) (Zoneraich et al, 1976) (York & Coleridge, 1988).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: Sudden asystole during a craniotomy procedure for aneurysm clipping after a mild subarachnoid hemorrhage is reported in a 49-year-old woman following a rapid unintentional infusion of 1500 mg of phenytoin. The patient was successfully resuscitated and the procedure completed, with full recovery noted by the ninth postoperative day (Berry et al, 1999).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) APNEA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A case of acute respiratory failure associated with eosinophilia was reported in an elderly patient who had been taking phenytoin 300 mg daily for 3 months. This and other signs were attributed to adverse drug reaction (Mahatma et al, 1989).
    2) WITH POISONING/EXPOSURE
    a) Apnea was reported in a fatal case (Laubscher, 1966).
    B) ACUTE RESPIRATORY INSUFFICIENCY
    1) WITH POISONING/EXPOSURE
    a) Irregular shallow respirations and mild ventilatory deficiency have been described (Mellick et al, 1989).
    C) PNEUMONIA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Reversible interstitial pneumonitis, probably due to hypersensitivity, was seen in a 36-year-old man on therapeutic amounts of phenytoin (Michael & Rudin, 1981).
    D) FIBROSIS OF LUNG
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A renopulmonary syndrome, requiring mechanical ventilation, was reported following 3 days of phenytoin therapy, including a 1000 mg loading dose. Lung biopsy revealed an interstitial fibrosis with lymphocytic and plasmacytic infiltration. No eosinophilic infiltration, vasculitis, or granuloma formation was present. Following discontinuation of phenytoin therapy and administration of corticosteroids, the patient recovered (Polman et al, 1998).
    E) RADIOLOGIC INCREASED DENSITY OF LUNG
    1) WITH THERAPEUTIC USE
    a) Four days after beginning phenytoin therapy, a 32-year-old man with seizures secondary to head trauma, developed symptoms which mimicked a renopulmonary syndrome. The patient was febrile, dyspneic and developed renal failure. On chest x-ray, he had bilateral pulmonary infiltrates. After 5 days, phenytoin was stopped and prednisolone started with all symptoms resolving completely (Polman et al, 1998a).
    b) Phenytoin-induced hypersensitivity reaction with bilateral pulmonary infiltrates and associated hypereosinophilia occurred in a 11-year-old boy receiving phenytoin 250 milligrams/day (Fruchter & Laptook, 1981).
    c) An acute hypersensitivity reaction occurred in a 47-year-old man receiving phenytoin 100 milligrams every 6 hours. Clinical features of the reaction were shaking chills, cough, dyspnea, rash, and diffuse reticuloendothelial pulmonary infiltrates (Bayer et al, 1976).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) TOXIC ENCEPHALOPATHY
    1) WITH THERAPEUTIC USE
    a) ACUTE ENCEPHALOPATHY: The acute CNS effects include tremor of the hands, ataxia, nystagmus, and drowsiness.
    b) CHRONIC ENCEPHALOPATHY: Chronic high blood concentrations may cause a "phenytoin encephalopathy" with increase in seizure frequency and the development of more tonic or opisthotonic components. Transient focal neurologic signs such as hemiparesis may be seen, especially in brain-damaged patients (Sandy, 1983).
    c) CASE REPORT: Focal demyelination and loss of large myelinated fibers has been reported in one patient receiving 30 years of phenytoin treatment. The effects were seen to be partially reversible after 16 months phenytoin free (Ramirez et al, 1986).
    d) Pleocytosis in the CSF is rare but has been reported as part of a delayed sensitivity reaction during therapeutic use (Duma et al, 1966).
    e) A series of 11 patients on chronic therapy who developed elevated phenytoin concentrations underwent MRI. Five patients were found to have moderate to severe cerebellar atrophy; 2 patients never developed symptoms of phenytoin intoxication (Luef et al, 1994).
    2) WITH POISONING/EXPOSURE
    a) ACUTE ENCEPHALOPATHY: The acute CNS effects include tremor of the hands, ataxia, nystagmus, and drowsiness (Turkdogan et al, 2002).
    b) CASE REPORT: Phenytoin toxicity resulting in encephalopathy and bilateral asterixis was reported in an 84-year-old man with a phenytoin serum concentration of 37 mg/L. On clinical examination, bilateral coarse intentional tremors, bilateral asterixis of the upper limbs, truncal ataxia, and a broad-based gait were reported. Signs of encephalopathy improved as his serum phenytoin concentration decreased (Chi et al, 2000).
    c) Patients with phenytoin toxicity may present with neurological symptoms suggestive of meningo-encephalitis.
    1) CASE REPORTS: A 2-year-old boy with a 15-day history of a head injury (linear fracture of the occipital bone), presented with fever, vomiting, lethargy, loss of speech, difficulty in walking and sitting, and multiple episodes of generalized tonic clonic seizures. Physical examination showed altered sensorium, terminal neck stiffness, nystagmus, decreased motor power, hyperreflexia in the lower limbs, and bilateral extensor plantar response. Although a diagnosis of meningo-encephalitis was made, all laboratory and diagnostic tests were normal. At this time, it was revealed that he had been receiving multiple loading doses of phenytoin at different healthcare facilities in the past 15 days without monitoring serum phenytoin concentrations. Laboratory results revealed a serum phenytoin concentration of 62.9 mcg/mL (normal: 10 to 20 mcg/mL). Following the discontinuation of phenytoin, his condition gradually improved with a repeat serum phenytoin concentration of 22.6 mcg/mL (Gupta et al, 2011).
    2) CASE REPORT: A 3-year-old boy (weight, 10 kg) with a 13-day history of a head injury (fracture left frontal bone with associated extra dural hemorrhage), presented with fever, vomiting, 2 episodes of generalized tonic clonic seizures, and inability to walk and stand. Physical examination showed altered sensorium, nystagmus, urinary retention, decreased motor power in lower limbs and ataxia. Although a diagnosis of encephalitis was made, all laboratory and diagnostic tests were normal. At this time, it was revealed that he had been receiving phenytoin syrup 375 mg daily for the last 15 days. Laboratory results revealed a serum phenytoin concentration of 62.5 mcg/mL (normal: 10 to 20 mcg/mL). Following the discontinuation of phenytoin, his condition gradually improved and he was asymptomatic on day 9 (Gupta et al, 2011).
    B) CEREBELLAR DISORDER
    1) WITH THERAPEUTIC USE
    a) Chronic phenytoin use has been implicated in chronic cerebellar degeneration causing ataxia unresponsive to discontinuation of the drug (Botez et al, 1985; Lindvall & Nilsson, 1984; Luef et al, 1996). Focal neurologic signs such as hemiparesis may also be present.
    b) AKINETIC MUTISM: Phenytoin toxicity was associated with akinetic mutism in a 12-year-old epileptic girl. With a serum concentration greater than 40 mcg/mL, the girl displayed cyclical sleep/awake states, lacked mental acuity, was unable to speak, had orofacial dyskinesia and incontinence, and displayed no movements with either spontaneous or noxious stimuli. Mild cerebellar atrophy was seen on MRI. Phenytoin was discontinued and the patient started on carbamazepine. Within 2 months motor and mental activity returned to normal (Tutuncuoglu et al, 1997).
    2) WITH POISONING/EXPOSURE
    a) Cerebellar atrophy has been reported as a delayed and unusual effect following severe, acute overdoses. Acute phenytoin intoxication in a 25-year-old woman has been implicated in cerebellar atrophy (Alioglu et al, 2000). Clinical signs of encephalopathy and cerebellar dysfunction were noted on hospital admission (ataxia, nystagmus, diplopia, dysarthria, dysmetria, tremor). No signs of cerebellar atrophy were seen on MRI, but 8 months later MRI did show cerebellar atrophy. Brandolese et al (2001) also report a case of phenytoin toxicity resulting in dysarthria, nystagmus, dysmetria, hemifacial dyskinesia, and mental status changes (Brandolese et al, 2001).
    b) CASE REPORT: A 21-year-old man had evidence of cerebellar atrophy by CT 4 weeks following an ingestion of 7 g phenytoin in a suicide attempt. Treatment included gastric lavage and plasmaphereses.
    1) The phenytoin concentrations in plasma were more than 50 mcg/mL during the first two weeks, 18 mcg/mL after 20 days, and not detectable at six weeks after ingestion. At an 18-month follow-up, moderate dysarthria and dysmetria were detected (Masur et al, 1989).
    2) There was no evidence of cerebellar atrophy by CT 8 months prior to intoxication. Four and one-half years after the suicide attempt, the CT showed the same degree of cerebellar atrophy and a moderate dysmetria was still present (Masur et al, 1990).
    C) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) Seizures are a characteristic of an unusual type of phenytoin toxicity referred to as phenytoin encephalopathy, characterized by increasing seizures, electroencephalogram (EEG) changes, alteration in mental function, and certain motor and sensory disturbances. It is usually associated with toxic doses (but may appear in patients on usual doses) and is generally reversible upon discontinuation of phenytoin (Gupta et al, 2011; VanDerLinde & Campbell, 1977a; Shuttleworth et al, 1974; Vallarta et al, 1974).
    b) CASE REPORT: Three patients, two with a prior history of seizure disorder, were reported to develop seizures following accidental or suicidal overdose of phenytoin.
    c) Plasma concentrations at the time of presentation were 47, 73, and 49 mcg/mL, respectively (Stilman & Masdeu, 1985). Absent nystagmus is a significant negative finding. EEG readings may show marked slowing of alpha rhythm and the appearance of diffuse delta wave activity.
    d) CASE SERIES: A retrospective study of 90 phenytoin-intoxicated patients (blood concentrations greater than 20 mcg/mL plus one or more clinical signs) revealed two cases with a highly probable causal relationship of seizures induced by phenytoin (Osorio et al, 1989).
    e) CASE SERIES: Seizures occurred in 19% of one group of phenytoin-toxic patients (Murray et al, 1996).
    D) COMA
    1) WITH POISONING/EXPOSURE
    a) CNS depression may be more pronounced in children following an acute overdose of oral phenytoin (Jacobsen et al, 1986-87). Coma has been reported in adults following overdoses of phenytoin (Lau et al, 2000).
    b) CASE REPORT: Coma (GCS 3) and EEG showing diffuse abnormalities related to coma were reported in a 26-year-old woman following an overdose of approximately 12 g of phenytoin and an unknown amount of valproic acid. The patient recovered following serial doses of activated charcoal and symptomatic care (Buchwald, 2000).
    c) CASE REPORT: Following the ingestion of several Chinese proprietary medicines, found later to be contaminated with phenytoin, a 33-year-old woman was admitted to the emergency department in a comatose condition with a phenytoin serum concentration of 48.5 mg/L. Her alertness gradually improved, but there was ataxia and bilateral ocular nystagmus 2 days later. Clinical signs resolved within 10 days with no permanent neurological defect (Lau et al, 2000).
    d) CASE REPORT: Phenytoin toxicity consistent with upper motor neuron disease was reported in a middle aged female with elevated free phenytoin due to hypoalbuminemia. On clinical examination, bulbar weakness, diplopia, asymmetric motor weakness, hyperreflexia, and fasciculations were reported. The patient developed chest sepsis likely secondary to aspiration. After normalization of the corrected phenytoin concentration, motor weakness and respiratory function improved within 2 weeks (Veenith et al, 2009).
    E) ATAXIA
    1) WITH POISONING/EXPOSURE
    a) Ataxia may be more pronounced in adults following an acute overdose of oral phenytoin (Jacobsen et al, 1986-87). Ataxia resulting in falls may occur as a result of phenytoin toxicity (Jessen, 2004; Pollak & Slayter, 1997). Ataxia has been reported following toxic serum concentrations of phenytoin (Mamiya et al, 2001).
    b) CASE SERIES: In a retrospective review of 94 cases of phenytoin toxicity, ataxia was observed in 59 patients (63%) (Curtis et al, 1989).
    c) CASE SERIES: In another study ataxia was seen in 88% of cases of phenytoin toxicity (Murray et al, 1996).
    d) COCAINE ADULTERATION (CASE REPORT): A 26-year-old man ingested several crack cocaine rocks and was transported to an ED. He developed symptoms of cocaine toxicity (ie, anxiety, pallor, diaphoresis, agitation, tachycardia, and hypertension) 2 hours postingestion. He was treated with IV benzodiazepine boluses, however, he developed confusion, truncal ataxia, diplopia, slurred speech, and multidirectional nystagmus. At this time, phenytoin toxicity from cocaine adulteration was suspected and laboratory results revealed an elevated phenytoin concentration 48 mcg/mL (therapeutic range: 10 to 20 mcg/mL). Following further supportive care, he was discharged 72 hours after admission without further sequelae (Roldan, 2014).
    F) MONOPLEGIA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Monoplegia was reported to be associated with a serum phenytoin concentration of 28.1 mcg/mL in a 39-year-old woman ho had a hypertensive intracerebral hemorrhage two years earlier (Abdulhadi et al, 1987).
    1) Symptoms resolved when the maintenance dose of phenytoin was decreased to produce a serum phenytoin concentration of 13.3 mcg/mL.
    G) CHOREOATHETOSIS
    1) WITH THERAPEUTIC USE
    a) Phenytoin therapy has been associated with choreoathetosis, characterized by abnormal or jerky movements, lip smacking, continuous tongue movements, slurred speech, ataxia, dysarthria, and dystonic posturing. Choreoathetosis has occurred after days or years of therapy, and with phenytoin serum levels in the therapeutic or toxic range (Chalhub & DeVivo, 1976; Zinsmeister & Marks, 1976; Luhdorf & Lund, 1977; Rasmussen & Kristensen, 1977; Rosenblum et al, 1974; Kooiker & Sumi, 1974; McLellan & Swash, 1974; Krishnamoorthy et al, 1983). Attacks resolve after phenytoin withdrawal.
    b) CASE REPORT: An elderly patient with hypoalbuminemia experienced choreoathetosis even at nontoxic blood phenytoin concentrations (Tomson, 1988).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: Three patients with chronic phenytoin intoxication developed choreoathetoid movements (Shuttleworth et al, 1974a).
    b) CASE REPORT: Choreoathetosis and opisthotonic posturing were reported in a 15-year-old boy after acute ingestion of 19.6 g of phenytoin(Mellick et al, 1989).
    c) CASE REPORT: Dyskinesias as the only manifestation of phenytoin overdose were reported in a 20-year-old woman (Roullet et al, 1987).
    d) CASE REPORT: Choreoathetosis, dystonia, ballismus, and asterixis have also been reported in patients with a phenytoin concentration above a therapeutic blood concentration (Lazaro, 1982).
    e) CASE REPORT: A 4-year-old child presented with a 12-hour history of altered sensorium, involuntary head nodding movements, nystagmus, and a Glasgow coma scale of 10/15. Since her father was taking phenytoin, phenytoin intoxication was suspected. Laboratory results revealed a serum phenytoin concentration of 88 mcg/mL (therapeutic concentration: 10 to 20 mcg/mL). Activated charcoal therapy was not considered because it was about 42 hours postingestion. A week later, a repeat phenytoin serum concentration was still elevated (94 mcg/mL) and her sensorium was essentially unchanged. She gradually recovered following 4 sessions of charcoal hemoperfusion. She developed mild thrombocytopenia (platelet count of 90 to 100,000/mm) after the first session of charcoal hemoperfusion, which resolved on day 3. About 24 hours after each hemoperfusion, serum phenytoin concentrations were 56 mcg/mL, 26 mcg/mL, 23 mcg/mL, and 12 mcg/mL, respectively (Kumar et al, 2012).
    H) DISTURBANCE IN SPEECH
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: An 18-year-old man presented to his physicians office with complete mutism after 13 weeks of therapy with phenytoin 200 mg twice daily. Other presenting signs/symptoms of toxicity included motor tics, lateral gaze nystagmus, lower extremity coarse tremors and upper extremity athetoid-like movements. His phenytoin serum concentration was measured at 64.2 mcg/mL(Berigan & Watt, 1994).
    b) Phenytoin toxicity was associated with akinetic mutism in a 12-year-old epileptic girl. With a serum level greater than 40 mcg/mL, the girl displayed cyclical sleep/awake states, lacked mental acuity, was unable to speak, had orofacial dyskinesia and incontinence, and displayed no movements with either spontaneous or noxious stimuli. Mild cerebellar atrophy was seen on magnetic resonance imaging (MRI). Phenytoin was discontinued and the patient started on carbamazepine. Within 2 months motor and mental activity returned to normal (Tutuncuoglu et al, 1997a).
    2) WITH POISONING/EXPOSURE
    a) Dysarthria may occur with toxic concentrations of phenytoin (Jessen, 2004; Grillone & Myssiorek, 1992).
    b) CASE REPORT: A 19-year-old man developed dysarthria, nystagmus, ataxia and bilateral ankle clonus following the recreational use of phenytoin syrup, marijuana and alcohol. Upon admission 2 days after exposure, a serum phenytoin concentration was 45 mcg/mL; a toxicology screen was negative. Symptoms gradually improved as the phenytoin concentration decreased (Jessen, 2004).
    I) CEREBELLAR INFARCTION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: One case of localized cerebellar necrosis has been associated with phenytoin overdose in a 32-month-old child. On admission, the phenytoin concentration was 72 mcg/mL and CT scan was normal; a repeat CT 6 months later revealed a zone of cerebellar necrosis without clinical signs (Cochat et al, 1987).
    b) CASE REPORT: Severe cerebellar atrophy was described in a 38-year-old man following inadvertent administration of phenytoin 600 mg/day (instead of 300 mg/day) for 2 to 3 weeks. On hospital admission, a serum phenytoin concentration of 83.5 mcg/mL was reported. He had severe ataxia, urine incontinence and encephalopathy. Mild generalized atrophic changes in the cerebrum and cerebellum were seen on magnetic resonance brain scan. Recovery was slow, and the patient was discharged 30 days after admission (Kuruvilla & Bharucha, 1997).
    J) HYPERREFLEXIA
    1) WITH POISONING/EXPOSURE
    a) Hyperreflexia is a frequent finding in overdose. The creatine kinase may be elevated due to agitation and muscle trauma (Mellick et al, 1989).
    K) ALTERED MENTAL STATUS
    1) WITH POISONING/EXPOSURE
    a) Phenytoin toxicity may result in mental status changes, including confusion, combative behavior, memory impairment, agitation, hallucinations or delirium (Klaasen, 1998) (Kuruvilla & Bharucha, 1997; Pitner et al, 1998).
    b) COCAINE ADULTERATION (CASE REPORT): A 26-year-old man ingested several crack cocaine rocks and was transported to an ED. He developed symptoms of cocaine toxicity (ie, anxiety, pallor, diaphoresis, agitation, tachycardia, and hypertension) 2 hours postingestion. He was treated with IV benzodiazepine boluses, however, he developed confusion, truncal ataxia, diplopia, slurred speech, and multidirectional nystagmus. At this time, phenytoin toxicity from cocaine adulteration was suspected and laboratory results revealed an elevated phenytoin concentration 48 mcg/mL (therapeutic range: 10 to 20 mcg/mL). Following further supportive care, he was discharged 72 hours after admission without further sequelae (Roldan, 2014).
    c) CASE REPORT: A 4-year-old child presented with a 12-hour history of altered sensorium, involuntary head nodding movements, nystagmus, and a Glasgow coma scale of 10/15. Since her father was taking phenytoin, phenytoin intoxication was suspected. Laboratory results revealed a serum phenytoin concentration of 88 mcg/mL (therapeutic concentration: 10 to 20 mcg/mL). Activated charcoal therapy was not considered because it was about 42 hours postingestion. A week later, a repeat phenytoin serum concentration was still elevated (94 mcg/mL) and her sensorium was essentially unchanged. She gradually recovered following 4 sessions of charcoal hemoperfusion. She developed mild thrombocytopenia (platelet count of 90 to 100,000/mm) after the first session of charcoal hemoperfusion, which resolved on day 3. About 24 hours after each hemoperfusion, serum phenytoin concentrations were 56 mcg/mL, 26 mcg/mL, 23 mcg/mL, and 12 mcg/mL, respectively (Kumar et al, 2012).
    d) CASE REPORTS: A 15-year-old child without a seizure disorder, ingested 1500 mg of phenytoin in a suicide attempt and presented 3 hours postingestion with ataxia, dysarthria, agitation, and generalized tonic-clonic seizure. Following supportive care, including multiple doses of activated charcoal and midazolam to control seizures and agitations, the child's condition gradually improved with extubation on day 4. The child's serum phenytoin concentrations at 14 hours and 21 hours postingestion were 30 mcg/mL and 39.64 mcg/mL, respectively, but gradually decreased to the therapeutic range 3 days postingestion (Gaies et al, 2011).
    e) Patients with phenytoin toxicity may present with neurological symptoms suggestive of meningo-encephalitis.
    1) CASE REPORTS: A 2-year-old boy with a 15-day history of a head injury (linear fracture of the occipital bone), presented with fever, vomiting, lethargy, loss of speech, difficulty in walking and sitting, and multiple episodes of generalized tonic clonic seizures. Physical examination showed altered sensorium, terminal neck stiffness, nystagmus, decreased motor power, hyperreflexia in the lower limbs, and bilateral extensor plantar response. Although a diagnosis of meningo-encephalitis was made, all laboratory and diagnostic tests were normal. At this time, it was revealed that he had been receiving multiple loading doses of phenytoin at different healthcare facilities in the past 15 days without monitoring serum phenytoin concentrations. Laboratory results revealed a serum phenytoin concentration of 62.9 mcg/mL (normal: 10 to 20 mcg/mL). Following the discontinuation of phenytoin, his condition gradually improved with a repeat serum phenytoin concentration of 22.6 mcg/mL (Gupta et al, 2011).
    2) CASE REPORT: A 3-year-old boy (weight, 10 kg) with a 13-day history of a head injury (fracture left frontal bone with associated extra dural hemorrhage), presented with fever, vomiting, 2 episodes of generalized tonic clonic seizures, and inability to walk and stand. Physical examination showed altered sensorium, nystagmus, urinary retention, decreased motor power in lower limbs and ataxia. Although a diagnosis of encephalitis was made, all laboratory and diagnostic tests were normal. At this time, it was revealed that he had been receiving phenytoin syrup 375 mg daily for the last 15 days. Laboratory results revealed a serum phenytoin concentration of 62.5 mcg/mL (normal: 10 to 20 mcg/mL). Following the discontinuation of phenytoin, his condition gradually improved and he was asymptomatic on day 9 (Gupta et al, 2011).
    L) DISORDER OF THE PERIPHERAL NERVOUS SYSTEM
    1) WITH THERAPEUTIC USE
    a) Sensory peripheral polyneuropathy has been observed in patients receiving phenytoin (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    b) The literature on phenytoin-associated peripheral neuropathies has been reviewed. The clinical manifestations of peripheral nerve impairment among epileptic patients on long-term phenytoin therapy are reflexia in the lower extremities and sensory signs or symptoms. A subclinical state of peripheral nerve dysfunction as revealed by nerve conduction velocities on electromyogram (EMG) may also be present. The duration of therapy in patients with peripheral neuropathies is usually longer than 5 years and serum phenytoin levels have generally been higher than in unaffected patients. Although there is no correlation with either serum vitamin B12 or folate levels, deficiency at the tissue level has not been ruled out. Reversibility of phenytoin-induced peripheral nerve disorders is still not clear. It is important to screen for peripheral nerve dysfunction with peripheral nerve conduction velocities and electromyogram (EMG). Signs and symptoms have resolved following discontinuation of therapy or a correction of dose (So & Penry, 1981a).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) NAUSEA AND VOMITING
    1) WITH POISONING/EXPOSURE
    a) Gastritis with nausea and vomiting may occur due to large amounts of undissolved drug in the stomach (Mellick et al, 1989).
    B) DRUG-INDUCED ILEUS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A dosing miscalculation resulting in supratherapeutic dosing (40 mg 3 times/day for 10 days) of phenytoin in a one-month-old infant resulted in abdominal distension and ileus. Phenytoin serum concentration was reported to be 91.8 mcg/mL. Symptoms resolved following discontinuation of phenytoin (Lowry et al, 2005).
    C) PANCREATITIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Pancreatitis, with elevated amylase and lipase (1370 Units/L and 810 Units/L, respectively), was reported in a 26-year-old woman following the ingestion of approximately 12 g of phenytoin and an unknown amount of valproic acid. The patient recovered following symptomatic therapy (Buchwald, 2000).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) TOXIC HEPATITIS
    1) WITH THERAPEUTIC USE
    a) Toxic hepatitis and liver damage have been reported with phenytoin therapy (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    b) The arene oxide metabolites of phenytoin may be involved in the pathogenesis of phenytoin-induced hepatotoxicity. Predisposition of some patients to phenytoin hepatotoxicity may be due to a heritable defect in response to arene oxides (Spielberg et al, 1981).
    c) Twenty cases of hepatic injury associated with phenytoin therapy are presented. The interval between the beginning of therapy with phenytoin and the adverse drug reaction ranged from 1 to 8 weeks. Accompanying symptoms included fever (75%), rash (62.5%), eosinophilia (89%), and leukocytosis (100%). The morphological lesions included ground-glass transformation of liver cells, hepatocellular degeneration and/or necrosis, granulomatous reactions and infiltration of malignant cells of reticuloendothelial origin (Mullick & Ishak, 1980a).
    B) LIVER ENZYMES ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Mild elevation in LDH and SGOT were reported in a case of massive acute oral ingestion (Mellick et al, 1989).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) BLOOD IN URINE
    1) WITH THERAPEUTIC USE
    a) There is no known clinical observation substantiating the reports which indicate phenytoin as a cause of red urine (Derby & Ward, 1983).
    B) KIDNEY DISEASE
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A case of nephrotic syndrome has been seen in a phenytoin-treated patient who also manifested a polyclonal gamma peak on serum electrophoresis. Other causes of renal signs were ruled out, and the patient improved dramatically after withdrawal of phenytoin therapy (Orlandini & Garini, 1989).
    b) CASE REPORT: Renal dysfunction and rhabdomyolysis associated with a toxic reaction to phenytoin were reported in an adult man who was taking 300 mg daily phenytoin and phenobarbital 60 mg daily (Korman & Olson, 1989).
    c) CASE REPORT: A 26-year-old man developed transient heavy proteinuria and glomerular podocytic lesions 3 weeks after beginning phenytoin therapy. He also had a fever and diffuse erythematous rash. Renal function was normal. Nephrotic range proteinuria (7.8 g/day) with 77% albumin was found. Serum albumin was 31 g/L and gamma-globulin was 14 g/L. A Coombs test was positive for IgG. After discontinuation of phenytoin, proteinuria decreased and was undetectable after 18 days (Messiaen et al, 1997).
    d) CASE REPORT: An 83-year old woman developed phenytoin-induced nephrotic syndrome while on a low dosage (100 mg/day); after discontinuation of phenytoin, the clinical picture improved dramatically in a few days and proteinuria remained absent through a follow-up period of 60 days (Orlandini & Garini, 1989a).
    C) RENAL FAILURE SYNDROME
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Renopulmonary syndrome is reported in early-onset phenytoin (4 days of therapy) toxicity in a 32-year-old man. Elevated serum urea of 18.5 mmol/L and serum creatinine of 607 micromoles/L were reported. Urinalysis revealed elevated red cells and leukocyturia with no casts. After stopping phenytoin and starting prednisolone, renal failure resolved by day 18 (Polman et al, 1998).
    D) INTERSTITIAL NEPHRITIS
    1) WITH THERAPEUTIC USE
    a) CASE REPORT/ADULT: A 63-year-old woman developed interstitial nephritis following treatment with phenytoin 600 mg/day for approximately 3 weeks. Although the patient was also receiving phenobarbital and cefazolin (known inducers of interstitial nephritis), it is believed that phenytoin was the most likely cause. Phenytoin-induced interstitial nephritis is believed to involve an immunologic mechanism which is consistent with the results of autopsy (chronic inflammatory infiltration of the liver, focal interstitial inflammatory infiltrate of the kidney and acute epidermal necrolysis) (Hoffman, 1981a).
    b) CASE REPORT/CHILD: A 6-year-old epileptic treated with phenytoin 50 mg three times daily for one month developed a generalized scaly rash associated with symptoms of lethargy, fever, vomiting, and a seizure. Upon hospitalization the patient was febrile (40 degrees Celsius), noted to have hepatomegaly and a palpable spleen. Phenytoin was discontinued but jaundice became evident on the third hospital day associated with elevated liver function tests with the development of generalized lymphadenopathy on the seventh day. Creatinine clearance studies on the 18th hospital day demonstrated a value of 58 liters/24 hours/square meter with a renal biopsy that demonstrated interstitial nephritis with minimal granular changes. The patient was initiated on prednisone 40 mg/day which resulted in a rapid decrease in the patient's liver size, lymphadenopathy and the resolution of laboratory studies towards normal. Prednisone dosage reduction to 7.5 mg every other day was attempted but resulted in proteinuria requiring return to prednisone 10 mg every other day. Prednisone therapy was successfully discontinued 6 months later (Sheth et al, 1977).
    E) PRIAPISM
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 34-year-old man on a total daily phenytoin dose of 540 mg experienced priapism, ataxia, confusion, and hyperactive reflexes. A blood phenytoin concentration was 38 mcg/mL and CT scan was normal. After treatment and reduction of phenytoin dose, priapism did not recur (Simsek & Ozyurt, 1988).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) MEGALOBLASTIC ANEMIA
    1) WITH THERAPEUTIC USE
    a) Megaloblastic anemia that is generally responsive to folic acid therapy has been reported in patients receiving phenytoin (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    B) GRANULOMA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Reversible bone marrow granulomata were associated with phenytoin administration in an adult man (Eisenstein & Coleman, 1989).
    C) THROMBOCYTOPENIC DISORDER
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Fifteen days following initiation of phenytoin therapy (300 mg/day), severe thrombocytopenia (platelets, 8000/mm(3)), with a serum phenytoin concentration of 20 mg/L, was reported in a 36-year-old woman. Concurrent medications included dexamethasone and analgesics. The patient's platelet count returned to baseline after stopping phenytoin and administering several units of platelets and immune globulin therapy (Holtzer & Reisner-Keller, 1997).
    b) CASE REPORT/TODDLER: A 2-year-old girl developed reversible thrombocytopenia 11 days after beginning phenytoin therapy. She had a Dandy-Walker malformation and ventriculoperitoneal shunt. She was started on phenytoin after a generalized tonic-clonic seizure following a fall with hematoma evacuation. At 11 days, she was lethargic and her platelet count was 14,000/microliter. Platelet counts returned to normal 5 days after phenytoin discontinuation (Alehan et al, 1999).
    D) APLASTIC ANEMIA
    1) WITH THERAPEUTIC USE
    a) Aplastic anemia has occurred secondary to phenytoin administration, with the usual onset being 4 to 13 months after initiation of therapy (average, 9 months) (Robins, 1962a; Tomita et al, 1985a).
    b) CASE REPORT: A 79-year-old man who had been taking phenytoin 200 mg/day for many years for seizure control presented with symptoms of pneumonia. He was found to have macrocytic unregenerative anemia, neutropenia, thrombocytopenia, and very low serum folate, and was diagnosed with aplastic anemia (AA). Serum phenytoin was 10 mcg/mL (therapeutic level 10 to 20 mcg/mL). Phenytoin was discontinued and he was treated with antibiotics, intravenous folic acid, and transfusions of packed red blood cells. One week later, hematological values were nearly normal, and bone marrow biopsy was normal. After prolonged convalescence, he was discharged without anticonvulsants and with normal hematological status. He experienced no seizures while hospitalized. The authors attributed the AA to the prolonged phenytoin treatment with concurrent folic acid deficiency and warned physicians to use caution when prescribing phenytoin for the elderly, in whom folic acid deficiency is not infrequent (Blain et al, 2002).
    E) LEUKOPENIA
    1) WITH THERAPEUTIC USE
    a) Leukopenia has been reported with phenytoin therapy (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    F) HEMATOLOGY FINDING
    1) WITH THERAPEUTIC USE
    a) A cohort study investigating frequency of serious blood dyscrasias in patients taking anticonvulsants has reported the total overall rate for all patients was 3 to 4 per 100,000 prescriptions. Rates did not differ between phenytoin, phenobarbital, carbamazepine, or valproate throughout all age groups (Blackburn et al, 1998).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) ERYTHEMA MULTIFORME
    1) WITH THERAPEUTIC USE
    a) Severe erythema multiforme, exfoliative dermatitis, and Stevens-Johnson syndrome have been reported as hypersensitivity reactions (Watts, 1962).
    B) LYELL'S TOXIC EPIDERMAL NECROLYSIS, SUBEPIDERMAL TYPE
    1) WITH THERAPEUTIC USE
    a) Toxic epidermal necrolysis has been reported as an adverse effect of phenytoin therapy, and is associated with high fever, eosinophilic necrosis of keratinocytes with subepidermal separation (Sun et al, 1994; Sherertz et al, 1985; Schmidt & Kluge, 1983a). Severe cutaneous drug reactions, including toxic epidermal necrolysis, have been associated with phenytoin therapy and with drug interactions between corticosteroids, H2-blockers, and phenytoin, all taken concurrently (Cohen et al, 1999).
    C) STEVENS-JOHNSON SYNDROME
    1) WITH THERAPEUTIC USE
    a) Drug-induced Stevens-Johnson syndrome, an uncommon reaction characterized by atypical target lesions with widespread blisters on the face, mucous membranes, trunk and extremities, has been reported in patients receiving concomitant cranial radiotherapy and phenytoin therapy (Khafaga et al, 1999).
    b) Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) have been described occasionally during the early stages of phenytoin therapy. In a case-control study of patients taking various antiepileptic agents, 73 cases of SJS or TEN were identified; of these, 14 were due to phenytoin ingestion. SJS and TEN generally occurred during the first 8 weeks of therapy (Rzany et al, 1999).
    c) A 2.5-year-old girl developed Stevens-Johnson syndrome following the administration of phenytoin. Four days after the discontinuation of phenobarbital and the initiation of phenytoin, bullous lesions appeared on the child. By the eighth day, severe sloughing of the skin occurred over 75% of the body and Stevens- Johnson syndrome was diagnosed (Crosby et al, 1986).
    D) EDEMA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Severe facial edema, including periorbital edema so severe that the eyelids cannot open, has been reported (Helmy & Tripplet, 1988).
    E) INJECTION SITE INFLAMMATION
    1) WITH THERAPEUTIC USE
    a) PURPLE GLOVE SYNDROME
    1) Purple glove syndrome (PGS) is described following intravenous infusions of phenytoin. The incidence of PGS in a series of 152 patients receiving IV phenytoin was 5.9% (9 patients). PGS is a progressive development of limb edema, discoloration, and pain after phenytoin administration, with some patients developing skin ulceration. Elderly patients and patients receiving large, multiple IV doses appear to be at greatest risk. The vehicles in phenytoin injections (propylene glycol, ethanol, and sodium hydroxide), which are irritants, may contribute to PGS, especially if extravasation occurs (O'Brien et al, 1998).
    F) LUPUS ERYTHEMATOSUS
    1) WITH THERAPEUTIC USE
    a) Rare cases of lupus erythematosus have been reported in patients receiving phenytoin (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    G) HYPERTRICHOSIS
    1) WITH THERAPEUTIC USE
    a) Hypertrichosis has been reported in patients receiving phenytoin (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    H) COARSE FEATURES
    1) WITH THERAPEUTIC USE
    a) The coarsening of facial features has been reported in patients taking phenytoin (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) REFLEX FINDING
    1) WITH THERAPEUTIC USE
    a) Hyper and hyporeflexia have commonly been reported and it is not uncommon for the examination to change over a short time period (Mellick et al, 1989).
    B) OSTEOMALACIA
    1) WITH THERAPEUTIC USE
    a) CHRONIC EXPOSURE: Osteomalacia has been reported in patients receiving long-term phenytoin therapy (greater than 10 years) (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPERGLYCEMIA
    1) WITH THERAPEUTIC USE
    a) Hyperglycemia (due to the inhibition of insulin release) and elevated serum glucose levels in diabetics have been reported in patients taking phenytoin (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    b) CASE REPORT: An 11-year-old boy treated with 200 to 300 mg/day for 3 years along with other anticonvulsants developed ataxia, headache, vomiting, drowsiness, and glycosuria (plasma glucose of 280 mg/dL). Oral glucose tolerance test revealed results indicative of a diabetic curve and in addition, the patient's insulin levels were noted to be raised. Plasma phenytoin level at that time was determined to be 4.4 mg/dl. Discontinuation of phenytoin resulted in the phenytoin level of 2.2 mg/dl 5 days later with normal neurological signs at 3 days. One month later, the patient had a normal glucose tolerance test and was potentially convulsion free on primidone 250 mg/day (Treasure & Toseland, 1971a).
    2) WITH POISONING/EXPOSURE
    a) Hyperglycemia may rarely be noted and is due to altered carbohydrate tolerance. No significant change in electrolytes, glucose, WBC, or alkaline phosphatase occurred after acute phenytoin overdose (Wagner & Leikin, 1986).
    B) HYPOGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A case of hypoglycemia following an intentional overdose of phenytoin (20 g) and zopiclone (225 mg) was reported in a 27-year-old man. There was no personal history of diabetes or alcohol abuse. Blood glucose initially increased, then declined and became refractory to treatment, which included hypertonic dextrose and a total of 32 g of dextrose, which was necessary to maintain blood glucose over 81 mg/dL between hour 23 and 25 (Manto et al, 1996a).
    1) No evidence of a pancreatic tumor was reported. Laboratory tests included normal plasma ACTH and cortisol and normal urine cortisone. The authors suggested a mechanism of an escape from the inhibitory effects of DPH on insulin secretion, and/or increased sensitivity of the tissues to insulin (Manto et al, 1996a).
    C) HYPOTHYROIDISM
    1) WITH POISONING/EXPOSURE
    a) Reversible hypothyroidism has been reported in multiple cases of chronic phenytoin toxicity (Betteridge & Fink, 2009; Sarich & Wright, 1996; Kushnir et al, 1985). One proposed mechanism dubbed "a vicious circle" involves the induction of hepatic monooxygenase by phenytoin leading to increased clearance of free T4. The resulting decreased serum concentrations of T4 results in decreased activity of hepatic NADPH cytochrome P-450 reductase leading to decreased rates of hydroxylation and inactivation of phenytoin (Sarich & Wright, 1996).

