MOBILE VIEW  | 

LAMOTRIGINE

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Lamotrigine is a phenyltriazine anticonvulsant with a pharmacological profile similar to phenytoin. It is chemically unrelated to the currently used antiepileptic drugs and has weak antifolate activity. It is currently used for the treatment of seizures and Bipolar I Disorder.

Specific Substances

    1) BW-430C
    2) 6-(2,3-dichlorophenyl)-1,2,4-triazine-3,5-diyldiamine
    3) 3,5-Diamino-6-(2,3-dichlorophenyl)-as-triazine
    4) Lamotrigiini
    5) Lamotrigin
    6) Lamotrigina
    7) Lamotriginum
    8) Lamotrijin
    9) CAS 84057-84-1
    1.2.1) MOLECULAR FORMULA
    1) C9H7N5Cl2

Available Forms Sources

    A) FORMS
    1) Lamotrigine is available in the United States in the following formulations (Prod Info LAMICTAL XR oral extended-release tablets, 2009; Prod Info LAMICTAL chewable dispersible oral tablets, oral tablets, orally disintegrating tablets, 2009):
    a) ORAL TABLETS - 25 mg, 100 mg, 150 mg, 200 mg
    b) CHEWABLE DISPERSIBLE TABLETS - 2 mg, 5 mg and 25 mg
    c) ORAL DISINTEGRATING TABLETS - 25 mg, 50 mg, 100 mg, 200 mg
    d) EXTENDED-RELEASE TABLETS - 25 mg, 50 mg, 100 mg, 200 mg
    B) USES
    1) SEIZURES
    a) Lamotrigine is indicated as adjunctive therapy for partial onset seizures, the generalized seizures of Lennox-Gastaut syndrome, and primary generalized tonic-clonic seizures in adults and pediatric patients 2 years and older. It is also indicated as monotherapy in the treatment of epilepsy in patients 16 years or older who are being converted from carbamazepine, phenobarbital, phenytoin, primidone or valproic acid as the single antiepileptic agent (Prod Info LAMICTAL XR oral extended-release tablets, 2009; Prod Info LAMICTAL chewable dispersible oral tablets, oral tablets, orally disintegrating tablets, 2009).
    2) BIPOLAR I DISORDER
    a) Lamotrigine is indicated as maintenance treatment of Bipolar I Disorder in patients 18 years or older (Prod Info LAMICTAL chewable dispersible oral tablets, oral tablets, orally disintegrating tablets, 2009).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Lamotrigine is a phenyltriazine anticonvulsant with a pharmacological profile similar to phenytoin. It is currently used for the treatment of seizures and Bipolar I Disorder.
    B) PHARMACOLOGY: The exact mechanism of action of lamotrigine has not been fully elucidated. One proposed mechanism is that lamotrigine inhibits sodium channels, resulting in neuronal membrane stabilization and control of excitatory neurotransmitter release.
    C) EPIDEMIOLOGY: Overdose is rare.
    D) WITH THERAPEUTIC USE
    1) COMMON: Weight gain, amnesia, nervousness, and thought disturbances. LESS COMMON: Headache, dizziness, visual disturbances, gastrointestinal disturbances, hypotension, leukopenia, anemia, hematuria, elevated liver enzymes, and rashes. RARE: Disseminated intravascular coagulation, acute renal failure, aseptic meningitis, Stevens Johnson Syndrome, and toxic epidermal necrolysis. Premarketing studies have revealed slight increases in the PR interval in a small number of patients on therapeutic doses. Rhabdomyolysis may result from lamotrigine therapeutic dose. It should be noted that generalized seizures may be a cause of rhabdomyolysis.
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Nausea, vomiting, anorexia, abdominal pain, somnolence, lethargy, slurred speech, xerostomia, blurred vision, diplopia, rash, confusion, nystagmus, ataxia, tremor, agitation, dyskinesia, hyperreflexia, and elevated liver enzymes.
    2) SEVERE TOXICITY: Tachycardia and conduction disturbances, hypokalemia, oculogyric crisis, seizures, rhabdomyolysis, encephalopathy, coma, and respiratory depression.
    0.2.20) REPRODUCTIVE
    A) Lamotrigine is classified as FDA pregnancy category C. Preliminary data have shown an association with cleft palate and/or cleft lip in infants exposed to lamotrigine during the first trimester of pregnancy. There is insufficient information concerning teratogenic effects of lamotrigine in the literature to recommend its use during pregnancy. Lamotrigine has been shown to be excreted in breast milk at low concentrations. Monitor breastfed infants closely for signs or symptoms of lamotrigine toxicity, including apnea, drowsiness, or poor sucking. Because the effects on nursing infants exposed to lamotrigine from the mother's breast milk are not known, administer lamotrigine cautiously to breastfeeding mothers.
    0.2.21) CARCINOGENICITY
    A) At the time of this review, the manufacturer does not report any carcinogenic potential of lamotrigine in humans.