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) LYMPHADENOPATHY
    1) WITH THERAPEUTIC USE
    a) Lymphadenopathy, including benign lymph node hyperplasia, pseudolymphoma, lymphoma and Hodgkin's disease, has been reported in patients receiving phenytoin. Lymph node involvement may occur with or without symptoms indicative of serum sickness, such as fever, rash and hepatic involvement (Prod Info Dilantin-125(R) oral suspension, 2011; Prod Info DILANTIN(R) INFATABS(R) oral chewable tablets, 2011).
    b) Lymphadenopathy that mimics malignant lymphoma has been commonly reported. A few cases exist in which patients on anticonvulsant therapy developed malignant lymphoma, but the association is unproved (Helmy & Tripplet, 1988).
    B) ACUTE ALLERGIC REACTION
    1) WITH THERAPEUTIC USE
    a) Hypersensitivity reactions are manifested by skin rashes, fever, neutropenia, and agranulocytosis and occur following chronic administration of therapeutic doses. Dermatitis and/or hepatitis is unlikely following acute overdose. Hypersensitivity reactions include severe erythema multiforme, exfoliative dermatitis, and Stevens-Johnson syndrome.
    b) Hepatitis may be a hypersensitivity reaction, usually appearing with fever, lymphadenopathy, blood dyscrasia (neutropenia, agranulocytosis) (Crawford & Jones, 1962; Ebeid et al, 1989; Black & Fivenson, 1989).
    c) TOXIC EPIDERMAL NECROLYSIS: A fatal necrolysis reaction was associated with prophylactic phenytoin therapy (Kelly & Hope, 1989).
    d) Renal failure (Micheal & Mitch, 1976) (Agarwal et al, 1977), encephalopathy (Ambrosetto et al, 1977), myositis (Chaiken et al, 1950), cerebrospinal fluid pleocytosis (Duma et al, 1966), rhabdomyolysis (Engel et al, 1986) , and pulmonary infiltrates with hypereosinophilia (Fruchter & Laptook, 1981; Bayer et al, 1976) have also been noted.
    e) VASCULITIS: Hypersensitivity vasculitis generally occurs 7 to 10 days after initiation of phenytoin therapy, but may occur after years of therapy. Most commonly, the skin is affected, with purpura or macular eruptions. Renal involvement, with hematuria, nephrotic syndrome and acute renal failure is also common. All organs may be affected, and autoimmune profile is generally negative (Parry et al, 1996).

Reproductive

    3.20.1) SUMMARY
    A) Ethotoin, phenytoin, and phenytoin sodium have been classified as FDA pregnancy category D. Fetal hydantoin syndrome occurs in 10% to 30% of exposed pregnancies. It may result in multisystem anomalies including CNS dysfunction, craniofacial anomalies, major malformations, and nail/digital hypoplasia. If used during pregnancy or if patient becomes pregnant, weigh the risks and benefits of use during pregnancy, inform the patient of the potential harm to the fetus, and measure phenytoin plasma concentrations periodically. Prevent potentially life-threatening bleeding disorders in the infant by administering vitamin K to the mother before delivery and then to the neonate after birth. Breastfeeding while taking phenytoin is not recommended. However, phenytoin is considered to be compatible with breastfeeding by the American Academy of Pediatrics and the World Health Organization. Monitor infant for side effects.
    3.20.2) TERATOGENICITY
    A) CONGENITAL ANOMALY
    1) In studies, increased frequencies of major malformations (eg, cardiac defects, orofacial clefts), minor abnormalities (eg, dysmorphic facial features, nail and digit hypoplasia), growth abnormalities (eg, microcephaly), and mental deficiency have been reported in children born to women administered phenytoin during pregnancy (Prod Info Dilantin-125(R) oral suspension, 2015). Eye malformations (eg, abnormality of the lacrimal apparatus, congenital glaucoma, epicanthal folds, hypertelorism, optic nerve hypoplasia, ptosis, retinoschisis, and strabismus) have also been observed (Grant & Schuman, 1993).
    2) FETAL HYDANTOIN SYNDROME: Chronic therapy with phenytoin during pregnancy has been associated with fetal hydantoin syndrome (FHS), characterized by intrauterine growth retardation, microcephaly, mental retardation, rigid metopic suture, inner epicanthal folds, eyelid ptosis, a broad depressed nasal bridge, nail and/or distal phalangeal hypoplasia, and inguinal hernia. Other effects are cardiovascular (patent ductus arteriosus), gastrointestinal, or genitourinary (renal anomalies, ambiguous genitalia, oral ectopia, pyloric stenosis) (Kogutt, 1984; Barr et al, 1974; Hanson et al, 1976; Hanson & Smith, 1975). The full syndrome has been found in 10% to 11% of exposed newborns and appears to have a genetic influence (Witter et al, 1981). Approximately 40% of newborns exposed to phenytoin in utero will exhibit some features of the disorder (Seeler et al, 1979). Isolated case reports describe polydactyly of the right foot (Yalcinkaya et al, 1997), placental lymphocytosis (Greco et al, 1973), and neonatal acne and suggest an association with phenytoin use during pregnancy (Stankler & Campbell, 1980).
    3) Neocerebellar hypoplasia and multiple other malformations were seen in an infant born to a mother taking 175 mg phenytoin and 1 g sodium valproate daily throughout the pregnancy (Squier et al, 1990). Midface and digital hypoplasia have been associated with anticonvulsant-exposed (ie, phenytoin, phenobarbital, and carbamazepine) children. In some cases, these children may have cognitive dysfunction. In the presence of these conditions further developmental evaluation is suggested (Holmes et al, 2005).
    4) Cardiac defects, cleft lip and palate, intrauterine growth retardation, and irregular ossification and hypoplasia of the distal phalanges have been noted (Schardein, 1985; Gaily, 1990).
    B) ANIMAL STUDIES
    1) Growth impairment, behavior abnormalities, and increased incidences of fetal malformations were observed when phenytoin was administered to animals at clinically relevant doses (Prod Info Dilantin-125(R) oral suspension, 2015).
    2) Increased resorption and malformation rates were reported in pregnant rabbits given phenytoin doses of 75 mg/kg or higher (approximately 120% of the maximum human loading dose or higher on a mg/m(2) basis) (Prod Info DILANTIN KAPSEALS(R) extended oral capsules, 2007).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) ETHOTOIN (Prod Info PEGANONE(R) oral tablets, 2007).
    2) PHENYTOIN (Prod Info Dilantin-125(R) oral suspension, 2015).
    3) PHENYTOIN SODIUM (Prod Info DILANTIN KAPSEALS(R) extended oral capsules, 2007).
    1) The manufacturers have classified the following as FDA pregnancy category D:
    2) If used during pregnancy or if patient becomes pregnant, weigh the risks and benefits of use during pregnancy, inform the patient of the potential harm to the fetus, and measure phenytoin plasma concentrations periodically. Prevent potentially life-threatening bleeding disorders in the infant by administering vitamin K to the mother before delivery and then to the neonate after birth (Prod Info Dilantin-125(R) oral suspension, 2015).
    B) PREGNANCY REGISTRY
    1) Encourage enrollment in the North American Antiepileptic Drug (NAAED) Pregnancy Registry (1-888-233-2334) for any woman who becomes pregnant while being treated phenytoin (refer to http://www.aedpregnancyregistry.org/ for more information) (Prod Info Dilantin-125(R) oral suspension, 2015).
    C) NEOPLASM
    1) There have been isolated reports of malignancies, including neuroblastoma, in offspring of phenytoin-treated women (Prod Info DILANTIN KAPSEALS(R) extended oral capsules, 2007). Neoplasms in infants with fetal hydantoin syndrome (FHS) also have been reported (Allen et al, 1980; Ehrenbard & Chaganti, 1981; Cohen, 1981), suggesting that phenytoin may be a human transplacental carcinogen. However, other researchers studied the incidence of reported cases of neuroblastoma associated with FHS between 1976 and 1988 and concluded that, while there may be a slightly increased risk of neuroblastoma in children with FHS, there was no association between phenytoin use in pregnancy and postnatal neuroblastoma (Koren et al, 1989). Phenytoin has been linked with transplacental tumorigenesis of the kidney, ureter, and bladder (RTECS, 2001).
    D) DEPRESSION OF CLOTTING FACTORS AND HEMORRHAGE
    1) Phenytoin therapy can cause depression of clotting factors and hemorrhage in the newborn. This condition can be prevented with vitamin K administration to the mother before delivery and to the neonate. Phenytoin serum level monitoring throughout pregnancy is also recommended (Prod Info Dilantin-125(R) oral suspension, 2015; Prod Info DILANTIN KAPSEALS(R) extended oral capsules, 2007). However, in one study, no increased incidence of neonatal blood coagulation disorders was reported in a series of women taking phenytoin throughout pregnancy (Hey, 1999). In addition, no increased incidence of vitamin K deficiency in neonates was reported (Hey, 1999)
    E) COGNITIVE IMPAIRMENT
    1) Phenytoin exposure during pregnancy has been associated with impaired neurobehavioral development resulting in lower cognitive abilities. In a prospective study, children exposed in utero to phenytoin had mean lower global intelligence quotient (IQ) points than their matched controls (Scolnik et al, 1994).
    F) MORE FREQUENT SEIZURES
    1) Seizures may occur more frequently in pregnant women when phenytoin is used during pregnancy (Prod Info Dilantin-125(R) oral suspension, 2015).
    G) ANIMAL STUDIES
    1) Embryofetal death was observed when phenytoin was administered at clinically relevant doses (Prod Info Dilantin-125(R) oral suspension, 2015).
    2) A syndrome comparable to fetal hydantoin syndrome has been produced in mice. Genetic strain differences were seen in susceptibility, and the induction of the disorder occurred in offspring of dams who did not have seizure disorders (Finnell et al, 1989). Some genetic differences have been linked with the H-2a haplotype in the H-2 major histocompatibility family on chromosome 17 in mice (Goldman et al, 1983), and others with a gene in the minor H-3 histocompatibility locus on chromosome 2 (Goldman, 1984).
    3) Male rats exposed prenatally to levels of phenytoin sodium of 50 or 100 mg/kg on day 17 of gestation through day 7 postpartum had permanent reductions in seminal vesicle weight; female reproduction was normal (Shapiro & Babalola, 1987).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) Breastfeeding while taking phenytoin is not recommended (Prod Info Dilantin-125(R) oral suspension, 2015). However, phenytoin is considered to be compatible with breastfeeding by the American Academy of Pediatrics (Anon, 2001) and the World Health Organization (Anon, 1995). Monitor infant for side effects.
    2) Phenytoin is excreted into human breast milk. The phenytoin milk to plasma ratios range from approximately 0.06 to 0.55 (Mirkin, 1971; Kaneko et al, 1979; Rane et al, 1974; Steen et al, 1982). Following a maternal dose of 100 mg 3 times a day, the peak concentration in breast milk was 4 mcg/mL (4 mg/L) 3 hours after oral ingestion (daily therapeutic dose in infants, 5 to 7 mg/kg) (Chaplin et al, 1982; Horning et al, 1975). With no further dosing, a stable level of 1 to 2 mcg/mL remained throughout the day (Horning et al, 1975).
    3) One case has been reported in which a breastfed infant developed drowsiness, ecchymoses, and methemoglobinemia. The infant's mother was reported to have received 400 mg phenytoin and 390 mg of phenobarbital daily during pregnancy and the puerperium. The infant was noted to be cyanotic on the fourth day of life (confirmed to be associated with the methemoglobinemia) and upon rechallenge with the mother's breast milk 5 days later became cyanotic again (Finch & Lorber, 1954; Illingworth, 1953).
    4) In one study, 38 breast-fed infants whose mothers received phenytoin 300 to 600 mg daily were followed. The infants did not develop any drowsiness or lethargy (Sparberg, 1963).
    3.20.5) FERTILITY
    A) LACK OF INFORMATION
    1) At the time of this review, no data were available to assess the potential effects on fertility from exposure to this agent (Prod Info Dilantin-125(R) oral suspension, 2015).

Carcinogenicity

    3.21.1) IARC CATEGORY
    A) IARC Carcinogenicity Ratings for CAS57-41-0 (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) IARC Classification
    a) Listed as: Phenytoin
    b) Carcinogen Rating: 2B
    1) The agent (mixture) is possibly carcinogenic to humans. The exposure circumstance entails exposures that are possibly carcinogenic to humans. This category is used for agents, mixtures and exposure circumstances for which there is limited evidence of carcinogenicity in humans and less than sufficient evidence of carcinogenicity in experimental animals. It may also be used when there is inadequate evidence of carcinogenicity in humans but there is sufficient evidence of carcinogenicity in experimental animals. In some instances, an agent, mixture or exposure circumstance for which there is inadequate evidence of carcinogenicity in humans but limited evidence of carcinogenicity in experimental animals together with supporting evidence from other relevant data may be placed in this group.
    B) IARC Carcinogenicity Ratings for CAS630-93-3 (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) Phenytoin is a suspect human carcinogen. Elevated risks for Hodgkin's disease, lymphosarcomas, and reticulum-cell sarcoma have been seen in patients receiving phenytoin therapy (HSDB , 1995). Patients treated with anticonvulsants have shown an elevated risk for cancers of the brain and CNS, but clustering within 10 years of hospitalization suggests that pre-existing brain tumors may have been the cause of the seizure disorders for which the phenytoin was prescribed (Olsen et al, 1989).
    B) There is no evidence that phenytoin can cause liver cancer in humans from the 60 years of experience in its use; therefore the induction of liver tumors in mice is deemed not to be relevant to the human situation (Dethloff et al, 1996).
    3.21.3) HUMAN STUDIES
    A) LYMPHOMA
    1) Phenytoin is a transplacental carcinogen and has been linked mainly with embryonal tumors of neural crest origin in the first 3 years of life. One case of childhood T-lymphocyte lymphoblastic lymphoma has been reported (Murray et al, 1996). One case of Epstein Barr virus-positive non-Hodgkin's lymphoma has been reported in a person after 20 years of phenytoin therapy; the lymphoma was preceded by benign reactive lymphadenopathy (Garcia-Suarez et al, 1996).
    B) NEUROBLASTOMA
    1) There have been isolated reports of malignancies, including neuroblastoma, in offspring of phenytoin-treated women (Prod Info Dilantin-125(R) oral suspension, 2015; Lipson & Bale, 1985; Koren et al, 1989a). Neoplasms in infants with fetal hydantoin syndrome (FHS) have also been reported (Allen et al, 1980; Ehrenbard & Chaganti, 1981; Cohen, 1981).
    3.21.4) ANIMAL STUDIES
    A) HEPATOCELLULAR TUMORS
    1) Increased incidences of hepatocellular tumors were observed in mice at phenytoin doses of 45 mg/kg/day (peak plasma levels below human therapeutic concentrations) and also in female mice at doses up to approximately 90 mg/kg/day (Prod Info Dilantin-125(R) oral suspension, 2015).
    B) LYMPHOMA
    1) Phenytoin sodium induced thymic lymphomas in female mice when given in the feed at a level of 60 mg/kg body weight/day for 168 days (HSDB , 1995). Thymic and mesenteric lymphomas and leukemias were induced in mice with IP doses of 0.6 mg/ animal/day for 66 days (HSDB , 1995).
    C) LACK OF EFFECT
    1) Phenytoin was not carcinogenic in rats when given at doses up to 100 mg/kg/day (peak plasma levels below human therapeutic concentrations) for 2 years. No increased incidences of tumors were observed when given to rats at doses up to approximately 120 mg/kg/day (Prod Info Dilantin-125(R) oral suspension, 2015).
    2) Phenytoin was not carcinogenic in rats when given in the diet at levels of 0.025 or 0.05% for 2 years (Jang et al, 1987). It was also not carcinogenic in mice at dietary levels of 0.006 or 0.12% for 78 weeks (Maeda et al, 1988).

Genotoxicity

    A) Phenytoin has been reported to be both genotoxic and inactive in a variety of short-term genetic assays (Kindig et al, 1992).
    B) The apparently conflicting results of genetic tests with phenytoin could be explained on the basis of a weakly active intermediate which requires metabolic activation. There is evidence, discussed in the REPRODUCTIVE EFFECTS section, which suggests that phenytoin is metabolized to an epoxide. Such an intermediate would be predicted to be genotoxic, and would be formed to varying extents under different conditions depending on the level of activating and deactivating enzymes.

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Obtain a basic metabolic panel, aspirin concentration, and acetaminophen concentration after deliberate overdose.
    B) Monitor phenytoin concentrations every 4 hours until clearly declining.
    C) Monitor serial neurologic examinations.
    D) Obtain an ECG and institute continuous cardiac monitoring during and after rapid intravenous infusion or intravenous overdose.
    E) Obtain serum albumin concentration as phenytoin toxicity may occur in the setting of normal total serum phenytoin concentrations, but elevated free phenytoin concentrations.
    F) Some clinical effects reported in elevated phenytoin concentrations include:
    1) Greater than 20 mcg/mL: far lateral nystagmus
    2) Greater than 30 mcg/mL: 45 degrees lateral gaze nystagmus and ataxia
    3) Greater than 40 mcg/mL: decreased mentation
    4) Greater than 100 mcg/mL: death
    4.1.2) SERUM/BLOOD
    A) Obtain a basic metabolic panel, aspirin concentration, and acetaminophen concentration after deliberate overdose.
    B) Monitor phenytoin concentrations every 4 hours until clearly declining.
    C) ASSAY INTERFERENCE
    1) One study reported undetectable total serum phenytoin levels in a 52-year-old man with adequate phenytoin therapy. All laboratory results, including serum IgG, IgA, and IgM concentrations were normal and Human anti mouse antibody (HAMA) was undetectable. However, elevated levels of serum free kappa light chains (20 mg/L), urine free kappa light chains (43.1 mg/L), and urine kappa/lambda light chain ratio (17.52) were observed. It was concluded that the undetectable total serum phenytoin levels may have been caused by the assay interference from excess immunoglobulins (Rentmeester et al, 2015).
    D) TOXICITY
    1) Total serum phenytoin (free phenytoin concentration when possible) will allow assessment of severity.
    2) Blood concentrations are available and generally seem to correlate with the acute neurological symptoms.
    a) Nystagmus usually appears at about 20 mcg/mL, ataxia at about 30 mcg/mL, and drowsiness at a concentration of 40 mcg/mL. Death can occur at phenytoin concentrations greater than 100 mcg/mL.
    b) Intoxication is rare with total phenytoin blood concentration under 20 mcg/mL, or unbound phenytoin concentrations below 1.5 mcg/L.
    3) In acute oral overdose, a minimum of 2 serial concentrations are recommended, due to delayed absorption, even if the initial concentrations are negligible.
    E) BLOOD/SERUM CHEMISTRY
    1) Monitor renal and hepatic function tests in patients with significant toxicity.
    4.1.4) OTHER
    A) OTHER
    1) MONITORING
    a) Serial neurologic examinations may be necessary during follow-up in patients with severe phenytoin-induced ataxia. Magnetic resonance volumetry has been used to assess damage to the cerebellum after overdoses (Luef et al, 1996).
    b) Obtain an ECG and institute continuous cardiac monitoring during and after rapid intravenous infusion or intravenous overdose.
    2) HAIR
    a) Tsatsakis et al (2000) reported hair sectional analysis with dissolution, liquid phase extraction procedures, and immunoassay or HPLC. These methods may determine a history of phenytoin drug use, with hair phenytoin concentration dependent on the drug dosage. Differences among drug doses according to hair color appeared significant (Tsatsakis et al, 2000).

Methods

    A) MULTIPLE ANALYTICAL METHODS
    1) These are very general guidelines, and considerable individual variation exists. Some phenytoin preparations contain phenobarbital. Several different methods are available for determining phenytoin concentrations.
    2) Care must be taken with the Svensmark and Kristensen method, since ethosuximide, primidone, mephenytoin, metharbital, bishydroxy coumarin, phenylbutazone, oil of cloves, and oil of sassafrass may alter the laboratory results.
    3) In general, current measurement methods are accurate and show good inter-test correlation (Painter et al, 1983).
    4) It should be noted that oxalate and citrate anticoagulation show statistically significant depression of serum phenytoin measurements by EMIT and GLC methods (Godolphin et al, 1983).
    5) Commonly used assays of phenytoin monitor total drug concentrations.
    a) Quantitation of free drug may be more appropriate in evaluating patient response when pathophysiologic conditions (renal insufficiency and hypoalbuminemia) alter the plasma protein binding of phenytoin (Toler et al, 1990; Phelps et al, 1993).

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 who have worsening symptoms after 4 to 6 hours or with significant ataxia or CNS depression, and those with rising serum concentrations, should be admitted to the hospital. If the CNS depression is severe enough that there is respiratory depression or concern of airway protection, the patient should be admitted to an intensive care setting. Otherwise, most patients can be safely admitted to a hospital ward bed. Criteria for discharge from the hospital should include clinical symptomatic improvement, which should be reflected by declining phenytoin serum concentrations.
    6.3.1.2) HOME CRITERIA/ORAL
    A) For unintentional ingestions where the patient is asymptomatic and the dose is less than 20 mg/kg, the patient can be observed at home.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) In patients with cardiac toxicity secondary to the intravenous formulation of phenytoin, it might be reasonable to consult a cardiologist. Neurologists are often very familiar with both the acute and chronic toxicity of phenytoin. Consult a medical toxicologist or poison center for patients with severe toxicity.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Any intentional overdose or inadvertent overdoses above 20 mg/kg, or if symptomatic, the patient should be sent to a healthcare facility for observation. Patients should be observed for at least 4 to 6 hours, and should not be sent home until symptoms are clearly improving, or they are asymptomatic and serum concentrations are clearly declining.
    B) Cardiac monitoring is not necessary following ORAL overdose (Wyte & Berk, 1989; Evers et al, 1997). This does not imply that all oral overdoses can safely be admitted to the medical floor. Some patients may require ICU admission for reasons other than cardiac monitoring (CNS or respiratory depression, suicide precautions, etc).
    1) In a retrospective 4 year chart review of 44 patients with a diagnosis of oral phenytoin overdose (serum concentrations greater than 20 mcg/mL), ECG readings at time of admission in all 44 patients revealed no clinically significant phenytoin related abnormalities. No cardiovascular dysrhythmias or deaths were reported, even with serum concentrations as high as 75 mcg/mL (Evers et al, 1997).
    6.3.2) DISPOSITION/PARENTERAL EXPOSURE
    6.3.2.4) PATIENT TRANSFER/PARENTERAL
    A) Cardiac monitoring is advisable following parenteral overdose or rapid infusions in adults and infants/pediatrics.
    B) Some patients may require ICU admission.

Monitoring

    A) Obtain a basic metabolic panel, aspirin concentration, and acetaminophen concentration after deliberate overdose.
    B) Monitor phenytoin concentrations every 4 hours until clearly declining.
    C) Monitor serial neurologic examinations.
    D) Obtain an ECG and institute continuous cardiac monitoring during and after rapid intravenous infusion or intravenous overdose.
    E) Obtain serum albumin concentration as phenytoin toxicity may occur in the setting of normal total serum phenytoin concentrations, but elevated free phenytoin concentrations.
    F) Some clinical effects reported in elevated phenytoin concentrations include:
    1) Greater than 20 mcg/mL: far lateral nystagmus
    2) Greater than 30 mcg/mL: 45 degrees lateral gaze nystagmus and ataxia
    3) Greater than 40 mcg/mL: decreased mentation
    4) Greater than 100 mcg/mL: death

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) ACTIVATED CHARCOAL
    1) PREHOSPITAL ACTIVATED CHARCOAL ADMINISTRATION
    a) Consider prehospital administration of activated charcoal as an aqueous slurry in patients with a potentially toxic ingestion who are awake and able to protect their airway. Activated charcoal is most effective when administered within one hour of ingestion. Administration in the prehospital setting has the potential to significantly decrease the time from toxin ingestion to activated charcoal administration, although it has not been shown to affect outcome (Alaspaa et al, 2005; Thakore & Murphy, 2002; Spiller & Rogers, 2002).
    1) In patients who are at risk for the abrupt onset of seizures or mental status depression, activated charcoal should not be administered in the prehospital setting, due to the risk of aspiration in the event of spontaneous emesis.
    2) The addition of flavoring agents (cola drinks, chocolate milk, cherry syrup) to activated charcoal improves the palatability for children and may facilitate successful administration (Guenther Skokan et al, 2001; Dagnone et al, 2002).
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    6.5.2) PREVENTION OF ABSORPTION
    A) ACTIVATED CHARCOAL
    1) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    B) MULTIPLE DOSE ACTIVATED CHARCOAL
    1) Multiple dose activated charcoal has not been shown to affect patient outcome, need for, or duration of hospitalization after phenytoin overdose. Routine use of multiple dose activated charcoal is generally not warranted; it may be considered in the rare patient with a life-threatening ingestion. Administration of a second dose of activated charcoal should be considered in patients with rising serum concentrations or worsening clinical condition despite gastric decontamination.
    2) STUDIES
    a) Administration of multiple doses of activated charcoal to volunteers receiving intravenous phenytoin resulted in a decrease in apparent half-life from 44.5 to 22.3 hours (Mauro et al, 1987).
    b) Rowden et al (1990) reported a significant decrease in the area under the curve and in total body clearance associated with administration of activated charcoal in 15 normal volunteers who received 15 mg/kg of phenytoin by intravenous infusion (Rowden et al, 1990).
    c) In a randomized, controlled study, patients with serum phenytoin concentrations of greater than 30 mg/L received either 50 g of activated charcoal orally every 4 hours (n=7 patients) or no activated charcoal (n=8 controls). Serum phenytoin concentrations were obtained every 6 hours for 1 day, then every 24 hours. A subtoxic phenytoin concentration was reached 11.6 to 196 hours (median time: 41.1 hours) in the control group and 13 to 33 hours (median time: 19.3 hours) in the multiple activated charcoal group (p=0.049). The multiple activated charcoal and control groups had median peak serum concentrations of 35.6 hours (range, 32.5 to 40 hours) and 40 hours (range, 32 to 47.6 hours), respectively (p=0.082). The multiple activated charcoal and control groups had median mini-mental status exam (MMSE) scores of 19.5 points (range, 16 to 29) and 20 points (range, 12 to 30), respectively. Overall, multiple doses of activated charcoal decreased the time to reach subtoxic concentrations of serum phenytoin. No adverse effects were observed in any patients (Skinner et al, 2012).
    d) GENETIC POLYMORPHISMS: Administration of multiple-dose activated charcoal (MDAC) to 2 patients with chronic phenytoin toxicity, secondary to genetic polymorphisms of CYP2C9 and 2C19, resulted in a rapid and large reductions in the measured elimination half-lives (before MDAC: 117 hours and no apparent elimination; after MDAC: 27 and 44 hours, respectively). Another patient was not treated with multiple-dose activated charcoal and had a very prolonged elimination of phenytoin (178 hours) (Chan et al, 2015).
    3) CASE REPORTS
    a) Administration of activated charcoal 30 grams every 6 hours to a 38-year-old woman who had ingested 10 to 15 grams of phenytoin resulted in a peak serum concentration 42.5 hours postingestion (compared to historical controls of at least 4 days) and a return to therapeutic concentrations by 5.8 days (Weichbrodt & Elliott, 1987).
    b) Weidle et al (1991) also found multiple dose activated charcoal to be useful when treating a 67-year-old woman with phenytoin toxicity (Weidle et al, 1991).
    c) Multiple-dose activated charcoal given in 4 doses was used in lowering serum phenytoin in a patient with acute on chronic phenytoin toxicity; serum concentration declined from 186.7 micromoles/liter to within the therapeutic range within 36 hours (Howard et al, 1994).
    d) In several pediatric cases, multiple-dose charcoal greatly decreased absorption of phenytoin (Dolgin et al, 1991; Ros & Black, 1989).
    6.5.3) TREATMENT
    A) SUPPORT
    1) Most cases of intoxications may be managed conservatively with good symptomatic and supportive care. Signs and symptoms of toxicity may persist up to 7 to 10 days after ingestion. Serial neurologic examinations may be necessary in patients with severe ataxia. No antidote is currently available for phenytoin overdoses.
    B) SEIZURE
    1) Seizures are extremely rare after acute overdose, and their occurrence should initiate a search for other causes, unless the patient is epileptic.
    2) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    3) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    4) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    5) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    6) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976b; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    7) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    C) 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).
    D) BRADYCARDIA
    1) ATROPINE/DOSE
    a) ADULT BRADYCARDIA: BOLUS: Give 0.5 milligram IV, repeat every 3 to 5 minutes, if bradycardia persists. Maximum: 3 milligrams (0.04 milligram/kilogram) intravenously is a fully vagolytic dose in most adults. Doses less than 0.5 milligram may cause paradoxical bradycardia in adults (Neumar et al, 2010).
    b) PEDIATRIC DOSE: As premedication for emergency intubation in specific situations (eg, giving succinylchoine to facilitate intubation), give 0.02 milligram/kilogram intravenously or intraosseously (0.04 to 0.06 mg/kg via endotracheal tube followed by several positive pressure breaths) repeat once, if needed (de Caen et al, 2015; Kleinman et al, 2010). MAXIMUM SINGLE DOSE: Children: 0.5 milligram; adolescent: 1 mg.
    1) There is no minimum dose (de Caen et al, 2015).
    2) MAXIMUM TOTAL DOSE: Children: 1 milligram; adolescents: 2 milligrams (Kleinman et al, 2010).
    E) MONITORING OF PATIENT
    1) Obtain a basic metabolic panel, aspirin concentration, and acetaminophen concentration after deliberate overdose.
    2) Monitor phenytoin concentrations every 4 hours until clearly declining.
    3) Monitor serial neurologic examinations.
    4) Obtain an ECG and institute continuous cardiac monitoring during and after rapid intravenous infusion or intravenous overdose.
    a) Cardiac telemetry monitoring is generally not necessary following oral overdoses (Evers et al, 1997).
    5) Obtain serum albumin concentration as phenytoin toxicity may occur in the setting of normal total serum phenytoin concentrations, but elevated free phenytoin concentrations.
    6) Some clinical effects reported in elevated phenytoin concentrations include:
    a) greater than 20 mcg/mL: far lateral nystagmus
    b) greater than 30 mcg/mL: 45 degrees lateral gaze nystagmus and ataxia
    c) greater than 40 mcg/mL: decreased mentation
    d) greater than 100 mcg/mL: death
    F) EXPERIMENTAL THERAPY
    1) ALBUMIN: Intravenous administration of albumin 25 g every 6 hours in an attempt to increase the bound fraction of phenytoin has been tried in a patient who overdosed on 6.2 g of phenytoin.
    a) Symptoms were markedly decreased at a phenytoin concentration of 61.8 mcg/mL and serum albumin 6.6 mg/dL compared to earlier marked ataxia and lethargy with a phenytoin concentration of 54.6 mcg/mL and normal serum albumin.
    b) Free serum phenytoin concentrations were not measured in this case, thus a beneficial effect cannot be directly attributed to the albumin (Black et al, 1987).