Laboratory Monitoring

    A) The value of plasma lamotrigine monitoring has not been established.
    B) Monitor vital signs, serial CBC with differential, serum electrolytes, liver enzymes, and renal function in symptomatic patients.
    C) Obtain an ECG and institute continuous cardiac monitoring following significant exposures. Overdoses have resulted in ECG abnormalities, including widening QRS complex and PR prolongation.
    D) Monitor for CNS and respiratory depression.
    E) Monitor CPK levels in patients with prolonged coma or seizures.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Management will primarily be symptomatic and supportive. Manage mild hypotension with IV fluids.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Treatment is symptomatic and supportive. Closely monitor neurologic function. Treat seizures with IV benzodiazepines; barbiturates or propofol may be needed if seizures persist or recur. Manage hypotension with IV fluids and pressors if needed. Treat hypokalemia with oral or IV potassium chloride. Consider intravenous lipid therapy early for patients with ventricular dysrhythmias or hypotension.
    C) DECONTAMINATION
    1) PREHOSPITAL: Prehospital gastrointestinal decontamination is not recommended because of the potential for CNS depression and subsequent aspiration.
    2) HOSPITAL: Consider activated charcoal after a potentially toxic ingestion of lamotrigine if the patient is able to maintain airway or in whom airway is protected, or if coingestants dictate it.
    D) AIRWAY MANAGEMENT
    1) Endotracheal intubation may be necessary if life-threatening cardiac dysrhythmias, significant CNS, or respiratory depression develop.
    E) ANTIDOTE
    1) None
    F) SEIZURES
    1) Administer IV benzodiazepines; barbiturates or propofol may be needed if seizures persist or recur.
    G) HYPOTENSION
    1) IV 0.9% NaCl at 10 mL to 20 mL/kg, dopamine, norepinephrine.
    H) VENTRICULAR DYSRHYTHMIAS
    1) Obtain an ECG, institute continuous cardiac monitoring and administer oxygen. Evaluate for hypoxia, acidosis, and electrolyte disorders (particularly hypokalemia, hypocalcemia, and hypomagnesemia). Sodium bicarbonate is generally first line therapy for QRS widening and ventricular dysrhythmias, administer 1 to 2 mEq/kg, repeat as needed to maintain blood pH between 7.45 and 7.55. In patients unresponsive to bicarbonate, consider lidocaine.
    I) LIPID EMULSION