Enhanced Elimination

    A) SYSTEMATIC REVIEW AND RECOMMENDATIONS
    1) In a systematic review of literature that included 51 studies, including 30 case reports/case series (31 patients), 17 pharmacokinetic studies (54 patients), 1 animal experiment, and 3 in vitro studies, the Extracorporeal Treatments in Poisoning (EXTRIP) workgroup (included international experts) concluded that phenytoin is moderately dialyzable, despite its high protein binding, and recommended the use of ECTR in selected patients with severe phenytoin toxicity. Despite a low quality of evidence for all recommendations, the following guideline was provided (Anseeuw et al, 2016):
    a) ECTR is suggested (Anseeuw et al, 2016):
    1) If prolonged coma is present or expected.
    b) ECTR is reasonable (Anseeuw et al, 2016):
    1) If prolonged incapacitating ataxia is present or expected.
    c) ECTR is NOT recommended (Anseeuw et al, 2016):
    1) If solely based on suspected ingested dose of phenytoin or based on serum phenytoin concentration.
    d) Discontinuation of ECTR is recommended (Anseeuw et al, 2016):
    1) In patients with apparent clinical improvement.
    e) The preferred ECTR is intermittent hemodialysis, with an acceptable alternative being intermittent hemoperfusion, if hemodialysis is not available (Anseeuw et al, 2016).
    B) HEMODIALYSIS
    1) The addition of 2.5% albumin to dialysate significantly enhance dialytic clearance of valproic acid and carbamazepine, but did not increase clearance of phenytoin in one study (Churchwell et al, 2009).
    C) PERITONEAL DIALYSIS
    1) Peritoneal dialysis or exchange transfusion appears to remove less than 1.5% and 11%, respectively, of total body burden and are of no value following acute phenytoin intoxication (Lindahl & Westerling, 1982).
    2) Peritoneal dialysis with a 5% albumin dialysate removed approximately 15% of the total body burden over 9 hours and was considered ineffective in a 3-year-old boy with chronic phenytoin overdose (Czajka et al, 1980).
    3) Narcy et al (1990) reported success in decreasing phenytoin concentrations in a newborn poisoned by an intravenous dose. Peritoneal dialysis removed 1.8 mg/hr of phenytoin and decreased the incidence of previously seen cardiac signs of toxicity. The authors postulated that peritoneal dialysis was successful in this infant due to a high unbound protein fraction and a high diffusion speed through the peritoneum (Narcy et al, 1990).
    D) DIURESIS
    1) Diuresis is ineffective since less than 4% of the drug is excreted unchanged in the urine.
    E) COMBINATION HIGH FLUX DIALYSIS AND HEMOPERFUSION
    1) CASE REPORT: A 35-year-old woman with a severe iatrogenic intravenous phenytoin overdose with a peak plasma concentration of 117 mg/L was started on a combination of high-flux hemodialysis and activated charcoal hemoperfusion. The patient received a cumulative phenytoin dose of approximately 3150 mg (the maximum recommended dose 1500 mg in first 24 hours followed by 500 mg the following day) intravenously over 48 hours. After 4 hours of hemodiaperfusion with activated charcoal, the patient showed signs of clinical improvement with a phenytoin concentration of 74 mg/L. Two more sessions were performed for ongoing clinical symptoms and an elevated phenytoin concentration. Calculated half-life was reduced to between 7 to 13 hours during these procedures, as compared to between 40 to 100 hours with no intervention. No adverse events occurred. The patient was clinically stable and made an uneventful recovery (Eyer et al, 2008).
    2) CASE REPORT: A 49-year-old woman with a recent history of hepatic failure, hyperammonemia, metabolic encephalopathy, coma and facial epilepsy was started on phenytoin. Because of ongoing severe hypoalbuminemia and persistent coma, a free phenytoin concentration was 4.7 mcg/mL (normal values 1 to 2 mcg/mL). The patient was started on a combination of high-flux dialyzer and hemoperfusion using a charcoal filter containing 300 g active charcoal. It was found that the combination of hemodialysis/hemoperfusion was able to significantly reduce the amount of free phenytoin. To evaluate the efficacy of each procedure separately, phenytoin concentrations were obtained before the dialyzer and between the dialyzer and charcoal filter. High-flux dialysis was found to lower free phenytoin fraction concentrations faster, with an average extraction of 38% (+/- 5%), as compared to 16% (+/- 6%) by charcoal. No rebound effects were observed. By day 3, the patient regained consciousness, but died 3 weeks later due to a bowel infarction with refractory lactic acidosis and shock (DeSchoenmakere et al, 2005). The authors concluded that the combination of therapies enhanced the overall clearance of phenytoin and improved clinical signs of toxicity. Further study with a larger patient group is suggested.
    3) CASE REPORT: A 32-year-old man presented to the ED with ataxia and nystagmus one hour after ingesting approximately 17.5 grams of phenytoin. A phenytoin concentration, obtained 6 hours postadmission, was 96 mg/L. Over the next several days, following decontamination with activated charcoal, the patient was intubated due to obtundation and an increased risk of aspiration following emesis. His phenytoin concentration, on hospital day 8, remained elevated at 76 mg/L, therefore hemodialysis (HD) was performed. Although his phenytoin concentration decreased to 51.7 after the 4-hour HD session, his clinical status remained unchanged. On day 10, hemoperfusion (HP) was performed. Again, after the 4-hour HP session, his phenytoin concentration decreased to 38 mg/L, however his clinical status remained unchanged, and his phenytoin concentration rebounded to 52 mg/L 30 hours later. Over the next 2 weeks the patient's phenytoin concentration continued to decrease and he was extubated. Over the next year, the patient continued to have residual cognitive and functional impairment that gradually improved with rehabilitation (Miller et al, 2006).
    F) CHARCOAL HEMOPERFUSION
    1) CASE REPORT: A 4-year-old child presented with a 12-hour history of altered sensorium, involuntary head nodding movements, nystagmus, and a Glasgow coma scale of 10/15. Since her father was taking phenytoin, phenytoin intoxication was suspected. Laboratory results revealed a serum phenytoin concentration of 88 mcg/mL (therapeutic concentration: 10 to 20 mcg/mL). Activated charcoal therapy was not considered because it was about 42 hours postingestion. A week later, a repeat phenytoin serum concentration was still elevated (94 mcg/mL) and her sensorium was essentially unchanged. She gradually recovered following 4 sessions of charcoal hemoperfusion. She developed mild thrombocytopenia (platelet count of 90 to 100,000/mm) after the first session of charcoal hemoperfusion, which resolved on day 3. About 24 hours after each hemoperfusion, serum phenytoin concentrations were 56 mcg/mL, 26 mcg/mL, 23 mcg/mL, and 12 mcg/mL, respectively (Kumar et al, 2012).
    2) CASE REPORT: A 77-year-old man being treated prophylactically with phenytoin 100 mg 3 times daily following a head injury developed ataxia and dizziness 7 days after the start of therapy. The phenytoin plasma concentration was 39.3 mg/L approximately 18 hours after the last dose, and thrombocytopenia and hematuria were also present on admission. Despite phenytoin withdrawal the plasma concentration remained elevated due to slow metabolism, and the patient underwent four 9-hour charcoal hemoperfusion sessions with the concentration decreasing to 3 mg/L. The patient also received colony stimulating factor and antibiotics for a gradual decline in his absolute neutrophil count with a nadir of 0.015 x 10(9)/L along with fever. No residual effects were reported and the patient was discharged on hospital day 26 (Sung et al, 2004).
    3) Kawasaki et al (2000) reported on the successful use of charcoal hemoperfusion in the treatment of a phenytoin overdose. Plasma concentrations of total and free phenytoin fell rapidly, from 40.0 mcg/mL and 3.6 mcg/mL to 16.2 mcg/mL and 1.5 mcg/mL, respectively, following 3 hours of hemoperfusion. The fraction of protein-bound phenytoin remained constant (approximately 90%). Bound phenytoin was reported to dissociate from plasma proteins in the presence of activated charcoal, becoming adsorbed to the charcoal. In this case, charcoal hemoperfusion was effective for the removal of a drug that binds to plasma proteins with a low binding constant (Kawasaki et al, 2000).
    G) PLASMAPHERESIS
    1) Plasmapheresis removes only a small amount of the burden of phenytoin, and in one case the plasma concentration had even increased after plasma exchange (White et al, 1987). Plasmapheresis is of limited value (Silberstein & Shaw, 1986; Larsen et al, 1986).
    H) HEMOFILTRATION
    1) Continuous venous-venous hemofiltration (CVVH) with charcoal filter has been used in an 8-month-old child who sustained bradyasystolic arrest after infusion of 750 mg fosphenytoin over 15 minutes. Serum phenytoin concentrations were 77.2 mcg/mL (total) 1 hour after infusion, 73.8 mcg/mL (total) and 13 mcg/mL (free) 8 hours later, and 82 mcg/mL (total) 16 hours later. Approximately 24 hours after the infusion CVVH was performed for 4 hours. About one hour after completion of CVVH, total and free serum phenytoin concentrations were 33.9 mcg/mL and 3.9 mcg/mL, respectively (Rose et al, 1998).
    I) MULTIPLE DOSE ACTIVATED CHARCOAL
    1) Multiple dose activated charcoal has not been shown to affect patient outcome, need for or duration of hospitalization after phenytoin overdose. Routine use of multiple dose activated charcoal is generally not warranted; it may be considered in the rare patient with a life threatening ingestion. Administration of a second dose of activated charcoal should be considered in patients with rising serum concentrations or worsening clinical condition despite gastric decontamination.
    2) STUDIES
    a) Administration of multiple doses of activated charcoal to volunteers receiving intravenous phenytoin resulted in a decrease in apparent half-life from 44.5 to 22.3 hours (Mauro et al, 1987).
    b) Rowden et al (1990) reported a significant decrease in the area under the curve and in total body clearance associated with administration of activated charcoal in 15 normal volunteers who received 15 mg/kg of phenytoin by intravenous infusion (Rowden et al, 1990).
    3) CASE REPORTS
    a) Administration of activated charcoal 30 g every 6 hours to a 38-year-old woman who had ingested 10 to 15 g of phenytoin resulted in a peak serum concentration 42.5 hours postingestion (compared to historical controls of at least 4 days) and a return to therapeutic concentrations by 5.8 days (Weichbrodt & Elliott, 1987).
    b) Weidle et al (1991) also found multiple dose activated charcoal to be useful when treating a 67-year-old woman with phenytoin toxicity (Weidle et al, 1991).
    c) Multiple-dose activated charcoal given in 4 doses was used in lowering serum phenytoin in a patient with acute on chronic phenytoin toxicity; a serum concentration declined from 186.7 micromoles/liter to within the therapeutic range within 36 hours (Howard et al, 1994).
    d) In several pediatric cases, multiple-dose charcoal greatly decreased absorption of phenytoin (Dolgin et al, 1991; Ros & Black, 1989).

Case Reports

    A) ADULT
    1) A 15-year-old boy ingested 19.6 g of phenytoin (392 mg/kg). He was found comatose with evidence of vomiting, shallow respirations, choreoathetoid movements, and occasional opisthotonus. Hyperreflexia and ankle clonus were present (Mellick et al, 1989).
    a) Gastric lavage was performed and multiple dose activated charcoal was initiated. The peak phenytoin concentration occurred on the second day at 100.8 mcg/mL. During the first 3 days, combativeness, severe agitation, and responsiveness only to pain were noted.
    b) Laboratory findings on the third day included elevated creatine phosphokinase (6267 Units/L) and elevated LFTs (LDH 2247 Units/L, SGOT 171 Units/L, total bilirubin 1.4 mg/dL). By the fifth day he was oriented and ataxic with serum concentrations of 42 mcg/mL. He was discharged to psychiatric care on day 8.
    B) PEDIATRIC
    1) A neonate born prematurely to a drug-abusing mother was accidentally dosed with 55 mg/kg phenytoin instead of the calcium gluconate that was ordered. Phenytoin blood concentration was 93.2 mcg/mL, and the infant became unresponsive and apneic. With supportive care, the patient recovered without incident. The erroneous medication administration was attributed to the similarity of the drug product packages (Lombardi et al, 1989).
    2) A one-month-old girl was unintentionally dosed with phenytoin 40 mg 3 times/day for 10 days, instead of the intended dose of 15 mg 3 times/day. The patient presented with hypothermia, abdominal distension and ileus, as well as poor responsiveness. Phenytoin serum concentration was reported to be 91.8 mcg/mL. Symptoms resolved following discontinuation of phenytoin (Lowry et al, 2005).

Summary

    A) TOXICITY: The minimum acute toxic oral overdose is 20 mg/kg. However, since phenytoin has dose-dependent kinetics, inadvertent intoxication from drug interactions or small dose adjustments can easily occur in patients taking the medication chronically. Fatalities have most often been associated with the intravenous formulation being given too rapidly, and is thought to be secondary to the propylene glycol diluent rather than the phenytoin itself.
    B) THERAPEUTIC DOSE: SEIZURE DISORDER: ADULT: Usual maintenance dose is 100 mg orally 3 to 4 times daily, up to a maximum of 600 mg/day. PEDIATRIC: Children less than 6 years of age: 5 mg/kg/day divided into 2 or 3 doses up to a maximum of 300 mg/day; usual maintenance dose is 4 to 8 mg/kg/day. Children older than 6 years of age: May require the minimum adult dose of 300 mg/day. STATUS EPILEPTICUS: ADULT: Loading dose is between 10 to 15 mg/kg IV with a maximum rate of 50 mg/minute. PEDIATRIC: The recommended loading dose is 15 to 20 mg/kg IV at a rate not exceeding 1 to 3 mg/kg/min or 50 mg/min (whichever is slower).

Therapeutic Dose

    7.2.1) ADULT
    A) SEIZURE DISORDER
    1) ETHOTOIN
    a) Initial dose, 1 g or less, daily, orally in 4 to 6 divided doses; may be increased over a period of several days up to a maintenance dose of 2 to 3 g daily (Prod Info Peganone(R) oral tablets, 2013).
    2) PHENYTOIN
    a) CHEWABLE ORAL TABLETS: 2 tablets (50 mg each) 3 times daily; usual maintenance dose 6 to 8 tablets/day (300 to 400 mg/day); MAX: 12 tablets/day (600 mg/day) (Prod Info Dilantin(R) Infatabs(R) oral chewable tablets, 2014)
    b) EXTENDED-RELEASE CAPSULES: 100 mg orally 3 times a day; usual maintenance dose is 100 mg orally 3 to 4 times a day. Doses up to 200 mg orally 3 times a day may be used if necessary. Patients established on 100 mg 3 times a day may take one 300-mg extended-release capsule once daily (Prod Info Dilantin(R) oral extended release capsules, 2014; Prod Info DILANTIN(R) extended capsule, oral, 2009).
    c) ORAL SUSPENSION: 125 mg (5 mL) 3 times daily; adjust dose every 7 to 10 days as necessary (maximum: 625 mg/day) (Prod Info DILANTIN-125(R) oral suspension, 2003).
    d) INJECTABLE SOLUTION: 100 to 200 mg IM every 4 hours during surgery and continued during the postoperative period (Prod Info phenytoin sodium intravenous, intramuscular injection, 2010)
    e) INJECTABLE SOLUTION, NONEMERGENT: 10 to 15 mg/kg IV loading dose (at a rate not exceeding 50 mg/min), followed maintenance doses 100 mg orally or IV every 6 to 8 hours (Prod Info Dilantin(R) intravenous injection solution, 2011; Prod Info phenytoin sodium intravenous, intramuscular injection, 2010).
    f) INJECTABLE SOLUTION, SUBSTITUTION FOR ORAL PHENYTOIN: Continue same total daily dose, at a rate not exceeding 50 mg/min IV (Prod Info Dilantin(R) intravenous injection solution, 2011).
    7.2.2) PEDIATRIC
    A) SEIZURE DISORDER
    1) ETHOTOIN
    a) Initial oral dose should not exceed 750 mg daily; maintenance dose ranges from 500 to 1 g daily (Prod Info Peganone(R) oral tablets, 2013).
    2) PHENYTOIN
    a) CHEWABLE ORAL TABLETS: 5 mg/kg/day in 2 or 3 divided doses; usual maintenance dose 4 to 8 mg/kg. MAX: 300 mg/day. Children over the age of 6 years may require minimum adult dose of 300 mg/day (Prod Info Dilantin(R) Infatabs(R) oral chewable tablets, 2014).
    b) EXTENDED-RELEASE CAPSULES: Initial dose, 5 mg/kg/day orally divided equally into 2 or 3 doses; maintenance dose, 4 to 8 mg/kg/day. MAX: 300 mg/day. Children over the age of 6 years may require minimum adult dose of 300 mg/day (Prod Info Dilantin(R) oral extended release capsules, 2014).
    c) INJECTABLE SOLUTION, NONEMERGENT: 15 to 20 mg/kg IV loading dose, at a rate not exceeding 1 to 3 mg/kg/min or 50 mg/min (whichever is slower) (Prod Info Dilantin(R) intravenous injection solution, 2011).
    d) INJECTABLE SOLUTION, SUBSTITUTION FOR ORAL PHENYTOIN: continue same total daily dose at a rate not exceeding 1 to 3 mg/kg/min or 50 mg/min (whichever is slower) (Prod Info Dilantin(R) intravenous injection solution, 2011).
    e) ORAL SUSPENSION, ORAL TABLETS (Infatabs(R)): 5 mg/kg/day divided equally into 2 or 3 doses to a subsequent MAXIMUM of 300 mg/day; usual maintenance dose is 4 to 8 mg/kg/day; children over the age of 6 years may require minimum adult dose (300 mg/day) (Prod Info DILANTIN(R) KAPSEALS(R) oral capsule, 2003; Prod Info DILANTIN-125(R) oral suspension, 2003).
    f) MAINTENANCE THERAPY: The average mg/kg/day oral dose required to achieve a phenytoin level of 15 mcg/mL was as follows: (Bauer & Blouin, 1983)
    g) 0.5 to 3 years of age, 9.7 mg/kg/day.
    h) 4 to 6 years of age, 7.5 mg/kg/day.
    i) 7 to 9 years of age, 7 mg/kg/day.
    j) 10 to 16 years of age, 6 mg/kg/day.

Minimum Lethal Exposure

    A) GENERAL
    1) ORAL: Estimates of the minimal lethal dosage are unreliable. Deaths are very rare even with massive acute oral overdosage and have been reported mostly with the relatively serious hypersensitivity reactions seen with chronic use. The lowest published lethal dose in a human child is 100 mg/kg (RTECS , 2001).
    B) CASE REPORTS
    1) PEDIATRIC
    a) One 7-year-old boy who ingested 2 g phenytoin (100 mg/kg) presented comatose, but later developed vigorous motor activity, tetanic spasms, and Kussmaul respirations. He died 42 hours postingestion (Schmeiser, 1952).
    b) A 4.5-year-old girl ingested 2 g phenytoin (160 mg/kg), and hyperactivity was the first sign of intoxication, but this progressed to ataxia, miosis, epigastric pain, and generalized pruritus. Later the ataxia worsened with the patient becoming lethargic and unresponsive and she lapsed into a coma. Eighty hours after the onset of symptoms the patient died (Laubscher, 1966).
    c) A 3-year-old boy who ingested 3.2 g phenytoin (220 mg/kg) became lethargic, then comatose without reflexes. He experienced several minor seizures and died 34 hours postingestion (Petty et al, 1957).
    2) ADULT
    a) CHRONIC THERAPY: A 35-year-old man with a history of traumatic brain injury, epilepsy and pulmonary tuberculosis in a vegetative state developed severe cardiotoxicity (ie, junctional bradycardia, hypotension) after receiving phenytoin 200 mg twice daily for approximately 5 months. After transvenous pacing and intensive supportive care, the patient returned to baseline within 2 weeks (Su et al, 2009). The authors suggested that underlying metabolic derangements and a possible drug interaction (receiving an anti-TB agent) may have contributed to phenytoin toxicity in this patient.
    b) ACUTE: Two adult men, a 37-year-old and an 18-year-old, were found dead in their homes after taking overdoses of phenytoin the previous evening. Phenytoin serum concentrations in each patient several hours postmortem were 45 mcg/mL and 48 mcg/mL, respectively (Coutselinis et al, 1975).
    C) ROUTE OF EXPOSURE
    1) INTRAVENOUS: Death may be seen after rapid intravenous administration of phenytoin from severe hypoperfusion with intractable shock, cardiac standstill, ventricular fibrillation, or respiratory arrest. These effects are nearly always rate related (Prod Info Dilantin(R) intravenous injection solution, 2011).

Maximum Tolerated Exposure

    A) GENERAL
    1) Many cases of mild to moderately acute overdoses with phenytoin are reported. Also a few cases of acute overdose are reported in which patients survived ingestion of large amounts of oral phenytoin.
    B) CASE REPORTS
    1) ADULT
    a) ORAL TOXICITY
    1) There have been reports in the literature of adults ingesting between 4.5 g (Robinson, 1940; Black et al, 1987; Weichbrodt & Elliott, 1987; Blair et al, 1968; Miller et al, 2006; Theil et al, 1961; Grosz, 1956) up to 25 g of phenytoin (Nauth-Misir, 1948).
    2) A 20-year-old epileptic male ingested 15 g of primidone and 12 g of phenytoin (17 mg/kg), and had a phenytoin plasma concentration of 130 mcg/mL approximately 96 hours after ingestion. Peritoneal dialysis was begun and phenytoin plasma concentrations decreased from 130 mcg/mL to 30 mcg/mL over a 12-hour period, with recovery of greater than 450 mg phenytoin in the dialysate. In this case, it appears that peritoneal dialysis contributed to the lowering of phenytoin plasma concentrations (Blair et al, 1968).
    3) An 18-year-old pregnant epileptic ingested phenobarbital 2.4 grams and phenytoin 21.5 grams. At 138 hours postingestion the phenytoin concentration was 50 mcg/mL. Because of a lack of improvement in the patient's condition, she received hemodialysis for 6 hours. Phenytoin plasma concentrations dropped from 50 mcg/mL to 4 mcg/mL following dialysis. Although attempts to measure phenytoin in the dialysate were unsuccessful, it appears that dialysis contributed to the decrease in phenytoin concentrations (Theil et al, 1961).
    4) A 15-year-old boy with absence seizures ingested 19.6 g of phenytoin. The peak phenytoin concentration occurred 2 days postingestion and was 100.8 mcg/mL (Mellick et al, 1989).
    5) A 38-year-old man with a history of a right ganglia hemorrhage, seizures and diabetes mellitus ingested an unknown amount of alcohol and 10 g of phenytoin (133 mg/kg) and was initially alert and complaining of nausea and vomiting with evidence of ataxia. Serum phenytoin concentration peaked on day 15 at 90 mcg/mL. Despite aggressive supportive care including whole bowel irrigation (ileus developed and treatment was stopped) and charcoal hemoperfusion, the patient had persistently elevated phenytoin concentrations and progressing cerebellar toxicity. Phenytoin concentrations eventually dropped with the administration of multidose charcoal, but the patient was discharged to a rehabilitation center 100 days after admission with permanent cerebellar dysfunction (Craig, 2004).
    6) A 32-year-old man presented to the ED with ataxia and nystagmus 1 hour after ingesting approximately 17.5 g of phenytoin. Despite decontamination with activated charcoal, the patient's serum phenytoin concentration peaked at 96 mg/L approximately 7 hours postingestion. Over the next several days, the patient was intubated, due to obtundation and a risk of aspiration following emesis. His phenytoin concentrations also fluctuated following hemodialysis and hemoperfusion sessions, although his clinical status remained unchanged. Gradually, his phenytoin concentration decreased and he was extubated. Over the next year, the patient continued to experience residual cognitive and functional impairment which slowly improved with rehabilitation (Miller et al, 2006).
    b) INTRAVENOUS TOXICITY
    1) INADVERTENT EXPOSURE: A peak phenytoin plasma concentration of 117 mg/L was reported in a 35-year-old woman after receiving a cumulative phenytoin intravenous dose of approximately 3150 mg over 48 hours. She developed symptoms of violent agitation, confusion, drowsiness with nystagmus, dysarthria and cerebellar ataxia. The patient recovered completely following several sessions of hemodialysis and hemoperfusion (Eyer et al, 2008).
    2) Following a change in phenytoin oral dosing (100 mg 3 times per day) to the same dose given intravenously, phenytoin toxicity was reported in a 52-year-old man 5 days later. Neurologic findings, including mandibular tremor, encephalopathy and nystagmus (not related to his hydrocephalus) were reported, as well as a phenytoin serum concentration of 42 mcg/mL (Turkdogan et al, 2002).
    2) PEDIATRIC
    a) A 2.5-year-old boy ingested approximately 2.8 g phenytoin suspension (249 mg/kg). At 70 hours postingestion, the phenytoin plasma concentration was 112 mcg/mL. Peritoneal dialysis was begun and 16 hours later the concentration dropped to 75 mcg/mL. After 36 hours of dialysis the phenytoin serum concentration was 35 mcg/mL. However, analysis of the dialysis showed only "trace" phenytoin. The contribution of the dialysis to the lowering of the plasma concentrations in the patient is questionable based on the dialysate findings and the slow decrease in phenytoin concentrations (Tenckhoff et al, 1968).
    C) OTHER
    1) Severe phenytoin intoxication may result from altered protein binding associated with disease states. Agitation, lethargy, lack of response to deep pain, and bilateral clonus developed in a 25-year-old HIV positive woman while receiving a maintenance dose of phenytoin of 400 mg/day. Her total serum phenytoin concentration was 10.9 mcg/mL with a free phenytoin concentration of 4.9 mcg/mL (normal 1 to 2 mcg/mL) (Toler et al, 1990).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) PHENYTOIN
    a) An upper limit for therapeutic phenytoin serum concentration is generally considered at 20 mcg/mL. In some cases, though, supratherapeutic concentrations are required to control seizures. In a 9-year-old girl, concentrations as high as 40 mcg/mL were necessary to control her seizure disorder (Kozer et al, 2002). No signs of toxicity, other than nystagmus, were evident at this serum concentration.
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) SUMMARY
    a) THERAPEUTIC CONCENTRATIONS: 10 to 20 mcg/mL (Blaser et al, 1989).
    b) TOXIC EFFECTS: Usually seen at concentrations greater than 30 mcg/mL (Boyd, 1986). Commonly, nystagmus develops at concentrations around 20 mcg/mL. At 30 mcg/mL, ataxia develops and lethargy is seen at about 40 mcg/mL (Gilman et al, 1990).
    c) CASE REPORTS
    1) CHRONIC THERAPY: A peak phenytoin plasma concentration of 91 mcg/mL was observed in a 35-year-old man with a history of traumatic brain injury, epilepsy and pulmonary tuberculosis in a vegetative state. Severe cardiotoxicity (ie, junctional bradycardia, hypotension) occurred after receiving phenytoin 200 mg twice daily for approximately 5 months (Su et al, 2009). The authors suggested that underlying metabolic derangements and a possible drug interaction (receiving an anti-TB agent) may have also contributed to phenytoin toxicity in this patient.
    2) A peak phenytoin plasma concentration of 117 mg/L was reported in a 35-year-old woman after receiving a cumulative phenytoin intravenous dose of approximately 3150 mg over 48 hours. She developed significant CNS toxicity and recovered completely following several sessions of hemodialysis and hemoperfusion (Eyer et al, 2008).
    3) A phenytoin concentration of 41 mcg/mL was reported at 9 hours following an ingestion of 100 capsules of phenytoin 100 mg in a 21-year-old woman (Griffiths et al, 1987).
    4) In one series of patients, admission phenytoin serum concentrations following an overdose were not a useful predictor of length of hospital stay (Curtis et al, 1989).
    5) Plasma concentrations as high as 112 and 108 mg/mL have been reported in intoxicated children (Larsen & Larsen, 1989).
    6) In a case of severe phenytoin toxicity following 10 days of therapeutic dosing (100 mg 3 times daily) in a patient homozygous for CYP2C9*3, a serum concentration of greater than 100 mg/L was reported. Clinical manifestations of dysarthria, nystagmus, dysmetria, left hemifacial dyskinesia, and mental status changes were reported (Brandolese et al, 2001).
    7) FREE PHENYTOIN: The proportion of phenytoin in the blood not bound to protein is an important measure of potential toxicity.
    a) Levels of free phenytoin less than 1.5 mcg/mL had no signs of toxicity; 1.5 to 3 mcg/mL was seen with mild intoxication; 3.5 to 5 mcg/mL was associated with some signs of toxicity in all subjects and concentrations greater than 5 mcg/mL were associated with severe intoxication in all subjects (Larsen & Larsen, 1989).