    1) Limited anecdotal evidence suggest that lipid emulsion my be useful in patients with severe cardiac or neurologic toxicity. Administer 1.5 mL/kg of 20% lipid emulsion over 2 to 3 minutes as an IV bolus, followed by an infusion of 0.25 mL/kg/min. Evaluate the patient's response after 3 minutes at this infusion rate. The infusion rate may be decreased to 0.025 mL/kg/min (ie, 1/10 the initial rate) in patients with a significant response. This recommendation has been proposed because of possible adverse effects from very high cumulative rates of lipid infusion. Monitor blood pressure, heart rate, and other hemodynamic parameters every 15 minutes during the infusion. If there is an initial response to the bolus followed by the re-emergence of hemodynamic instability during the lowest-dose infusion, the infusion rate may be increased back to 0.25 mL/kg/min or, in severe cases, the bolus could be repeated. A maximum dose of 10 mL/kg has been recommended by some sources. Where possible, lipid resuscitation therapy should be terminated after 1 hour or less, if the patient's clinical status permits. In cases where the patient's stability is dependent on continued lipid infusion, longer treatment may be appropriate.
    J) RHABDOMYOLYSIS
    1) Administer sufficient 0.9% saline to maintain urine output of 2 to 3 mL/kg/hr. Monitor input and output, serum electrolytes, CK, and renal function. Diuretics may be necessary to maintain urine output. Urinary alkalinization is NOT routinely recommended.
    K) ENHANCED ELIMINATION
    1) Hemodialysis has not been established as an effective means of removing lamotrigine from the blood. Approximately 20% (range, 5.6% to 35.1%) of the amount of lamotrigine present in the body was eliminated by hemodialysis during a 4-hour session. In 6 renal failure patients, approximately 17% of the amount of lamotrigine in the body was removed during 4 hours of hemodialysis.
    L) PATIENT DISPOSITION
    1) OBSERVATION CRITERIA: All patients with deliberate self-harm ingestions should be evaluated in a healthcare facility and monitored until symptoms resolve. Children with unintentional ingestions who are symptomatic should be observed in a healthcare facility.
    2) ADMISSION CRITERIA: Patients with a deliberate ingestions demonstrating cardiotoxicity, or other persistent neurotoxicity should be admitted.
    3) CONSULT CRITERIA: Call a Poison Center for assistance in managing patients with severe toxicity or in whom the diagnosis is unclear.
    M) PITFALLS
    1) When managing a suspected lamotrigine overdose, the possibility of coingestants should be determined.
    N) PHARMACOKINETICS
    1) Lamotrigine is rapidly and completely absorbed after oral administration with an absolute bioavailability of 98%. Protein binding: Approximately 55%. Vd: approximately 0.9 to 1.3 L/kg. Metabolized in the liver primarily by glucuronic acid conjugation into inactive metabolites. Renal excretion: 94% of the drug recovered in the urine. Elimination half-life: 25.4 to 32.8 hours.
    O) DIFFERENTIAL DIAGNOSIS
    1) Includes overdose ingestions of other antiepileptic agents, neurologic disorders.

Range Of Toxicity

    A) TOXICITY: Adults have survived overdoses of greater than 4000 mg with supportive therapy. A man developed generalized myoclonus status epilepticus and coma several hours after ingesting 6 g of lamotrigine. He recovered following supportive care. A 3-year-old girl survived an overdose of 1.15 g of lamotrigine. Another child, a 2-year-old, also survived an overdose of 800 mg. Fatalities have been reported following overdoses of up to 15 g.
    B) THERAPEUTIC: Adult maintenance doses of lamotrigine for bipolar disorder can be up to 400 mg/day and up to 500 mg/day for anti-seizure therapy. For children 2 to 12 years of age, the maintenance dose for the anti-seizure therapy can be up to 15 mg/kg/day (max 400 mg/day). For children over the age of 12, the maintenance dose for the anti-seizure therapy can be up to 500 mg/day.

Summary Of Exposure

    A) USES: Lamotrigine is a phenyltriazine anticonvulsant with a pharmacological profile similar to phenytoin. It is currently used for the treatment of seizures and Bipolar I Disorder.
    B) PHARMACOLOGY: The exact mechanism of action of lamotrigine has not been fully elucidated. One proposed mechanism is that lamotrigine inhibits sodium channels, resulting in neuronal membrane stabilization and control of excitatory neurotransmitter release.
    C) EPIDEMIOLOGY: Overdose is rare.
    D) WITH THERAPEUTIC USE
    1) COMMON: Weight gain, amnesia, nervousness, and thought disturbances. LESS COMMON: Headache, dizziness, visual disturbances, gastrointestinal disturbances, hypotension, leukopenia, anemia, hematuria, elevated liver enzymes, and rashes. RARE: Disseminated intravascular coagulation, acute renal failure, aseptic meningitis, Stevens Johnson Syndrome, and toxic epidermal necrolysis. Premarketing studies have revealed slight increases in the PR interval in a small number of patients on therapeutic doses. Rhabdomyolysis may result from lamotrigine therapeutic dose. It should be noted that generalized seizures may be a cause of rhabdomyolysis.
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Nausea, vomiting, anorexia, abdominal pain, somnolence, lethargy, slurred speech, xerostomia, blurred vision, diplopia, rash, confusion, nystagmus, ataxia, tremor, agitation, dyskinesia, hyperreflexia, and elevated liver enzymes.
    2) SEVERE TOXICITY: Tachycardia and conduction disturbances, hypokalemia, oculogyric crisis, seizures, rhabdomyolysis, encephalopathy, coma, and respiratory depression.

Vital Signs

    3.3.3) TEMPERATURE
    A) WITH THERAPEUTIC USE
    1) FEVER
    a) Fever has been reported with overdose (Dagtekin et al, 2011).
    b) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. One patient developed hyperthermia (temperature greater than 38 degrees C) (Moore et al, 2013).
    c) CASE REPORT: Increased temperature related to leukopenia and sepsis has been reported following 10 days of lamotrigine therapy (Nicholson et al, 1995). Other drugs used by the patient included valproate sodium and propranolol.
    d) CASE REPORT: A 45-year-old woman developed fever, rash, rhabdomyolysis, disseminated intravascular coagulation, and acute renal failure 14 days after beginning lamotrigine therapy (Schaub et al, 1994). Other medications included clonazepam and carbamazepine.
    3.3.5) PULSE
    A) WITH POISONING/EXPOSURE
    1) TACHYCARDIA
    a) CASE REPORT: Tachycardia (150 bpm) was present in a 29-year-old man after intentionally ingesting an unknown number of 200 milligram lamotrigine tablets along with ethanol (Schwartz & Geller, 2007).
    b) CASE REPORT: Increased pulse rate (120 BPM) in a 33-year-old woman following a 1500 to 2000 milligrams lamotrigine overdose (Harchelroad et al, 1994) and in a 26-year-old man following a 1350 milligram lamotrigine overdose (Buckley et al, 1993) have been reported.