Workplace Standards

    A) ACGIH TLV Values for CAS57-41-0 (American Conference of Governmental Industrial Hygienists, 2010):
    1) Not Listed

    B) ACGIH TLV Values for CAS630-93-3 (American Conference of Governmental Industrial Hygienists, 2010):
    1) Not Listed

    C) NIOSH REL and IDLH Values for CAS57-41-0 (National Institute for Occupational Safety and Health, 2007):
    1) Not Listed

    D) NIOSH REL and IDLH Values for CAS630-93-3 (National Institute for Occupational Safety and Health, 2007):
    1) Not Listed

    E) Carcinogenicity Ratings for CAS57-41-0 :
    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): 2B ; Listed as: Phenytoin
    a) 2B : The agent (mixture) is possibly carcinogenic to humans. The exposure circumstance entails exposures that are possibly carcinogenic to humans. This category is used for agents, mixtures and exposure circumstances for which there is limited evidence of carcinogenicity in humans and less than sufficient evidence of carcinogenicity in experimental animals. It may also be used when there is inadequate evidence of carcinogenicity in humans but there is sufficient evidence of carcinogenicity in experimental animals. In some instances, an agent, mixture or exposure circumstance for which there is inadequate evidence of carcinogenicity in humans but limited evidence of carcinogenicity in experimental animals together with supporting evidence from other relevant data may be placed in this group.
    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

    F) Carcinogenicity Ratings for CAS630-93-3 :
    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

    G) OSHA PEL Values for CAS57-41-0 (U.S. Occupational Safety, and Health Administration (OSHA), 2010):
    1) Not Listed

    H) OSHA PEL Values for CAS630-93-3 (U.S. Occupational Safety, and Health Administration (OSHA), 2010):
    1) Not Listed

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) PHENYTOIN
    1) LD50- (ORAL)MOUSE:
    a) 150 mg/kg (RTECS, 2001)
    2) LD50- (ORAL)RAT:
    a) 1635 mg/kg (RTECS, 2001)

Pharmacologic Mechanism

    A) Phenytoin is an antiepileptic drug that inhibits the spread of seizure activity in the motor cortex. Through the promotion of sodium efflux from neurons, phenytoin possibly tends to stabilize the threshold against hyperexcitability caused by excessive stimulation or changes in environment that are capable of reducing membrane sodium gradient. Reduction of posttetanic potentiation at synapses occurs which prevents cortical seizure foci from detonating adjacent cortical areas. The maximal activity of brain stem centers that are responsible for the tonic phase of tonic-clonic seizures is reduced by phenytoin (Prod Info Dilantin(R) intravenous injection solution, 2011; Prod Info DILANTIN(R) extended capsule, oral, 2009).

Toxicologic Mechanism

    A) HYPOALBUMINEMIA: Since phenytoin is so highly protein bound, hypoalbuminemic patients will have a greater amount of free (active) drug in the blood and experience higher concentrations for the dose administered.
    1) A report of 4 critically ill patients who were started on phenytoin following seizures during their ICU stay developed elevated free phenytoin concentrations and clinical signs of phenytoin toxicity with mildly elevated or normal total phenytoin concentrations. All patients had serum albumin concentrations less than 2 g/dL (Lindow & Wijdicks, 1994).
    2) These patients are at risk of overdosage when given normal therapeutic doses and frequently manifest signs of toxicity (Toler et al, 1990; Driscoll et al, 1988; Tomson, 1988).
    3) Patients at high risk for hypoproteinemia include the elderly, those who are HIV positive, and those with chronic debilitating disease.
    B) Patients with chronic renal failure and neonates have increased free phenytoin; hepatic dysfunction due to liver damage, genetic defects in metabolism, and inhibition of metabolism by other drugs will also predispose patients to toxicity at therapeutic concentrations of phenytoin (Ellenhorn & Barceloux, 1988).
    C) Viral infections can cause transient increases in serum phenytoin and resultant toxicity (Baselt & Cravey, 1989).
    D) GINGIVAL HYPERPLASIA: Zhou et al (1996) hypothesized that the proliferative inflammation following phenytoin use in some patients may be initiated by formation of reactive metabolites, which may be catalyzed by one or more cytochrome P450 isoenzymes located in the gingiva. Gingival cellular injury and an induced reactive inflammatory response, followed by fibroblastic proliferation may then occur (Zhou et al, 1996).
    E) The intravenous form of phenytoin is dissolved in 40% propylene glycol and 10% ethanol at a pH of 12. This formulation can have its own toxicity secondary to cardiac toxicity of propylene glycol (mechanism unknown) and tissue necrosis from infiltration (secondary to the alkaline nature of the formulation) (Prod Info Dilantin(R) intravenous injection solution, 2011).

Physical Characteristics

    A) Phenytoin is a powder with a bitter, soapy taste (HSDB , 1991; Budavari, 1996).
    B) Ethotoin is a white crystalline powder which is practically insoluble in water (S Sweetman , 2001).
    C) Mephenytoin is a white crystalline powder with 1 part soluble in 1400 parts of water (S Sweetman , 2001).

Molecular Weight

    A) Phenytoin: 252.26
    B) Ethotoin: 204.2
    C) Mephenytoin: 218.3

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) AIHA: 2006 Emergency Response Planning Guidelines and Workplace Environmental Exposure Level Guides Handbook, American Industrial Hygiene Association, Fairfax, VA, 2006.
    13) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    14) AMA: Department of Drugs: AMA Drug Evaluation, 5th ed, American Medical Association, Chicago, IL, 1983.
    15) Abdel-Hamid HA, Abdel-Rahman MS, & Abdel-Rahman SA: Teratogenic effect of diphenylhydantoin and/or fluphenazine in mice. J Appl Toxicol 1996; 16:221-225.
    16) Abdulhadi MH, Notman DD, & Cardon GC: Phenytoin toxicity: A cause of reversible monoplegia. Cleve Clin J Med 1987; 54:438-439.
    17) Abernethy DR & Greenblatt DJ: Phenytoin disposition in obesity: Determination of loading dose. Arch Neurol 1985; 42:468-471.
    18) Adams SL, Mathews J, & Grammer LC: Drugs that may exacerbate myasthenia gravis. Ann Emerg Med 1984; 13:532-538.
    19) Addy V, McElnay JC, Eyre DG, et al: Risk factors in phenytoin-induced gingival hyperplasia. J Periodontol 1983; 54:373.
    20) Agarwal BN, Cabebe FG, & Hoffman BI: Diphenylhydantoin-induced acute renal failure. Nephron 1977; 18:249-251.
    21) Ahmad S, Laidlaw J, & Houghton GW: Involuntary movements caused by phenytoin intoxication in epileptic patients. J Neurol Neurosurg Psych 1975; 38:225-331.
    22) Ahmad S: Amiodarone and phenytoin: interaction (letter). JAGS 1995; 43:1449-1450.
    23) Ahmad S: Nifedipine-phenytoin interaction (letter). J Am Coll Cardiol 1984; 3:1582.
    24) Ahmad S: Renal insensitivity to frusemide caused by chronic anticonvulsant therapy. Br Med J 1974; 3:657.
    25) Ahmad S: Ticrynafen-induced phenytoin toxicity: an interaction. J R Soc Med 1981; 74:162-163.
    26) Al-Kawas FH, Lenes BA, & Sacher RA: Cimetidine and agranulocytosis (letter). Ann Intern Med 1979; 90:992-993.
    27) 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.
    28) Albertson TE, Fisher CJ, & Shragg TA: A prolonged severe intoxication after ingestion of phenytoin and phenobarbital. West J Med 1981; 135:418-422.
    29) Alehan PK, Russell EC, & Pellock JM: Phenytoin-associated thrombocytopenia in a toddler. J Child Neurol 1999; 14:130-131.
    30) Algozzine GJ, Stewart RB, & Springer PK: Decreased clearance of phenytoin with cimetidine. Ann Intern Med 1981; 95:244-245.
    31) Alhaj E & Alhaj N: Hypothermia and phenytoin toxicity: a case report. Clin Neuropharmacol 2001; 24:239-241.
    32) Alioglu Z, Sari A, & Velioglu SK: Cerebellar atrophy following acute phenytoin intoxication. J Neuroradiol 2000; 27:52-55.
    33) Allen RW, Ogden B, Bentley FL, et al: Fetal hydantoin syndrome, neuroblastoma, and hemorrhagic disease in a neonate. JAMA 1980; 244:1464.
    34) Ambrosetto G, Tassinari CA, & Baruzzi A: Phenytoin encephalopathy as probable idiosyneratic reaction: case report. Epilepsia 1977; 18:405-408.
    35) 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.
    36) Anon: American academy of pediatrics committee on drugs: transfer of drugs and other chemicals into human milk. Pediatrics 2001; 108(3):776-789.
    37) Anon: Breastfeeding and Maternal Medication. World Health Organization, Geneva, Switzerland, 1995.
    38) Anon: Neurogenic nystagmus. In: Duke-Elder (Ed): System of Ophthalmology, CV Mosby, St Louis, MO, 1971, pp 861.
    39) Anon: Vitamin supplements. Med Lett Drugs Ther 1985; 27:66-68.
    40) Anseeuw K, Mowry JB, Burdmann EA, et al: Extracorporeal Treatment in Phenytoin Poisoning: Systematic Review and Recommendations from the EXTRIP (Extracorporeal Treatments in Poisoning) Workgroup. Am J Kidney Dis 2016; 67(2):187-197.
    41) Arnold K & Gerber N: The rate of clearing of diphenylhydantoin in human plasma. Clin Pharmacol Ther 1970; 11:121.
    42) Arya R, Gulati S, Kabra M, et al: Folic acid supplementation prevents phenytoin-induced gingival overgrowth in children. Neurology 2011; 76(15):1338-1343.
    43) Ashworth B & Horn DB: Evidence of osteomalacia in an outpatient group of adult epileptics. Epilepsia 1977; 18:37.
    44) Australian Government Department of Health and Ageing Therapeutic Goods Administration: Prescribing medicines in pregnancy database. Australian Government Department of Health and Ageing Therapeutic Goods Administration. Woden, Australia. 2012. Available from URL: http://www.tga.gov.au/hp/medicines-pregnancy.htm. As accessed 2012-05-31.
    45) Baciewicz AM, Self TH, & Bekemeyer WB: Update of rifampin drug interactions. Arch Intern Med 1987; 147:565-568.
    46) Baier WK, Beck U, & Hirsch W: CT findings following diphenylhydantoin intoxication. Pediatr Radiol 1985; 15:220-221.
    47) Ballek RE, Reidenberg MM, & Orr L: Inhibition of diphenylhydantoin metabolism by chloramphenicol. Lancet 1973; 1:150.
    48) Banerjee A, Brotherston TM, Lamberty BG, et al: Cancer chemotherapy agent-induced perivenous extravasation injuries. Postgrad Med J 1987; 63(735):5-9.
    49) Bardy AH, Hiilesmaa VK, & Teramo KAW: Serum phenytoin during pregnancy, labor and puerperium. Acta Neurol Scand 1987; 75:374-375.
    50) Barr M Jr, Poznanski AK, Schmickel RD, et al: Digital hypoplasia and anticonvulsants during gestation: a teratogenic syndrome?. J Pediatr 1974; 84:254-256.
    51) Barron SA: Cardiac arrhythmias after small IV dose of phenytoin. N Engl J Med 1976; 295:678.
    52) Bartle WR, Walker SE, & Shapero T: Dose-dependent effect of cimetidine on phenytoin kinetics. Clin Pharmacol Ther 1983; 33:649-655.
    53) Baselt RC & Cravey RH: Disposition of Toxic Drugs and Chemicals in Man, 3rd ed. Year Book Medical Publishers, Chicago, IL, 1989.
    54) Baselt RC: Disposition of Toxic Drugs and Chemicals in Man, 2nd ed, Biomedical Publications, Davis, CA, 1982, pp 643-647.
    55) Baselt RC: Disposition of Toxic Drugs and Chemicals in Man, 5th ed, Chemical Toxicology Institute, Foster City, CA, 2000.
    56) Battino D, Estienne M, & Avanzini G: Clinical pharmacokinetics of antiepileptic drugs in paediatric patients. Clin Pharmacokinet 1995; 29:341-369.
    57) Bauer LA & Blouin RA : Phenytoin Michaelis-Menten pharmacokinetics in Caucasian paediatric patients. Clin Pharmacokinet 1983; 8(6):545-549.
    58) Bauer LA & Blouin RA: Age and phenytoin kinetics in adult epileptics. Clin Pharmacol 1982; 31:301-304.
    59) Bayer AS, Targan SR, Pitchon HE, et al: Dilantin toxicity: miliary pulmonary infiltrates and hypoxemia (letter). Ann Intern Med 1976; 85:475-476.
    60) Bayer AU, Thiel HJ, & Zrenner E: Color vision tests for early detection of antiepileptic drug toxicity. Neurology 1997; 48:1394-1397.
    61) Bazemore RP & Zuckermann FC: On the problem of diphenylhydantoin induced seizures. Arch Neurol 1974; 31:243.
    62) Beech E, Mathur SVS, & Harrold BP: Phenytoin toxicity produced by tolbutamide. Br Med J 1988; 297:1613-1614.
    63) Bellin MF, Jakobsen JA, Tomassin I, et al: Contrast medium extravasation injury: guidelines for prevention and management. Eur Radiol 2002; 12(11):2807-2812.
    64) Benton J: Systemic lupus erythematosus occurring during anticonvulsant drug therapy. JAMA 1962; 180:115.
    65) Berg JM, Rivey MP, & Vern BA: Phenytoin and folic acid: individualized drug-drug interaction. Ther Drug Monitor 1983; 5:395-399.
    66) Berigan T & Watt TJ: Dilantin toxicity presenting as mutism following severe head injury: case report. Military Medicine 1994; 159:533-534.
    67) Berry JM, Kowalski A, & Fletcher SA: Sudden asystole during craniotomy: unrecognized phenytoin toxicity. J Neurosurg Anesthesiol 1999; 11:42-45.
    68) Betteridge T & Fink J : Phenytoin toxicity and thyroid dysfunction. N Z Med J 2009; 122(1303):102-104.
    69) Biechel J: Hydantoin derivatives and malignancies of the haematopoietic system. Acta Med Scand 1975; 198:327.
    70) Binder L, Trujillo J, & Parker D: Association of intravenous phenytoin toxicity with demographic, clinical, and dosing parameters. Am J Emerg Med 1996; 14:398-401.
    71) Black DM & Fivenson DP: Case report of antiepileptic drug hypersensitivity reaction: pseudolymphoma syndrome. Fam Pract Res J 1989; 8:107-111.
    72) Black J, Hannaman T, & Malone C: The relationship of serum albumin level to phenytoin toxicity. J Clin Pharmacol 1987; 27:249-250.
    73) Blackburn SCF, Oliart AD, Garcia Rodriguez LA, et al: Antiepileptics and blood dyscrasias: a cohort study. Pharmacotherapy 1998; 18(6):1277-1283.
    74) Blain H, Hamdan KA, Blain A, et al: Aplastic anemia induced by phenytoin: a geriatric case with severe folic acid deficiency (letter). J Am Geriatr Soc 2002; 50(2):396-401.
    75) Blair AAD, Hallpike JF, & Lascelles PT: Acute diphenylhydantoin and primidone poisoning treated by peritoneal dialysis. J Neurol Neurosurg Psychiatr 1968; 31:520-523.
    76) Blaser KU, Vozeh S, & Landolt H: Intravenous phenytoin: a loading scheme for desired concentrations. Ann Intern Med 1989; 110:1029-1031.
    77) Blum RA, Wilton JH, & Hilligoss DM: Effect of fluconazole on the disposition of phenytoin. Clin Pharmacol Ther 1991; 49:420-425.
    78) Blumson D & Seabrook M: Oral diphenylhydantoin sodium and cardiovascular toxicity. S Afr Med J 1970; 44:1207.
    79) Blyden GT, Scavone JM, & Greenblatt DJ: Metronidazole impairs clearance of phenytoin but not alprazolam or lorazepam. J Clin Pharmacol 1988; 28:240-245.
    80) Botez MI, Gravel J, & Attig E: Reversible chronic cerebellar ataxia after phenytoin intoxication: possible role of cerebellum in cognitive thought. Neurology 1985; 35:1152-1157.
    81) Bramhall D & Levine M: Possible interaction of ranitidine with phenytoin. Drug Intell Clin Pharm 1988; 22:979-980.
    82) Bramhall D & Levine M: Possible interaction of ranitidine with phenytoin. Drug Intell Clin Pharm 1988a; 22:979-980.
    83) Brandolese R, Scordo MG, & Spina E: Severe phenytoin intoxication in a subject homozygous for CYP2C9*3. Clin Pharmacol Ther 2001; 70:391-394.
    84) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    85) Brouwers PJ, de Boer L, & Guchelaar HJ: Ciprofloxacin-phenytoin interaction (letter). Ann Pharmacother 1997; 31:498.
    86) Brown AS, Hoelzer DJ, & Piercy SA: Skin necrosis from extravasation of intravenous fluids in children. Plast Reconstr Surg 1979; 64(2):145-150.
    87) Brown CG, Kaminsky MJ, & Feroli ER: Delirium with phenytoin and disulfiram administration. Ann Emerg Med 1983; 12:310-313.
    88) Browne TR, Szabo GK, & Evans JE: Carbamazepine increases phenytoin serum concentration and reduces phenytoin clearance (Abstract). Neurology 1988; 38:1146-1150.
    89) Brundage RC, Cloyd JC, & Leppik IE: Effect of progabide on serum phenytoin and carbamazepine concentrations. Clin Neuropharmacol 1987; 10:545-554.
    90) Bruni J, Gallo JM, & Lee CS: Interaction of valproic acid with phenytoin. Neurology 1980; 30:1233-1236.
    91) Buchanan RA: Metabolism of diphenylhydantoin following once-daily administration. Neurology 1972; 22:126.
    92) Buchwald AL: Mephenytoin overdose - phenytoin poisoning incognito? Case report and mephenytoin/phenytoin comparison. Clin Toxicol 2000; 38:781-785.
    93) Budavari S: The Merck Index, 12th ed, Merck & Co, Inc, Whitehouse Station, NJ, 1996.
    94) Bulau P, Stoll KD, & Froscher W: Oxcarbazepine versus carbamazepine, in Wolf P, Dam M, Janz D et al (eds): Advances in Epilepsy, 16, Raven Press, New York, NY, 1987, pp 531-536.
    95) Cadle RM, Zenon GJ, & Rodriguez-Barradas MC: Fluconazole-induced symptomatic phenytoin toxicity. Ann Pharmacother 1994; 28:191-195.
    96) Campbell WW Jr: Periodic alternating nystagmus in phenytoin intoxication. Arch Neurol 1980; 37:178-180.
    97) Chaiken BH, Goldberg BI, & Aegar JP: Dilantin sensitivity: report of a case of hepatitis with jaundice, pyrexia and exfoliative dermatitis. Engl J Med 1950; 242:897-898.
    98) Chaikin P & Adir J: Unusual absorption profile of phenytoin in a massive overdose case. J Clin Pharmacol 1987; 27:70-73.
    99) Chait LA & Dinner MI: Ulceration caused by cytotoxic drugs. SA Med J 1975; 49:1935-1936.
    100) Chalhub EG & DeVivo DC: Phenytoin-induced choreoathetosis. J Pediatr 1976; 89:153.
    101) Chamberlain JM, Altieri MA, & Futterman C: A prospective, randomized study comparing intramuscular midazolam with intravenous diazepam for the treatment of seizures in children. Ped Emerg Care 1997; 13:92-94.
    102) Chan BS, Sellors K, Chiew AL, et al: Use of multi-dose activated charcoal in phenytoin toxicity secondary to genetic polymorphism. Clin Toxicol (Phila) 2015; 53(2):131-133.
    103) Chang SI & McAuley JW: Pharmacotherapeutic issues for women of childbearing age with epilepsy. Ann Pharmacother 1998; 32:794-801.
    104) Chaplin S, Sanders GL, & Smith JM: Drug excretion in human breast milk. Adv Drug React Ac Pois Rev 1982; 1:255-287.
    105) Chi WM, Chua KSG, & Kong KH: Phenytoin-induced asterixis - uncommon or under-diagnosed?. Brain Injury 2000; 14:847-850.
    106) Chien LT: Diphenylhydantoin fatal hepatic necrosis. Alabama J Med Sci 1970; 7:318.
    107) Chin RF , Neville BG , Peckham C , et al: Treatment of community-onset, childhood convulsive status epilepticus: a prospective, population-based study. Lancet Neurol 2008; 7(8):696-703.
    108) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    109) Choovivathanavanich P: Pseudolymphoma induced by diphenylhydantoin. J Pediatr 1970; 76:621.
    110) Christensen LK & Skovsted L: Inhibition of drug metabolism by chloramphenicol. Lancet 1969; 2:1397.
    111) Chua HC, Venketasubramanian N, & Tjia H: Elimination of phenytoin in toxic overdose. Clin Neurol Neurosurg 2000; 102:6-8.
    112) Churchwell MD, Pasko DA, Smoyer WE, et al: Enhanced clearance of highly protein-bound drugs by albumin-supplemented dialysate during modeled continuous hemodialysis. Nephrol Dial Transplant 2009; 24(1):231-238.
    113) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    114) Clarke WR, Horn JR, & Kawabori I: Potentially serious drug interactions secondary to high-dose diltiazem used in the treatment of pulmonary hypertension. Pharmacotherapy 1993; 13:402-405.
    115) Cleland PG: Risk-benefit assessment of anticonvulsants in women of child-bearing potential. Drug Saf 1991; 6:70-81.
    116) Closson RG: Interaction between valproic acid and phenytoin. West J Med 1983; 138:108.
    117) Cochat P, Hartemann E, & Duc H: Necrose cerebelleuse localisee apres surdosage en diphenyl-hydantoine. Arch Fr Pediatr 1987; 44:824-825.
    118) Cohan RH, Ellis JH, & Garner WL: Extravasation of radiographic contrast material: recognition, prevention, and treatment. Radiology 1996; 200(3):593-604.
    119) Cohen AD, Reichental E, & Halevy S: Phenytoin-induced severe cutaneous drug reactions: suspected interactions with corticosteroids and H2- blockers. Isr Med Assoc J 1999; 1:95-97.
    120) Cohen MM: Neoplasia and the fetal alcohol and hydantoin syndromes. Neurobehav Toxicol Tetratol 1981; 3:161.
    121) Comer JB: Extravasation from intravenous phenytoin. J Intravenous Ther Clin Nutr 1984; 11:23-29.
    122) Coustan D: Antiarrhythmic agents during pregnancy (letter). JAMA 1982; 247:303.
    123) Coutselinis A, Dimopoulos G, & Versame P: Fatal intoxication with diphenylhydantoin: report of two cases. Forensic Sci 1975; 6:131-133.
    124) Craig S: Phenytoin overdose complicated by prolonged intoxication and residual neurological deficits. Emerg Med Aust 2004; 16:361-365.
    125) Craig S: Phenytoin poisoning. Neurocrit Care 2005; 3(2):161-170.
    126) Crawford SE & Jones CK: Fatal liver necrosis and diphenylhydantoin sensitivity. Pediatrics 1962; 595-600.
    127) Creamer JD, Whittaker SJ, & Kerrmuir M: Phenytoin-induced toxic epidermal necrolysis - a case report. Clin Exp Dermatol 1996; 21:116-120.
    128) Crosby SS, Murray KM, Marvin JA, et al: Management of Stevens-Johnson syndrome. Clin Pharm 1986; 5:682-689.
    129) Curtis DL, Piibe R, & Ellenhorn MJ: Phenytoin toxicity: a review of 94 cases. Vet Hum Toxicol 1989; 32:164-165.
    130) Czajka PA, Anderson WH, & Christoph RA: A pharmacokinetic evaluation of peritoneal dialysis for phenytoin intoxication. J Clin Pharmacol 1980; 20:565-569.
    131) 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.
    132) 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.
    133) Daroff RB: Ocular oscillations. Ann Otol Rhinol Laryngol 1977; 86:102.
    134) Day RE & Chambers RE: Salivary anticonvulsant levels in children with epilepsy. Br J Clin Pract 1981; 33:25.
    135) DeSchoenmakere G, DeWaele J, Terryn W, et al: Phenytoin intoxication in critically ill patients. Am J Kidney Dis 2005; 45(1):189-192.
    136) Demers-Desrosiers LA: Acute psychosis precipitated by withdrawal of anticonvulsant medication. Am J Psychiatry 1978; 135:981.
    137) Dent CE, Richens A, & Rowe DJ: Osteomalacia with long-term anticonvulsant therapy in epilepsy. Br Med J 1970; 4:69-72.
    138) Derby BM & Ward JW: The myth of red urine due to phenytoin. JAMA 1983; 249:1723-1724.
    139) Dethloff LA, Graziano MJ, & Goldenthal E: Perspective on the carcinogenic potential of phenytoin based on rodent tumor bioassays and human epidemiological data. Hum Exp Toxicol 1996; 15:335-348.
    140) Dolgin JG, Nix DE, & Sanchez J: Pharmacokinetic simulation of the effect of multiple-dose activated charcoal in phenytoin poisoning-report of two pediatric cases. DICP 1991; 25:646-649.
    141) Dommisse J: Organic mania induced by phenytoin. Can J Psych 1990; 35:457.
    142) Donahue SR, Flockhart DA, & Abernethy DR: Ticlopidine inhibition of phenytoin metabolism mediated by potent inhibition of CYP2C19. Clin Pharmacol Ther 1997; 62:572-577.
    143) Dorn JM: A case of phenytoin toxicity possibly precipitated by trazodone. J Clin Psychiatry 1986; 47:89-90.
    144) Dorr RT & Fritz WL: Cancer Chemotherapy Handbook, Elsevier, New York, NY, 1980.
    145) Driscoll DF, McMahon M, & Blackburn GL: Phenytoin toxicity in a critically ill, hypoalbuminemic patient with normal serum drug concentrations. Crit Care Med 1988; 16:1248-1249.
    146) Duma RJ, Hendry CN, & Donahoo JS: Hypersensitivity to Diphenylhydantoin (Dilantin): a case report with toxic hepatitis. South Med J 1966; 59:168-170.
    147) Duncan JS, Shorvon SD, & Trimble MR: Withdrawal symptoms from phenytoin, carbamazepine and sodium valproate. J Neurol Neurosurg Psychiatry 1988; 51:924-928.
    148) 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/.
    149) Eadie MJ: Plasma level monitoring of anticonvulsants. Clin Pharmacokinet 1976; 1:52.
    150) Earnest MP, Marx JA, & Drury LR: Complications of intravenous phenytoin for acute treatment of seizures. JAMA 1983; 249:762-765.
    151) Eastwood JB, Phillips ME, & De Wardner HE: Effect of vitamin D3 (Cholecalciferol) in anticonvulsant osteomalacia. Metab Bone Dis Rel Res 1980; 2:83-86.
    152) Ebeid MR, Feldman S, & Parks B: An atypical hypersensitivity reaction. Hosp Pract 1989; 24:57-62.
    153) Edwards JJ & Bosek V: Extravasation injury of the upper extremity by intravenous phenytoin. Anesth Analg 2002; 94(3):672-673.
    154) Ehrenbard LT & Chaganti RSK: Cancer in the fetal hydantoin syndrome. Lancet 1981; 2:97.
    155) Eisenstein SJ & Coleman GC: Reversible bone marrow granulomata and fever induced by phenytoin administration. J Fam Pract 1989; 29:564-565.
    156) Eldridge R: Baltic myoclonus epilepsy: hereditary disorder of childhood made worse by phenytoin. Lancet 1983; 2:838.
    157) Ellenhorn MJ & Barceloux DG: Medical Toxicology: Diagnosis and Treatment of Human Poisoning, Elsevier, New York, NY, 1988.
    158) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    159) Engel JN, Mellul VG, & Goodman DBP: Phenytoin hypersensitivity: a case of severe acute rhabdomyolysis. Am J Med 1986; 81:928-930.
    160) Esser KJ, Kotlarek F, & Habedank M: Chromosomal investigations in epileptic children during long-term therapy with phenytoin or primidone. Hum Genet 1981; 56:345-348.
    161) Evers ML, Izhar A, & Aqil A: Cardiac monitoring after phenytoin overdose. Heart Lung 1997; 26:325-328.
    162) Eyer F, Felgenhauer N, Pfab R, et al: Treatment of severe intravenous phenytoin overdose with hemodialysis and hemoperfusion. Medical science monitor - international medical journal of experimental and clinical research 2008; 14(12):CS145-CS148.
    163) FDA: Fluoxetine-phenytoin interaction. FDA: FDA Med Bull 1994; 24:3-4.
    164) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    165) Fariss BL & Lutcher CL: Diphenylhydantoin-induced hyperglycemia and impaired insulin release: effect of dosage. Diabetes 1971; 20:177.
    166) Fenoglio JJ Jr, McAllister HA Jr, & Mullick FG: Drug related myocarditis. I. Hypersensitivity myocarditis. Hum Pathol 1981; 12:900-907.
    167) Finch E & Lorber J: Methaemoglobinaemia in the newborn. J Obstet Gynecol Br Emp Common 1954; 61:833.
    168) Fine A, Henderson IS, & Morgan DR: Malabsorption of frusemide caused by phenytoin. Br Med J 1977; 4:1061-1062.
    169) Finnell RH, Abbott LC, & Taylor SM: The fetal hydantoin syndrome: Answers from a mouse model. Reprod Toxicol 1989; 3:127-133.
    170) Flowers FP, Araujo OE, & Hamm KA: Phenytoin hypersensitivity syndrome. J Emerg Med 1987; 5:103-108.
    171) Fogelman I: Do anticonvulsant drugs commonly induce osteomalacia?. Scot Med J 1982; 27:136.
    172) Frantzen E: Phenytoin (dilantin) intoxication. Acta Neurol Scand 1967; 43:440.
    173) Frigo GM, Lecchini S, & Caravaggi M: Reduction in phenytoin clearance caused by cimetidine. Eur J Clin Pharmacol 1983; 25:135-137.
    174) Fruchter L & Laptook A: Diphenylhydantoin hypersensitivity reaction associated with interstitial pulmonary infiltrates and hypereosinophilia. Ann Allergy 1981; 47(6):453-455.
    175) Fuerst RH, Graves NM, & Leppik IE: Felbamate increases phenytoin but decreases carbamazepine concentrations. Epilepsia 1988; 29:488-491.
    176) Gaies E, Charfi R, Trabelsi S, et al: Acute phenytoin intoxication: two cases report and literature review. Therapie 2011; 66(5):461-463.
    177) Gaily E: Distal phalangeal hypoplasia in children with prenatal phenytoin exposure: results of a controlled anthropometric study. Am J Med Genet 1990; 35:574-578.
    178) Garcia-Suarez J, Dominguez-Franjo P, & Delcampo JF: EBV-positive non-Hodgkins lymphoma developing after phenytoin therapy. Br J Haematol 1996; 95:376-379.
    179) Garrison SJ & Henson HK: Dilantin toxicity and vegetative depression: a report of two cases. Arch Phys Med Rehabil 1990; 71:422-423.
    180) Gellerman GL & Martinez C: Fatal ventricular fibrillation following intravenous sodium diphenylhydantoin therapy. JAMA 1967; 200:161-162.
    181) Gennis MA, Vemuri R, & Burns EA: Familial occurrence of hypersensitivity to phenytoin. Am J Med 1991; 91:631-634.
    182) Gill MA, Kern JW, & Kaneko J: Phenytoin overdose kinetics. West J Med 1978; 128:246-248.
    183) Gillman MA & Sandyk R: Phenytoin toxicity and co-trimoxazole. Ann Intern Med 1985; 102:559.
    184) Gilman AG, Goodman LS, & Gilman A: Goodman and Gilman's The Pharmacological Basis of Therapeutics, 6th ed, Macmillan Publishing Co Inc, New York, NY, 1980, pp 453.
    185) Gilman AG, Goodman, & Rall TW: Goodman and Gilman's The Pharmacological Basis of Therapeutics, 8th ed, Macmillan Publishing Co, New York, NY, 1990.
    186) Godolphin W, Trepanier J, & Farrell K: Serum and plasma for total and free anticonvulsant drug analyses: effects on EMIT(R) assays and ultrafiltration devices. Therap Drug Monit 1983; 5:319-323.
    187) Goldman AS, Fishman CL, & Baker MK: Phenytoin teratogenicity in the primary and secondary mouse embryonic palate is influenced by the H-2 histocompatibility locus. Proc Soc Exp Biol Med 1983; 173:82-86.
    188) Goldman AS: Biochemical mechanism of glucocorticoid-and phenytoin-induced cleft palate. Curr Top Dev Biol 1984; 19:217-239.
    189) Goldschlager AW & Karliner JS: Ventricular standstill after intravenous diphenylhydantoin. Am Heart J 1967; 74:410-412.
    190) Goldstein N: Drug eruptions from anticonvulsant drugs. Arch Dermatol 1963; 87:118.
    191) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    192) Gore JM, Haffajee CI, & Alpert JS: Interaction of amiodarone and diphenylhydantoin. Am J Cardiol 1984; 54:1145.
    193) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    194) Grant SM & Heel RC: Vigabatrin: a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in epilepsy and disorders of motor control. Drugs 1991; 41:889-926.
    195) Grant WM & Schuman JS: Toxicology of the Eye, 4th ed, Charles C Thomas, Springfield, IL, 1993a.
    196) Grant WM & Schuman JSGrant WM & Schuman JS: Toxicology of the Eye, 4th. Charles C Thomas, Springfield, IL, 1993.
    197) Graves NM, Holmes GB, & Fuerst RH: Effect of felbamate on phenytoin and carbamazepine serum concentrations. Epilepsia 1989; 30:225-229.
    198) Greco MA, Klein S, & Bigelow B: Lymphocytosis in the first-trimester placenta of a mother taking diphenylhydantoin. N Engl J Med 1973; 289(16):867-868.
    199) Griffiths ML, Kaplan H, & Monteagudo FSE: Phenytoin overdose. S Afr Med J 1987; 71:471.
    200) Grillone G & Myssiorek D: Otolaryngologic manifestations of phenytoin toxicity. Clin Otolaryngol 1992; 17:185-191.
    201) Grillone G & Myssiorek D: Otolaryngologic manifestations of phenytoin toxicity. Clin Otolaryngol 1992a; 17(185):191.
    202) Grinder D, Guastella CP, & Pellegrino M: Soft-tissue damage and intravenous phenytoin. Drug Intell Clin Pharm 1988; 22:725-726.
    203) Grosz HJ: Dilantin(R) intoxication - with a report of one case. Am Pract Digest Treat 1956; 7:1633-1636.
    204) 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.
    205) Gupta V, Yadav TP, & Yadav A: Phenytoin toxicity presenting as acute meningo-encephalitis in children. Neurol India 2011; 59(1):66-67.
    206) HSDB : Hazardous Substances Data Bank. National Library of Medicine. Bethesda, MD (Internet Version). Edition expires 1991; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    207) HSDB : Hazardous Substances Data Bank. National Library of Medicine. Bethesda, MD (Internet Version). Edition expires 1995; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    208) Hagan HJ III & Hastings H: Extravasation of phenytoin in the hand. J Hand Surg 1988; 13A:942-943.
    209) Hahn TJ & Halstead LR: Anticonvulsant drug-induced osteomalacia: alterations in mineral metabolism and response to vitamin D3 administration. Calc Tissue Intern 1979; 27:13-18.
    210) Hall TG, Cuddy PG, & Glass CJ: Effect of sucralfate on phenytoin bioavailability. Drug Intell Clin Pharm 1986; 20:607-611.
    211) Hannon MG & Lee SK: Extravasation injuries. J Hand Surg Am 2011; 36(12):2060-2065.
    212) Hansen JM, Kampmann JP, & Siersbaek-Nielsen K: The effect of different sulfonamides on phenytoin metabolism in man. Acta Med Scand Suppl 1979; 624:106-110.
    213) Hansen JM, Siersboek-Nielsen K, & Skovsted L: Carbamazepine-induced acceleration of diphenylhydantoin and warfarin metabolism in man. Clin Pharmacol Ther 1971; 12:539-543.
    214) Hansen JM: Dicumarol-induced diphenylhydantoin intoxication. Lancet 1966; 2:265.
    215) Hanson JW & Smith DW: The fetal hydantoin syndrome. J Pediatr 1975; 87:285.
    216) Hanson JW, Myrianthopoulos NC, Harvey MA, et al: Risks to the offspring of women treated with hydantoin anticonvulsants with emphasis on the fetal hydantoin syndrome. J Pediatr 1976; 89:662-668.
    217) Hansten PD: Drug Interactions Newsletter: Warfarin (Coumadin) and phenytoin (Dilantin), 4, Applied Therapeutics, Inc, Spokane, WA, 1984, pp 25-28.
    218) Hansten PD: Interactions between anticonvulsant drugs: primidone, diphenylhydantoin and phenobarbital. Northwest Med J 1974; 1:17.
    219) Harney J & Glasberg MR: Myopathy and hypersensitivity to phenytoin. Neurology () 1983; 33:790.
    220) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    221) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    222) Helmy ES & Tripplet R: Phenytoin syndrome developing after administration of dilantin for an enflurane-induced seizure. J Oral Maxillofac Surg 1988; 46:52-58.
    223) Hempel E & Klinger W: Drug stimulated biotransformation of hormonal steroid contraceptives; clinical implications. Drugs 1976; 12:442-448.
    224) Hetzel DJ, Bochner F, & Hallpike JF: Cimetidine interaction with phenytoin. Br Med J 1981; 282:1512.
    225) Hey E: Effect of maternal anticonvulsant treatment on neonatal blood coagulation. Arch Dis Child Fetal Neonatal Ed 1999; 81:F208-F210.
    226) Hicks R, Dysken MW, & Davis JM: The pharmacokinetics of psychotropic medication in the elderly: a review. J Clin Psychiatry 1981; 42:374-385.
    227) Hirsh JD & Conlon PF: Implementing guidelines for managing extravasation of antineoplastics. Am J Hosp Pharm 1983; 40:1516-1519.
    228) Hoff JV, Beatty PA, & Wade JL: Dermal necrosis from dobutamine. N Engl J Med 1979; 300:1280.
    229) Hoffman EW: Phenytoin-induced interstitial nephritis. South Med J 1981; 74:1160-1161.
    230) Hoffman EW: Phenytoin-induced interstitial nephritis. South Med J 1981a; 74:1160-1161.
    231) Hoikka V, Savolainen K, & Karjalainen P: Treatment of osteomalacia in institutionalized epileptic patients on long-term anticonvulsant therapy. Ann Clin Res 1982; 14:72-75.
    232) Holcomb R, Lynn R, & Harvey B: Intoxication with 5,5-diphenylhydantoin. J Pediatr 1972; 80:627-632.
    233) Holmes LB, Coull BA, Dorfman J, et al: The correlation of deficits in IQ with midface and digit hypoplasia in children exposed in utero to anticonvulsant drugs. J Pediatr 2005; 146(1):118-122.
    234) Holtzer CD & Reisner-Keller LA: Phenytoin-induced thrombocytopenia. Ann Pharmacother 1997; 31:435-437.
    235) Horning MG, Butler CM, Howlin I, et al: Drug metabolism in the human neonate. Life Sci 1975; 16:651-671.
    236) Houghton GW & Richens A: The effect of benzodiazepines and pheneturide on phenytoin metabolism in man. Br J Clin Pharmacol 1974; 1:344P-345P.
    237) Houtkooper MA, Lammertsma A, & Meyer JWA: Oxcarbazepine (GP 47.680): a possible alternative to carbamazepine?. Epilepsia 1987; 28:693-698.
    238) Howard CE, Roberts RS, & Ely DS: Use of multiple-dose activated charcoal in phenytoin toxicity. Ann Pharmacother 1994; 28:201-203.
    239) Howitt KM & Oziemski MA: Phenytoin toxicity induced by fluconazole. Med J Aust 1989; 20:603-604.
    240) Hunter J, Maxwell JD, & Stewart DA: Altered calcium metabolism in epileptic children on anticonvulsants. Br Med J 1971; 4:202-204.
    241) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976b; 1:161.
    242) Hvidberg EI & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    243) Hvidberg EI & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976a; 1:161.
    244) 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.
    245) 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.
    246) 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.
    247) 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.
    248) 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.
    249) 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.
    250) Ignoffo RJ & Friedman MA: Therapy of local toxicities caused by extravasation of cancer chemotherapeutic drugs. Cancer Treat Rev 1980; 7(1):17-27.
    251) Illingworth RS: Abnormal substances excreted in human milk. Practitioner 1953; 171:533.
    252) 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.
    253) 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.
    254) Jacobs J: Systemic lupus erythematosus in childhood. Report of thirty-five cases, with discussion of seven apparently induced by anticonvulsant medication, and of prognosis and treatment. Pediatrics 1963; 132:257.
    255) Jacobsen D, Alvik A, & Bredesen JE: Pharmacokinetics of phenytoin in acute adult and child intoxication. Clin Toxicol 1986-87; 24:519-531.
    256) Jalil P: Toxic reaction following the combined administration of fluoxetine and phenytoin: two case reports. J Neurol Neurosurg Psychiatry 1992; 55:412-413.
    257) Jang JJ, Takahashi M, & Furukawa F: Long-term in vivo carcinogenicity study of phenytoin (5,5-diphenylhydantoin) in F344 rats. Food Chem Toxicol 1987; 25:697-702.
    258) Janz D & Schmidt D: Anti-epileptic drugs and failure of oral contraceptives (Letter). Lancet 1974; 1:1113.
    259) Jessen K: Recreational use of phenytoin, marijuana, and alcohol: a case report. Neurology 2004; 62:2330.
    260) Johnson J & Freeman HL: Phenytoin and INH drug interaction report. Br J Psychiatry 1976; 129:511.
    261) Kaneko S, Sato T, & Suzuki K: The levels of anticonvulsants in breast milk. Br J Clin Pharmacol 1979; 7:624-627.
    262) Karlin JM & Kutt H: Acute diphenylhydantoin intoxication following halothane anesthesia. J Pediatr 1970; 76:941-944.
    263) Kawasaki C, Nishi R, & Uekihara S: Charcoal hemoperfusion in the treatment of phenytoin overdose. Am J Kid Dis 2000; 35:323-326.
    264) Kay L, Kampmann JP, & Svendsen TL: Influence of rifampin and isoniazid on the kinetics of phenytoin. Br J Clin Pharmacol 1985; 20:323-326.
    265) Keilholz U & Guthrie GP Jr: Adverse effect of phenytoin on mineralocorticoid replacement with fludrocortisone in adrenal insufficiency. Am J Med Sci 1986; 291:280-283.
    266) Kelly DF & Hope DG: Fatal phenytoin-related toxic epidermal necrolysis: case report. Neurosurg 1989; 25:976-978.
    267) Khafaga YM, Jamshed A, & Allam AAK: Stevens-Johnson syndrome in patients on phenytoin and cranial radiotherapy. Acta Oncologica 1999; 38:111-116.
    268) Kim PM & Wells PG: Phenytoin-initiated hydroxyl radical formation - characterization by enhanced salicylate hydroxylation. Molec Pharmacol 1996; 49:172-181.
    269) Kindig D, Garriott ML, & Parton JW: Diphenylhydantoin is not genotoxic in a battery of short-term cytogenetic assays. Teratogen Carcinogen Mutagen 1992; 12:43-50.
    270) 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.
    271) Kogutt MS: Fetal hydantoin syndrome. South Med J 1984; 77:657-658.
    272) Kooiker JC & Sumi SM: Movement disorder as a manifestation of diphenylhydantoin intoxication. Neurology 1974; 24:68.
    273) Koranyi L & Gero L: Influence of diphenylhydantoin on the effect of streptozotocin. Br Med J 1979; 1:127.
    274) Koren G, Brand N, Halkin H, et al: Kinetics of intravenous phenytoin in children. Pediatr Pharmacol 1984a; 4:31-38.
    275) Koren G, Demitrakoudis D, & Weksberg R: Neuroblastoma after prenatal exposure to phenytoin: Cause and effect?. Teratology 1989a; 40:157-162.
    276) Koren G, Demitrakoudis D, Weksberg R, et al: Neuroblastoma after prenatal exposure to phenytoin: cause and effect?. Teratology 1989; 40:157-162.
    277) Korman LB & Olson MJ: Phenytoin-induced hepatitis, rhabdomyolysis, and renal dysfunction. Clin Pharm 1989; 8:514-515.
    278) Koup JR, Gibaldi M, & McNamara P: Interaction of chloramphenicol with phenytoin and phenobarbital: case report. Clin Pharmacol Ther 1978; 24:571-575.
    279) Kozer E, Parvez S, & Minassian BA: How high can we go with phenytoin?. Ther Drug Monitor 2002; 24:386-389.
    280) Krishnamoorthy KS, Zalneraitis EL, Young RSK, et al: Phenytoin-induced choreoathetosis in infancy: case reports and a review. Pediatrics 1983; 72:831-834.
    281) Kruse K: On the pathogenesis of anticonvulsant drug-induced alterations of calcium metabolism. Eur J Pediatr 1982; 138:202.
    282) Kulkarni PS, Mondkar VP, & Sonawalla AB: Chromosomal studies of peripheral blood from epileptic patients treated with phenobarbital and/or diphenylhydantoin. Food Chem Toxicol 1984; 22:1009-1012.
    283) Kumar PP, Lingappa L, Shah MA, et al: Charcoal hemoperfusion for phenytoin intoxication. Indian Pediatr 2012; 49(2):152-153.
    284) Kurata K, Kido H, & Kobayashi K: Long-lasting movement disorder induced by intravenous phenytoin administration for status epilepticus: a case report. Clin Neuropharmacol 1988; 11:467-471.
    285) Kuruvilla T & Bharucha NE: Cerebellar atrophy after acute phenytoin intoxication. Epilepsia 1997; 38:500-502.
    286) Kushnir M, Weinstein R, & Landau B: Hypothyroidism and phenytoin intoxication. Ann Intern Med 1985; 102:341-342.
    287) Kutt H: Diphenylhydantoin metabolism, blood levels and toxicity. Arch Neurol 1964; 11:642.
    288) Lance S & Ternouth I : Fluoxetine-induced phenytoin toxicity: a clinical reminder about the perils of polypharmacy. N Z Med J 2015; 128(1422):78-79.
    289) Lander CM, Smith MT, & Chalk JB: Bioavailability and pharmacokinetics of phenytoin during pregnancy. Eur J Clin Pharmacol 1984; 27:105-110.
    290) Larsen JR & Larsen LS: Clinical features and management of poisoning due to phenytoin. Med Toxicol Adverse Drug Exp 1989; 4:229-245.
    291) Larsen LS, Sterrett JR, & Whitehead B: Adjunctive therapy of phenytoin overdose: a case report using plasmaphoresis. Clin Toxicol 1986; 24:37-49.
    292) Larson DL: Treatment of tissue extravasation by antitumor agents. Cancer 1982; 49:1796-1799.
    293) Lau KK, Lai CK, & Chan AYW: Phenytoin poisoning after using Chinese proprietary medicines. Hum Experiment Toxicol 2000; 19:385-386.
    294) Laubscher FA: Fatal diphenylhydantoin poisoning: a case report. JAMA 1966; 198:1120-1121.
    295) Lazar JD & Wilner KD: Drug interactions with fluconazole. Rev Infect Dis 1990; 12(Suppl 3):S327-333.
    296) Lazaro RP: Involuntary movements induced by anticonvulsant drugs. Mt Sinai J Med 1982; 49:274-281.
    297) Lee S: Activation of systemic lupus erythematosus by drugs. Arch Intern Med 1966; 117:620.
    298) Leppik IE, Dreifuss FE, & Pledger GW: Felbamate for partial seizures: results of a controlled clinical trial. Neurology 1991; 41:1785-1789.
    299) Letteri JM: Diphenylhydantoin metabolism in uremia. N Engl J Med 1971; 285:648.
    300) Levine M & Sheppard I: Biphasic interaction of phenytoin with warfarin. Clin Pharm 1984; 3:200-203.
    301) Levy RH & Koch KM: Drug interactions with valproic acid. Drugs 1982; 24:543-556.
    302) Lindahl S & Westerling D: Detoxification with peritoneal dialysis and blood exchange after diphenylhydantoin intoxication. Acta Paediatr Scand 1982; 71:665-666.
    303) Lindow J & Wijdicks EFM: Phenytoin toxicity associated with hypoalbuminemia in critically ill patients. Chest 1994; 105:602-604.
    304) Lindvall O & Nilsson B: Cerebellar atrophy following phenytoin intoxication. Ann Neurol 1984; 16:258-260.
    305) Linnoila M, Viukari M, & Vaisanen K: Effect of anticonvulsants on plasma haloperidol and thioridazine levels. Am J Psychiatry 1980; 137:819-821.
    306) Lipson A & Bale P: Ependymoblastoma associated with prenatal exposure to diphenylhydantoin and methylphenobarbitone. Cancer 1985; 55:1859-1862.
    307) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    308) Loiseau P: Treatment of concomitant illneses in patients receiving anticonvulsants. Drug interactions of clinical significance. Drug Safety 1998; 19:495-510.
    309) Lombardi TP, Gailey RA, & Bryant BG: Phenytoin overdose in neonate attributable to product packaging similarities. Am J Hosp Pharm 1989; 46:1999-2000.
    310) Loth TS & Eversmann WW Jr: Treatment methods for extravasation of chemotherapeutic agents. J Hand Surg 1986; 388-396.
    311) Lowry JA, Vandover JC, Degreeff J, et al: Unusual presentation of iatrogenic phenytoin toxicity in a newborn. J Med Toxicol 2005; 1(1):26-29.
    312) Luef G, Burtscher J, & Kremser C: Magnetic resonance volumetry of the cerebellum in epileptic patients after phenytoin overdosages. Eur Neurol 1996; 36:273-277.
    313) Luef G, Chemelli A, & Birbamer G: Phenytoin overdosage and cerebellar atrophy in epileptic patients: clinical and MRI findings. Eur Neurol 1994; 34(suppl 1):79-81.
    314) Luhdorf K & Lund M: Phenytoin-induced hyperkinesia. Epilepsy 1977; 18:409.
    315) Lumholtz B, Siersbaek-Nielsen K, & Skovsted L: Sulfamethizole-induced inhibition of diphenylhydantoin, tolbutamide, and warfarin metabolism. Clin Pharmacol Ther 1975; 17:731-734.
    316) Lund L, Alvan G, Berlin A, et al: Pharmacokinetics of single and multiple doses of phenytoin in man. Eur J Clin Pharmacol 1974; 7:81.
    317) Lund L: Pharmacokinetics of single and multiple doses of phenytoin in man. Eur J Clin Pharmacol 1974; 7:81.
    318) Lunde PK, Rane A, & Yaffe SJ: Plasma protein binding of diphenylhydantoin in man. Clin Pharmacol Ther 1970; 11:846-855.
    319) Luscher TF, Siegenthaler-Zuber G, & Kuhlmann U: Severe hypernatremic coma due to diphenylhydantoin intoxication. Clin Nephrol 1983; 20:268-269.
    320) Lusins JO & Jutkowitz R: Residual cerebellar systems dysfunction and peripheral neuropathy after diphenylhydantoin therapy. Mount Sinai J Med 1972; 39:617.
    321) Lynch DJ, Key JC, & White RR: Management and prevention of infiltration and extravasation injury. Surg Clin North Am 1979; 59:939-949.
    322) Maeda T, Sano N, & Togei K: Lack of carcinogenicity of phenytoin in (C57BL/6 x C3H)F sub(1) mice. J Toxicol Environ Health 1988; 24:111-119.
    323) Mahatma M, Haponik EF, & Nelson S: Phenytoin-induced acute respiratory failure with pulmonary eosinophilia. Am J Med 1989; 87:93-94.
    324) Mamiya K, Kojima K, & Yukawa E: Phenytoin intoxication induced by fluvoxamine. Ther Drug Monitor 2001; 23:75-77.
    325) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    326) Manto M, Preiser JC, & Vincent JL: Hypoglycemia associated with phenytoin intoxication. Clin Toxicol 1996a; 34:205-208.
    327) Manto M, Preiser JC, & Vincent JL: Hypoglycemia associated with phenytoin intoxication. J Toxicol Clin Toxicol 1996; 34:205-208.
    328) Marin LL, Garcia-Penas JJ, Herguedas JL, et al: Phenytoin-induced visual disturbances mimicking Delirium Tremens in a child. Eur J Paediatr Neurol 2010; 14(5):460-463.
    329) Masur H, Elger CE, & Ludolph AC: Cerebellar atrophy following acute intoxication with phenytoin. Neurology 1989; 39:432-433.
    330) Masur H, Fahrendorf G, & Oberwittler C: Cerebellar atrophy following acute intoxication with phenytoin. Neurology 1990; 40:1800.
    331) Mathers W, Kattan H, & Earill J: Development of presenile cataracts in association with high serum levels of phenytoin. Ann Ophthalmol 1987; 19:291-292.
    332) Mauro LS, Mauro VF, & Brown DL: Enhancement of phenytoin elimination by multiple-dose activated charcoal. Ann Emerg Med 1987; 16:1132-1135.
    333) McCluggage LK, Voils SA, & Bullock MR: Phenytoin Toxicity Due to Genetic Polymorphism. Neurocrit Care 2008; Epub:--.
    334) McElnay JC, Uprichard G, & Collier PS: The effect of activated dimethicone and a proprietary antacid preparation containing this agent on the absorption of phenytoin. Br J Clin Pharmacol 1982; 13:501-505.
    335) McFee AF, Tice RR, & Shelby MD: In vivo cytogenetic activity of diphenylhydantoin in mice. Mutat Res 1992; 278:61-68.
    336) McGovern B, Geer VR, & LaRaia PJ: Possible interaction between amiodarone and phenytoin. Ann Intern Med 1984; 101:650-651.
    337) McLain LW Jr, Martin JT, & Allen JH: Cerebellar degeneration due to chronic phenytoin therapy. Ann Neurol 1980; 7:18-23.
    338) McLellan DL & Swash M: Choreoathetosis and encephalopathy induced by phenytoin. BMJ 1974; 2:204.
    339) McLelland J & Jack W: Phenytoin/dexamethasone interaction: a clinical problem (letter). Lancet 1978; 1:1096-1097.
    340) Mellick LB, Morgan JA, & Mellick GA: Presentations of acute phenytoin overdose. AM J Emerg Med 1989; 7:61-67.
    341) Menitove JE: Antigranulocyte antibodies and deranged immune function associated with phenytoin-induced serum sickness. Am J Hematol 1981; 10:277.
    342) Messiaen TG, Plaisier EM, Rossert J, et al: Heavy proteinuria, podocyte hypertrophy and severe systemic manifestations in a patient on phenytoin therapy. Nephrol Dial Transplant 1997; 12:2723-2725.
    343) Michael JR & Rudin ML: Acute pulmonary disease caused by phenytoin. Ann Intern Med 1981; 95:452-454.
    344) Miller MA, Crystal CS, & Patel MM: Hemodialysis and hemoperfusion in a patient with an isolated phenytoin overdose. Am J Emerg Med 2006; 24(6):748-749.
    345) Miller RR, Porter J, & Greenblatt DJ: Clinical importance of the interaction of phenytoin and isoniazid: a report from the Collaborative Drug Surveillance Program. Chest 1979; 75:356-358.
    346) Miller WI: Phenytoin chronic toxicity and associated psychosis. Drug Intell Clin Pharm 1988; 22:1003-1004.
    347) Miners JO, Attwood J, & Birkett DJ: Determinants of acetaminophen metabolism: effect of inducers and inhibitors of drug metabolism on acetaminophen's metabolic pathways. Clin Pharmacol Ther 1984; 35:480-486.
    348) Mirkin B: Diphenylhydantoin: placental transport, fetal localization, neonatal metabolism, and possible teratogenic effects. J Pediatr 1971; 78:329-337.
    349) Mitchell AS & Holland JT: Fluconazole and phenytoin: a predictable interaction. Br Med J 1989; 298:1315.
    350) Mochizuki Y: Peripheral neuropathy in children on long-term phenytoin therapy. Brain Dev 1981; 3:375.
    351) Monks A, Boobis S, & Wadsworth J: Plasma protein binding interaction between phenytoin and valproic acid in vitro. Br J Clin Pharmacol 1978; 6:487-492.
    352) Morijiri Y & Sato T: Factors causing rickets in institutionalized handicapped children on anticonvulsant therapy. Arch Dis Child 1981; 56:446-449.
    353) Mrvos R, Wezorek C, & Krenzelok E: Phenytoin injection resulting in "purple glove syndrome" (abstract). J Tox - Clin Tox 1997; 35(5):502.
    354) Mullick FG & Ishak KG: Hepatic injury associated with diphenylhydantoin therapy. Am J Clin Pathol 1980; 74:442.
    355) Mullick FG & Ishak KG: Hepatic injury associated with diphenylhydantoin therapy. Am J Clin Pathol 1980a; 74:442.
    356) Murray JC, Hill RM, & Hegemier S: Lymphoblastic lymphoma following prenatal exposure to phenytoin. J Pediatr Hematol Oncol 1996; 18:241-243.
    357) NFPA: Fire Protection Guide to Hazardous Materials, 13th ed., National Fire Protection Association, Quincy, MA, 2002.
    358) 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.
    359) 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.
    360) 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.
    361) 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.
    362) Nappi JM: Warfarin and phenytoin interaction (letter). Ann Intern Med 1979; 30:852.
    363) Narcy P, Zorza G, & Taburet AM: Intoxication grave par la phenytoine intraveineuse chez le nouveau-ne. Arch Fr Pediatr 1990; 47:591-593.
    364) 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.
    365) 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.
    366) 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.
    367) 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.
    368) 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.
    369) 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.
    370) 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.
    371) 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.
    372) 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.
    373) 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.
    374) 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.
    375) 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.
    376) 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.
    377) 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.
    378) 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.
    379) 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.
    380) 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.
    381) 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.
    382) 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.
    383) 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.
    384) 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.
    385) 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.
    386) 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.
    387) 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.
    388) 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.
    389) 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.
    390) 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.
    391) 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.
    392) 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.
    393) 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.
    394) 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.
    395) 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.
    396) 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.
    397) 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.
    398) 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.
    399) 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.
    400) 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.
    401) 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.
    402) 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.
    403) 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.
    404) 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.
    405) 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.
    406) 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.
    407) 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.
    408) 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.
    409) 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.
    410) 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.
    411) 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.
    412) 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.
    413) 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.
    414) 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.
    415) 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.
    416) 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.
    417) 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.
    418) 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.
    419) 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.
    420) 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.
    421) 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.
    422) 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.
    423) 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.
    424) 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.
    425) 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.
    426) 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.
    427) 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.
    428) 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.
    429) 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.
    430) 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.
    431) 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.
    432) 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.
    433) 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.
    434) 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.
    435) 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.
    436) 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.
    437) 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.
    438) 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.
    439) National Institutes of Health Clinical Center Nursing Department: SOP: care of the patient receiving intravenous cytotoxics or biological agents. NIH. Bethesda, MDAvailable from URL: http://www.cc.nih.gov/nursing/ivctxsop.html.
    440) National Research Council : Acute exposure guideline levels for selected airborne chemicals, 5, National Academies Press, Washington, DC, 2007.
    441) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 6, National Academies Press, Washington, DC, 2008.
    442) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 7, National Academies Press, Washington, DC, 2009.
    443) National Research Council: Acute exposure guideline levels for selected airborne chemicals, 8, National Academies Press, Washington, DC, 2010.
    444) Nauth-Misir TN: A case of gross overdosage of soluble phenytoin. Br Med J 1948; 2:646.
    445) Neef C & de Voogd-van der Straaten I: An interaction between cytostatic and anticonvulsant drugs. Clin Pharmacol Ther 1988; 43:372-375.
    446) Neumar RW , Otto CW , Link MS , et al: Part 8: adult advanced cardiovascular life support: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2010; 122(18 Suppl 3):S729-S767.
    447) Neuvonen PJ, Lehtovaara R, & Bardy A: Antipyretic analgesics in patients on antiepileptic drug therapy. Eur J Clin Pharmacol 1979; 15:263-268.
    448) Neuvonen PJ, Penttila O, & Lehtovaara R: Effect of antiepileptic drugs on the elimination of various tetracycline derivatives. Eur J Clin Pharmacol 1975; 9:147-154.
    449) Nolan PE Jr, Marcus FI, & Hoyer GL: Pharmacokinetic interaction between intravenous phenytoin and amiodarone in healthy volunteers. Clin Pharmacol Ther 1989; 46:43-50.
    450) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    451) Notelovitz M, Tjapkes J, & Ware M: Interaction between estrogen and dilantin in a menopausal woman (letter). N Engl J Med 1981; 304:788-789.
    452) O'Brien TJ, Cascino GD, & So EL: Incidence and clinical consequence of the purple glove syndrome in patients receiving intravenous phenytoin. Neurology 1998; 51:1034-1039.
    453) Oates RK & Tonge RE: Phenytoin and the pseudolymphoma syndrome. Med J Aust 1971; 2:371.
    454) Odar-Cederlof I & Borga U: Kinetics of diphenylhydantoin in uremic patients: consequences of decreased plasma protein binding. Eur J Clin Pharmacol 1974; 7:31.
    455) Oellerich M & Muller-Vahl H: Ultrafiltration technique compared with equilibrium dialysis and ultracentrifugation to determine protein binding of phenytoin. Clin Pharmacokinetics 1984; 9:61-70.
    456) Olesen OV: Disulfiram (and abuse) as inhibitor of phenytoin metabolism. Acta Pharmacol Toxicol 1966; 24:317.
    457) Olsen JH, Boice JD Jr, & Jensen JP: Cancer among epileptic patients exposed to anticonvulsant drugs. J Natl Cancer Inst 1989; 81:803-808.
    458) Orlandini G & Garini G: Phenytoin-induced nephrotic syndrome (letter). Nephron 1989a; 52(1):109.
    459) Orlandini G & Garini G: Phenytoin-induced nephrotic syndrome. Nephron 1989; 52:109.
    460) Osorio I, Burnstine TH, & Remler B: Phenytoin-induced seizures: a paradoxical effect at toxic concentrations in epileptic patients. Epilepsia 1989; 30:230-234.
    461) Otero MJ, Moran D, & Valverde MP: Interaction between phenytoin and ciprofloxacin (letter). Ann Pharmacother 1999; 33:251-252.
    462) Painter PC, Evans JH, & Lyon JM: A rate nephelometric phenobarbital and phenytoin assay: analytical and correlation studies of a new single-point calibration immunoprecipitation inhibition method. Therap Drug Monit 1983; 5:461-466.
    463) Parry RG, Gordon P, & Mason JC: Phenytoin-associated vasculitis and ANCA positivity: a case report. Nephrol Dial Transplant 1996; 11:357-359.
    464) Patel I, Levy R, & Cutler R: Phenobarbital-valproic acid interaction. Clin Pharmacol Ther 1980; 4:515-521.
    465) 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.
    466) Penttila O, Neuvonen PJ, & Aho K: Interaction between doxycycline and some antiepileptic drugs. Br Med J 1974; 2:470-472.
    467) Perucca E & Richens A: Interaction between phenytoin and imipramine. Br J Clin Pharmacol 1977; 4:485-486.
    468) Perucca E, Hebdige S, & Frigo GM: Interaction between phenytoin and valproic acid: plasma protein binding and metabolic effects. Clin Pharmacol Ther 1980; 28:779-789.
    469) Perucca E: Free level monitoring of antiepileptic drugs: clinical usefulness and case studies. Clin Pharmacokinetics 1984; 9:71-78.
    470) Perucca E: Pharmacokinetic interactions with antiepileptic drugs. Clin Pharmacokinet 1982; 7:57-84.
    471) Petereit LB & Meikle AW: Effectiveness of prednisolone during phenytoin therapy. Clin Pharmacol Ther 1977; 22:912-916.
    472) Peterson GM, Khoo BH, & von Witt RJ: Clinical response in epilepsy in relation to total and free serum levels of phenytoin. Ther Drug Monit 1991; 13:415-419.
    473) Peterson GM, Mclean S, & Aldous S: Plasma protein binding of phenytoin in 100 epileptic patients. Br J Clin Pharmacol 1982; 14:298-300.
    474) Petro DJ, Vannucci RC, & Kulin HE: Diazoxide-diphenylhydantoin interaction (letter). J Pediatr 1976; 89:331-332.
    475) Petty CS, Muelling RJ, & Sindell HW: Accidental, fatal poisoning with diphenylhydantoin (Dilantin). J Forensic Sci 1957; 2:279-286.
    476) Phelps SJ, Baldree LA, & Boucher BA: Neuropsychiatric toxicity of phenytoin: importance of monitoring phenytoin levels. Clin Pediatr 1993; 32:107-110.
    477) Pisani F, Fazio A, & Artesi C: Elevation of plasma phenytoin by viloxazine in epileptic patients: a clinically significant drug interaction. J Neurol Neurosurg Psychiatry 1992; 55:126-127.
    478) Pitner JK, Long LD, & Meyer RP: Phenytoin toxicity in an older patients with slow metabolism and atypical presentation. Pharmacother 1998; 18:218-225.
    479) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    480) Pollak PT & Slayter KL: Hazards of doubling phenytoin dose in the face of an unrecognized interaction with ciprofloxacin. Ann Pharmacother 1997; 31:61-64.
    481) Polman AJ, van der Werf TS, & Tiebosch AT: Early-onset phenytoin toxicity mimicking a renopulmonary syndrome. Eur Respir J 1998; 11:501-503.
    482) Polman AJ, van der Werf TS, Tiebosch ATMC, et al: Early-onset phenytoin toxicity mimicking a renopulmonary syndrome. Eur Respir J 1998a; 11:501-503.
    483) Praveen-kumar S & Desai M: Ocular motor abnormalities in a patient with phenytoin toxicity--case report and minireview. Clin Neurol Neurosurg 2014; 127:116-117.
    484) Product Information: DILANTIN INFATABS(R) oral tablets, phenytoin oral tablets. Parke-Davis, New York, NY, 2006.
    485) Product Information: DILANTIN KAPSEALS(R) extended oral capsules, phenytoin sodium extended oral capsules. Parke-Davis, New York, NY, 2007.
    486) Product Information: DILANTIN(R) INFATABS(R) oral chewable tablets, phenytoin oral chewable tablets. Parke-Davis (per DailyMed), New York, NY, 2011.
    487) Product Information: DILANTIN(R) KAPSEALS(R) oral capsule, phenytoin sodium oral capsule. Parke-Davis, New York, NY, 2003.
    488) Product Information: DILANTIN(R) extended capsule, oral, phenytoin sodium extended capsule, oral. Parke-Davis, New York, NY, 2009.
    489) Product Information: DILANTIN(R) oral extended release capsules, phenytoin sodium oral extended release capsules. Parke-Davis (per DailyMed), New York, NY, 2011.
    490) Product Information: DILANTIN-125(R) oral suspension, phenytoin oral suspension. Warner Lambert Co, New York, NY, 2003.
    491) Product Information: Dilantin(R) Infatabs(R) oral chewable tablets, phenytoin oral chewable tablets. Parke-Davis (per DailyMed), New York, NY, 2014.
    492) Product Information: Dilantin(R) intravenous injection solution, phenytoin sodium intravenous injection solution. Pfizer (Per FDA), New York, NY, 2011.
    493) Product Information: Dilantin(R) oral extended release capsules, phenytoin sodium oral extended release capsules. Parke-Davis (per DailyMed), New York, NY, 2014.
    494) Product Information: Dilantin(R), phenytoin. Parke-Davis, Morris Plains, NJ, 1994.
    495) Product Information: Dilantin-125(R) oral suspension, phenytoin oral suspension. Parke-Davis (per DailyMed), New York, NY, 2011.
    496) Product Information: Dilantin-125(R) oral suspension, phenytoin oral suspension. Parke-Davis (per FDA), New York, NY, 2015.
    497) Product Information: PEGANONE(R) oral tablets, ethotoin oral tablets. Abbott Laboratories, North Chicago, IL, 2009.
    498) Product Information: PEGANONE(R) oral tablets, ethotoin oral tablets. Ovation Pharmaceuticals, Inc, Deerfield, IL, 2007.
    499) Product Information: Peganone(R) oral tablets, ethotoin oral tablets. Recordati Rare Diseases Inc. (per DailyMed), Lebanon, NJ, 2013.
    500) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    501) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    502) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    503) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    504) Product Information: phenytoin sodium intravenous, intramuscular injection, phenytoin sodium intravenous, intramuscular injection. Hospira, Inc. (per manufacturer), Lake Forest, IL, 2010.
    505) Pugh RN, Geddes AM, & Yeoman WB: Interaction of phenytoin with chlorpheniramine (letter). Br J Clin Pharmacol 1975; 2:173-175.
    506) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 1995; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    507) RTECS : Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2001; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    508) RTECS: Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health, Cincinnati, Ohio (Internet Version). MICROMEDEX, Greenwood Village, CO, 2001.
    509) RTECS: Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2001a; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    510) Rabinowicz AL, Hinton DR, & Dyck P: High-dose tamoxifen in treatment of brain tumors: interaction with antiepileptic drugs. Epilepsia 1995; 36:513-515.
    511) Rafi JA, Frazier LM, & Driscoll-Bannister SM: Effect of over-the-counter cimetidine on phenytoin concentrations in patients with seizures. Ann Pharmacother 1999; 33:769-774.
    512) Rainey PM, Rogers KE, & Roberts WL: Metabolite and matrix interference in phenytoin immunoassays. Clin Chem 1996; 42:1645-1653.
    513) Rallison M: Lupus erythematosis and Stevens-Johnson Syndrome. Am J Dis Child 1961; 101:81.
    514) Rambeck B: Pharmacological interactions of methsuximide with phenobarbital and phenytoin in hospitalized epileptic patients. Epilepsia 1979; 20:147.
    515) Rameis H: On the interaction between phenytoin and digoxin. Eur J Clin Pharmacol 1985; 29:49-53.
    516) Ramirez JA, Mendell JR, & Warmolts JR: Phenytoin neuropathy: structural changes in the sural nerve. Ann Neurol 1986; 19:162-167.
    517) Rane A, Garle M, Borga O, et al: Plasma disappearance of placentally transferred diphenylhydantoin in the newborn studied by mass fragmentography. Clin Pharmacol Ther 1974; 15:39-45.
    518) Rao VK, Feldman PD, & Dibbell DG: Extravasation injury to the hand by intravenous phenytoin. J Neurosurg 1988; 68:967-969.
    519) Rasmussen S & Kristensen M: Choreoathetosis during phenytoin treatment. Acta Med Scand 1977; 201:239.
    520) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    521) Rentmeester L, Clark R, Joshua A, et al: Undetectable total phenytoin in a patient with elevated kappa light chains: 2015 Annual Meeting of the North American Congress of Clinical Toxicology (NACCT). Clin Toxicol 2015; 53(7):658-658.
    522) Richens A & Rowe DJ: Disturbance of calcium metabolism by anticonvulsant drugs. Br Med J 1970; 4:73-76.
    523) Riedel L & Obe G: Mutagenicity of antiepileptic drugs. II. Phenytoin, primidone and phenobarbital. Mutat Res 1984; 138:71-74.
    524) Rimmer EM & Richens A: Interaction between vigabatrin and phenytoin. Br J Clin Pharmacol 1989; 27:27S-33S.
    525) Rivinus TM: Psychiatric effects of the anticonvulsant regimens. J Clin Psychopharmacol 1982; 2:165-192.
    526) Robins MM: Aplastic anemia secondary to anticonvulsants. Am J Dis Child 1962; 104:614-624.
    527) Robins MM: Aplastic anemia secondary to anticonvulsants. Am J Dis Child 1962a; 104:614-624.
    528) Robinson LJ: Case of acute poisoning from dilantin sodium with recovery. JAMA 1940; 115:289-290.
    529) Roe TF, Podosin RL, & Blaskovics ME: Drug interactions: diazoxide and diphenylhydantoin. J Pediatr 1975; 87:480-484.
    530) Rolan PE, Somogyi AA, & Drew MJ: Phenytoin intoxication during treatment with parenteral miconazole. Br Med J 1983; 287:1760.
    531) Roldan CJ: Phenytoin toxicity from cocaine adulteration. West J Emerg Med 2014; 15(2):127-130.
    532) Ros SP & Black LE: Multiple-dose activated charcoal in management of phenytoin overdose. Pediatr Emerg Care 1989; 5:169-170.
    533) Rose JQ, Choi HK, & Schentag JJ: Intoxication caused by interaction of chloramphenicol and phenytoin. JAMA 1977; 237:2630-2631.
    534) Rose R, Cisek J, & Mickell J: Fosphenytoin-induced bradyasystole arrest in an infant treated with charcoal hemofiltration (abstract). J Tox-Clin Tox 1998; 36:473.
    535) Rosenblum E, Rodichok L, & Hanson PA: Movement disorder as a manifestation of diphenylhydantoin toxicity. Pediatrics 1974; 54:364.
    536) Rothermich NO: Diphenylhydantoin intoxication. Lancet 1966; 2:640.
    537) Roullet E, Koskas P, & Mahieux F: Dyskinesies aigues recidivantes sueles manifestations d'un surdosage en phenytoine. Rev Neurol 1987; 143:836-838.
    538) Rowden AM, Spoor JE, & Bertino JS Jr: The effect of activated charcoal on phenytoin pharmacokinetics. Ann Emerg Med 1990; 19:1144-1147.
    539) Russell MA & Bousvaros G: Fatal results from diphenylhydantoin administered intravenously. JAMA 1968; 206:2118-2119.
    540) 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.
    541) S Sweetman : Martindale: The Complete Drug Reference. Pharmaceutical Press. London, England (Internet Version). Edition expires 2001; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    542) Salem RB, Breland BD, & Mishra SK: Effect of cimetidine on phenytoin serum levels. Epilepsia 1983; 24:284-288.
    543) Sanderson DR: Drug interaction between risperidone and phenytoin resulting in extrapyramidal symptoms (letter). J Clin Psychiatr 1996; 57:177.
    544) Sandy KR: Transient hemiparesis - a rare complication of phenytoin toxicity. Postgrad Med J 1983; 59:601-602.
    545) Sandyk R: Phenytoin toxicity induced by interaction with ibuprofen. S Afr Med J 1982; 62:592.
    546) Sandyk R: Total external ophthalmoplegia induced by phenytoin. S Afr Med J 1984; 65:141-142.
    547) Sarich TC & Wright JM: Hypothyroxinemia and phenytoin toxicity: a vicious circle. Drug Metabol Drug Interact 1996; 13(2):155-160.
    548) Sazie E & Jaffe JP: Severe granulocytopenia with cimetidine and phenytoin (letter). Ann Intern Med 1980; 93:151-152.
    549) Schardein JL: Chemically Induced Birth Defects, Marcel Dekker Inc, Inc, New York, 1985.
    550) Schaumann B, Johnson SB, & Wang N: Sister chromatid exchanges in adult epileptic patients on phenytoin therapy. Environ Mutagen 1985; 7:711-714.
    551) Schaumann BA, Winge VB, & Garry VF: Lack of sister chromatid exchange induction in phenytoin-treated patients with epilepsy. Epilepsia 1989; 30:240-245.
    552) Scheyer RD, Hochholzer JM, & Mattson RH: Near-patient rapid assay of phenytoin concentration. Epilepsia 1995; 36:1152-1154.
    553) Schmeiser M: Todliche vergiftung mit zentropil beim kind. Kinderarztl Prax 1952; 20:158-161.
    554) Schmidt D & Kluge W: Fatal toxic epidermal necrolysis following reexposure to phenytoin: a case report. Epilepsia 1983a; 24:440-443.
    555) Schmidt D & Kluge W: Fatal toxic epidermal necrolysis following reexposure to phenytoin: a case report. Epilepsia 1983; 24:440-443.
    556) Schmidt D, Rohde M, & Wolf P: Clobazam for refractory focal epilepsy. Arch Neurol 1986; 43:824-826.
    557) Schulmeister L: Extravasation management: clinical update. Semin Oncol Nurs 2011; 27(1):82-90.
    558) Scolnik D, Nulman I, & Rovet J: Neurodevelopment of children exposed in utero to phenytoin and carbamazepine monotherapy. JAMA 1994; 271:767-770.
    559) Scott R, Besag FMC, & Neville BGR: Buccal midazolam and rectal diazepam for treatment of prolonged seizures in childhood and adolescence: a randomized trial. Lancet 1999; 353:623-626.
    560) Seeler RA, Israel JN, Royal JE, et al: Ganglioneuroblastoma and fetal hydantoin-alcohol syndromes. Pediatrics 1979; 63:524-527.
    561) Selhorst JB: Diphenylhydantoin-induced cerebellar degeneration. Arch Neurol 1972; 27:453.
    562) Sennoune S, Mesdjian E, & Bonneton J: Interactions between clobazam and standard antiepileptic drugs in patients with epilepsy. Ther Drug Monit 1992; 14:269-274.
    563) Sezzano P: Mutagenicity of diphenylhydantoin and some of its metabolites towards Salmonella typhimurium strains. Mutat Res 1982; 103:219.
    564) Shapiro BH & Babalola GO: Toxicol Lett 1987; 39:165-175.
    565) Sherertz EF, Jegasothy BF, & Lazarus GS: Phenytoin hypersensitivity reaction presenting with toxic epidermal necrolysis and severe hepatitis. J Am Acad Dermatol 1985; 12:178-181.
    566) Sheth KJ, Casper JT, & Good TA: Interstitial nephritis due to phenytoin hypersensitivity. J Pediatr 1977; 91:438.
    567) Sheth KJ: Interstitial nephritis due to phenytoin hypersensitivity. J Pediatr 1977; 91:438.
    568) Shulman LM, Singer C, & Weiner WJ: Phenytoin-induced focal chorea. Movement Disorders 1996; 11:111-113.
    569) Shuttleworth E, Wise G, & Paulson G: Choreoathetosis and diphenylhydantoin intoxication. JAMA 1974a; 230:1170-1171.
    570) Shuttleworth F, Wise G, & Paulson G: Choreo-athetosis and diphenylhydantoin intoxication. JAMA 1974; 230:1170.
    571) Siersbaek-Nielsen K, Skovsted L, & Kampmann J: Sulfamethazole-induced inhibition of diphenylhydantoin and tolbutamide metabolism in man (abstract). Clin Pharmacol Ther 1973; 14:148.
    572) Silberstein LE & Shaw LM: Effect of plasma exchange on phenytoin plasma concentration. Therap Drug Monit 1986; 8:172-176.
    573) Simsek U & Ozyurt M: Phenytoin toxicity causing priapism. Br J Urol 1988; 61:261-268.
    574) Skinner CG, Chang AS, Matthews AS, et al: Randomized controlled study on the use of multiple-dose activated charcoal in patients with supratherapeutic phenytoin levels. Clin Toxicol (Phila) 2012; 50(8):764-769.
    575) Skovsted L, Kristensen M, & Hansen M: The effect of different oral anticoagulants on diphenylhydantoin (DPH) and tolbutamide metabolism. Acta Med Scand 1976; 199:513-515.
    576) Smart HL, Somerville KW, & Williams J: The effects of sucralfate upon phenytoin absorption in man. Br J Clin Pharmacol 1985; 20:238-240.
    577) So EL & Penry JK: Adverse effects of phenytoin on peripheral nerves and neuromuscular junction: a review. Epilepsia 1981a; 22:467-473.
    578) So EL & Penry JK: Adverse effects of phenytoin on peripheral nerves and neuromuscular junction: a review. Epilepsia 1981; 22:467-473.
    579) Sokol DK, Dahlmann A, & Dunn DW: Hyaluronidase treatment for intravenous phenytoin extravasation. J Child Neurol 1998; 13:246-247.
    580) Solomon GE, Hilgartner MW, & Kutt H: Coagulation defects caused by diphenylhydantoin. Neurology 1972; 22:1165-1171.
    581) Sparberg M: Diagnostically confusing complications of diphenylhydantoin therapy: a review. Ann Intern Med 1963; 59:914.
    582) Spengler RF, Arrowsmith JB, & Kilarski DJ: Severe soft-tissue injury following intravenous infusion of phenytoin. Arch Intern Med 1988; 148:1329-1333.
    583) Spielberg SP, Gordon GB, Blake DA, et al: Predisposition to phenytoin hepatotoxicity assessed in vitro. N Engl J Med 1981; 305:722-727.
    584) Spiller HA & Bosse GM: Management of acute anticonvulsant overdose (review). CNS Drugs 1996; 6(2):113-129.
    585) Spiller HA & Rogers GC: Evaluation of administration of activated charcoal in the home. Pediatrics 2002; 108:E100.
    586) Squier W, Hope PL, & Lindenbaum RH: Neocerebellar hypoplasia in a neonate following intra-uterine exposure to anticonvulsants. Dev Med Child Neurol 1990; 32:725-742.
    587) Sreenath TG, Gupta P, Sharma KK, et al: Lorazepam versus diazepam-phenytoin combination in the treatment of convulsive status epilepticus in children: A randomized controlled trial. Eur J Paediatr Neurol 2009; Epub:Epub.
    588) Stalnikowicz R, Mosseri M, & Shaley O: Phenytoin-induced arthritis. Neurology 1982; 32:1317-1318.
    589) Stankler L & Campbell AGM: Neonatal acne vulgaris: a possible feature of the fetal hydantoin syndrome. Br J Dermatol 1980; 103:453.
    590) Steen B, Rane A, Lonnerholm G, et al: Phenytoin excretion in human breast milk and plasma levels in nursed infants. Ther Drug Monit 1982; 4:331-334.
    591) Stilman N & Masdeu JC: Incidence of seizures with phenytoin toxicity. Neurology 1985; 35:1769-1772.
    592) Su CM, Kung CT, Wang YC, et al: Life-threatening cardiotoxicity due to chronic oral phenytoin overdose. Neurol India 2009; 57(2):200-202.
    593) Sun DK, Reiner D, Frishman W, et al: Adverse dermatologic reactions from antiarrhythmic drug therapy. J Clin Pharmacol 1994; 34:953-966.
    594) Sung SF, Chiang PC, Tung HH, et al: Charcoal hemoperfusion in an elderly man with life-threatening adverse reactions due to poor metabolism of phenytoin. J Formos Med Assoc 2004; 103(8):648-652.
    595) Svendsen TL, Kristensen MB, & Hansen JM: The influence of disulfiram on the half-life and metabolic clearance rate of diphenylhydantoin and tolbutamide in man. Eur J Clin Pharmacol 1976; 9:439.
    596) Sylvester RK, Lewis FB, & Caldwell KC: Impaired phenytoin bioavailability secondary to cisplatinum, vinblastine, and bleomycin. Ther Drug Monit 1984; 6:302-305.
    597) Taliercio CP, Olney BA, & Lie JT: Myocarditis related to drug hypersensitivity. Mayo Clin Proc 1985; 60:463-468.
    598) Taneja N, Jain S, & Maheswari MC: Induction of sister chromatid exchanges in phenytoin treated & untreated patients with epilepsy. Indian J Med Res 1992; 96:302-305.
    599) Taylor JW, Alexander B, & Kathol RG: Pharmacokinetic analysis of the phenytoin-disulfiram interaction (abstract). Drug Intell Clin Pharm 1984; 18:499.
    600) Taylor JW, Alexander B, & Lyong LW: Oral anticoagulant-phenytoin interactions. Drug Intell Clin Pharm 1980; 14:669.
    601) Tenckhoff H, Sherrard DJ, & Hickman RO: Acute diphenylhydantoin intoxication. Am J Dis Child 1968; 116:422-425.
    602) Thakore S & Murphy N: The potential role of prehospital administration of activated charcoal. Emerg Med J 2002; 19:63-65.
    603) Thakral A, Shenoy R, & Deleu D: Acute visual dysfunction following phenytoin-induced toxicity. Acta Neurol Belg 2003; 103(4):218-220.
    604) The University of Kansas Hospital: Guide to extravasation management in adult & pediatric patients. The University of Kansas Hospital. Kansas City, KS. 2009. Available from URL: http://www2.kumc.edu/pharmacy/guidelines/Extravasations%20diagram.pdf. As accessed 2012-09-11.
    605) Theil GB, Richter RW, & Powell MR: Acute dilantin poisoning. Neurology 1961; 11:138-142.
    606) Tichner JB & Enselberg CD: Suicidal Dilantin poisoning: case report. N Engl J Med 1951; 245:723-725.
    607) Toler SM, Wilkerson MA, & Porter WH: Severe phenytoin intoxication as a result of altered protein binding in AIDS. DICP 1990; 24:698-700.
    608) Tomita S, Kurokawa T, & Ueda K: Aplastic anaemia induced by intravenous phenytoin and lidocaine administration. Eur J Pediatr 1985; 144:207-208.
    609) Tomita S, Kurokawa T, Ueda K, et al: Aplastic anaemia induced by intravenous phenytoin and lidocaine administration. Eur J Pediatr 1985a; 144:207-208.
    610) Tomsick RS: The phenytoin syndrome. Cutis 1983; 32:535-541.
    611) Tomson T: Choreoathetosis induced by ordinary phenytoin levels, explained by high free fraction? A case report. Ther Drug Monit 1988; 10:239-241.
    612) Travin M, Macris NT, & Block JM: Reversible common variable immunodeficiency syndrome induced by phenytoin. Arch Intern Med 1989; 149:1421-1422.
    613) Treasure T & Toseland PA: Hyperglycemia due to phenytoin toxicity. Arch Dis Child 1971; 46:563.
    614) Treasure T & Toseland PA: Hyperglycemia due to phenytoin toxicity. Arch Dis Child 1971a; 46:563.
    615) Treiman DM & Ben-Menachem E: Inhibition of carbamazepine and phenytoin metabolism by nafimidone, a new antiepileptic drug. Epilepsia 1987; 28:699-705.
    616) Tsan MF: Dilantin, agranulocytosis, and phagocytic marrow histiocytes. Ann Intern Med 1976; 84:710.
    617) Tsatsakis AM, Psillakes T, & Paritsis N: Phenytoin concentration in head hair sections: a method to evaluate the history of drug use. J Clin Psychopharmacol 2000; 20:560-573.
    618) Tse CST & Iagmin P: Phenytoin and ranitidine interaction. Ann Intern Med 1994; 120:892-893.
    619) Tse CST, Akinwande KI, & Biallowons: Phenytoin concentration elevation subsequent to ranitidine administration. Ann Pharmacother 1993; 27:1448-1451.
    620) Turck D, Largilliere C, & Dupuis B: Interaction between diazoxide and phenytoin (letter). Presse Med 1986; 15:31.
    621) Turkdogan D, Onat F, & Ture U: Phenytoin toxicity with mandibular tremor secondary to intravenous administration. Int J Clin Pharmacol Ther 2002; 40:18-19.
    622) Tutuncuoglu S, Kantar M, & Tegul H: Akinetic mutism due to diphenylhydantoin toxicity. Turk J Ped 1997; 39:403-407.
    623) Tutuncuoglu S, Kantar M, Tegul H, et al: Akinetic mutism due to diphenylhydantoin toxicity. Turk J Pediatr 1997a; 39:403-407.
    624) Tyndel F: Interaction of gabapentin with other antiepileptics (letter). Lancet 1994; 343:1363-1364.
    625) 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.
    626) 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.
    627) 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-.
    628) 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.
    629) 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.
    630) 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.
    631) 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.
    632) 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-.
    633) U.S. Occupational Safety, and Health Administration (OSHA): Process safety management of highly hazardous chemicals. 29 CFR 2010 2010; 29(1910.119):348-.
    634) USP: Drug Information for the Health Care Professional, 11th ed, 1, US Pharmacopeial Convention, Inc, Rockville, MD, 1989.
    635) Ueno K, Miyal K, & Mitsuzane K: Phenytoin-tizanidine interaction. DICP 1991; 25:1273.
    636) Unger AH & Sklaroff HJ: Fatalities following intravenous use of sodium diphenylhydantoin for cardiac arrhythmias: a report of two cases. JAMA 1967; 200:159-160.
    637) 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.
    638) Upton J, Mulliken JB, & Murray JE: Major intravenous extravasation injuries. Am J Surg 1979; 137(4):497-506.
    639) Upton J, Mulliken JB, & Murray JE: Major intravenous extravasation injuries. Am J Surg 1979a; 137:497-506.
    640) Vajda FJE, Prineas RJ, & Lovell RRH: Interaction between phenytoin and the benzodiazepines. Br Med J 1971; 1:346.
    641) Vajda FJE: The value of phenytoin plasma levels in the treatment of epilepsy. Med J Aust 1970; 2:1074.
    642) Vallarta JM, Bell DB, & Reichert A: Progressive encephalopathy due to chronic hydantoin toxicity. Am J Dis Child 1974; 128:27.
    643) Vallarta JM: Progressive encephalopathy due to chronic hydantoin toxicity. Am J Dis Child 1974; 128:27.
    644) VanDerLinde LP & Campbell RK: Guidelines for the intravenous administration of drugs. Drug Intell Clin Pharm 1977; 11:30-55.
    645) VanDerLinde LP & Campbell RK: Guidelines for the intravenous administration of drugs. Drug Intell Clin Pharm 1977a; 11:30-55.
    646) Veenith T, Jellema K, Pearce A, et al: Phenytoin toxicity masquerading as motor neurone disease. Anaesthesia 2009; 64(3):337-337.
    647) Vittorio CC & Muglia JJ: Anticonvulsant hypersensitivity syndrome. Arch Intern Med 1995; 155:2285-2290.
    648) Wagner KJ & Leikin JB: Metabolic effects of phenytoin toxicity (letter). Ann Emerg Med 1986; 15:509-510.
    649) Walubo A & Aboo A: Phenytoin toxicity due to concomitant anti-tuberculosis therapy. S Afr Med J 1995a; 85:1175-1176.
    650) Walubo A & Aboo A: Phenytoin toxicity due to concomitant antituberculosis therapy. S Afr Med J 1995; 85:1175-1176.
    651) Watts JC: A fatal case of erythema multiforme exudativum (Stevens-Johnson's syndrome) following therapy with dilantin. Pediatrics 1962; 592-594.
    652) Weichbrodt GD & Elliott DP: Treatment of phenytoin toxicity with repeated doses of activated charcoal. Ann Emerg Med 1987; 16:1387-1389.
    653) Weidle PJ, Skiest DJ, & Forrest A: Multiple-dose activated charcoal as adjunct therapy after chronic phenytoin intoxication. Clin Pharm 1991; 10:711-714.
    654) Wengstrom Y, Margulies A, & European Oncology Nursing Society Task Force: European Oncology Nursing Society extravasation guidelines. Eur J Oncol Nurs 2008; 12(4):357-361.
    655) White RL, Garnett WR, & Allen JH: Phenytoin removal during plasma exchange. J Clin Apheresis 1987; 3:147-150.
    656) Wilder BJ, Buchanan RA, & Serrano EE: Correlation of acute diphenylhydantoin intoxication with plasma levels and metabolite excretion. Neurology 1973a; 23:1329-1332.
    657) Wilder BJ, Serrano EE, & Ramsay RE: Plasma diphenylhydantoin levels after loading and maintenance doses. Clin Pharmacol Ther 1973; 14:797.
    658) Wilson JT, Huff JG, & Kilroy AW: Prolonged toxicity following acute phenytoin overdose in a child. J Pediatr 1979; 95:135-138.
    659) Witmer DR & Ritschel WA: Phenytoin-isoniazid interaction: a kinetic approach to management. Drug Intell Clin Pharm 1984; 18:483-486.
    660) Witter FR, King TM, & Blake DA: Adverse effects of cardiovascular drug therapy on the fetus and neonate. Obstet Gynecol 1981; 58:100S-105S.
    661) Woods DJ, Coulter DM, & Pillans P: Interaction of phenytoin and fluoxetine. N Z Med J 1994; 107:19.
    662) Wyte CD & Berk WA: Severe oral phenytoin overdose does not cause cardiovascular morbidity. Ann Emerg Med 1991; 20:508-512.
    663) Wyte CE & Berk WA: Severe oral phenytoin intoxication: lack of cardiovascular morbidity (abstract 206). Ann Emerg Med 1989; 18:480.
    664) Yalcinkaya C, Tuysuz B, Somay G, et al: Polydactyly and fetal hydantoin syndrome: an additional component of the syndrome?. Clin Genet 1997; 51:343-345.
    665) York RC & Coleridge ST: Cardiopulmonary arrest following intravenous phenytoin loading. Am J Emerg Med 1988; 6:255-259.
    666) Yosowitz P, Ekland DA, & Shaw RC: Peripheral intravenous infiltration necrosis. Ann Surg 1975; 182:553-556.
    667) Yue CP, Mann KS, & Chan KH: Severe thrombocytopenia due to combined cimetidine and phenytoin therapy. Neurosurgery 1987; 20:963-964.
    668) Zerwekh JE, Homan R, Tindall R, et al: Decreased serum 24,25-dihydroxyvitamin D concentration during long-term anticonvulsant therapy in adult epileptics. Ann Neurol 1982; 12:184.
    669) Zerwekh JE: Decreased serum 24,25-dihydroxyvitamin D concentration during long-term anticonvulsant therapy in adult epileptics. Ann Neurol 1982; 12:184.
    670) Zhou LX, Pihlstrom B, & Hardwick JP: Metabolism of phenytoin by the gingiva of normal humans: The possible role of reactive metabolites of phenytoin in the initiation of gingival hyperplasia. Clin Pharmacol Ther 1996; 60:191-198.
    671) Zielinski JJ & Haidukewych D: Dual effects of carbamazepine-phenytoin interaction. Ther Drug Monit 1987; 9:21-23.
    672) Zielinski JJ, Haidukewych D, & Leheta BJ: Carbamazepine-phenytoin interaction: elevation of plasma phenytoin concentrations due to carbamazepine comedication. Ther Drug Monit 1985; 7:51-53.
    673) Zifkin B, Sherwin A, & Andermann F: Phenytoin toxicity due to interaction with clobazam. Neurology 1991; 41:313-314.
    674) Zinsmeister S & Marks RE: Acute athetosis as a result of phenytoin toxicity in a child. Am J Dis Child 1976; 130:75.
    675) de Caen AR, Berg MD, Chameides L, et al: Part 12: Pediatric Advanced Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S526-S542.