Heent

    3.4.3) EYES
    A) WITH THERAPEUTIC USE
    1) OCULOGYRIC CRISIS: Three children with no history of movement disorders developed oculogyric crisis (1 to 20 episodes per day; 2 seconds to several hours duration) after taking increasing doses of lamotrigine. All patients recovered following the reduction of lamotrigine doses (Veerapandiyan et al, 2011).
    a) A 10-year-old girl who was taking lamotrigine 22 mg/kg/day (plasma concentration 6.7 mcg/mL) presented with oculogyric crisis (a few to 20 episodes/day for 3 days; duration of each episode, 2 to 3 hours; associated symptoms: tremulousness, irritability) about a week after her lamotrigine dose was increased to 25 mg/kg/day (plasma concentration 15.8 mcg/mL). MRI revealed diffuse cerebral and cerebellar atrophy. Her symptoms resolved after lamotrigine dose was decreased to 20 mg/kg/day (plasma concentration 8.5 mcg/mL).
    b) A 3-year-old boy who was taking lamotrigine 2 mg/kg/day (plasma concentration 2.1 mcg/mL) developed oculogyric crisis (1 episode/day for 1 month; duration of each episode, few seconds to 1 minute) after his lamotrigine dose was increased to 7.8 mg/kg/day. His symptoms resolved after decreasing the lamotrigine dose.
    c) An 11-year-old boy who was taking lamotrigine 5.5 mg/kg/day for absence seizures, developed recurrent episodes of oculogyric crisis (5 to 7 episodes/day for 3 months; duration of each episode, 2 to 4 seconds) after his lamotrigine dose was increased to 7 mg/kg/day (plasma concentration 9.3 mcg/mL). His symptoms resolved after his lamotrigine dose was decreased to 4 mg/kg/day.
    B) WITH POISONING/EXPOSURE
    1) OCULOGYRIC CRISIS: A 25-year-old man who was taking lamotrigine 400 mg/day (6 mg/kg/day; plasma concentration 12.1 mcg/mL) for generalized tonic-clonic seizures developed oculogyric crisis (2 to 3 episodes of sustained upward deviation of both eyes) after inadvertently ingesting 1600 mg/day (23.5 mg/kg/day; plasma concentration 16.5 mcg/mL) of lamotrigine during a 24-hour period. His symptoms resolved after his lamotrigine dose was decreased to 6 mg/kg/day (plasma concentration 7 mcg/mL) (Veerapandiyan et al, 2011).
    2) NYSTAGMUS: Following a massive overdose, nystagmus may be a major clinical feature (Oh et al, 2006; O'Donnell & Bateman, 2000; Buckley et al, 1993). Nystagmus developed in 8 of 493 (1.6%) patients with lamotrigine overdose (Lofton & Klein-Schwartz, 2004).
    a) CASE REPORT: Two patients developed reversible downbeat nystagmus as a result of lamotrigine toxicity. Both patients had intractable epilepsy and developed oscillopsia and incoordination while taking lamotrigine in combination with other anticonvulsants. The first patient was taking lorazepam 1 mg twice daily, lamotrigine 100 milligrams four times daily, and gabapentin 400 milligrams three times daily. She had large amplitude downbeat nystagmus in the primary gaze, which became more pronounced in both right and left gaze. Downbeat nystagmus in down gaze and minimal nystagmus in up gaze were noted. In addition, she had moderate truncal ataxia. Serum lamotrigine levels peaked at 19.9 mcg/mL (therapeutic range 1-14 mcg/mL). After lamotrigine dose was reduced, her nystagmus resolved without further sequelae (AlKawi et al, 2005).
    1) A 34-year-old woman developed intermittent diplopia and oscillopsia over a period of 2 years. She was taking lamotrigine 200 milligrams every morning, 100 milligrams at noon, 200 milligrams every evening, and valproic acid 250 milligrams five times daily which was switched to oxcarbazepine 300 milligrams three times daily the day before her evaluation. Low amplitude, downbeat oblique nystagmus in primary gaze that increased in lateral gaze was observed. The nystagmus was absent in up gaze and was of low amplitude in down gaze with frequent square wave jerks. Serum lamotrigine levels peaked at 27 mcg/mL (therapeutic range 4-18 mcg/mL using a different laboratory than the case above). The valproate dose was reduced slowly and lamotrigine serum level decreased rapidly; two days later, the serum level normalized. Valproic acid, an inhibitor of hepatic enzymes, can increase lamotrigine half-life from about 25 hours to 59 hours and increase lamotrigine levels (AlKawi et al, 2005).
    b) CASE REPORT: An 18-year-old woman with a history of epilepsy presented agitated, disoriented, noncooperative with a Glasgow Coma Scale (GCS) score of 12 about an hour after ingesting an unknown amount of lamotrigine 100 mg tablets and sertraline 50 mg tablets. Physical examination revealed vertical nystagmus and myoclonia. She responded to questions with dysphasia within an hour of receiving IV lipid emulsion therapy (bolus of 100 mL and 0.5 mL/kg/min infusion for 2 hours; total dose, 3100 mL). She had a GCS score of 15 and mild dysarthria 4 hours later (Eren Cevik et al, 2014).
    3) BLURRED VISION: In a series of 493 patients with lamotrigine overdose, 7 (1.4%) developed blurred vision (Lofton & Klein-Schwartz, 2004). Eleven percent to 25% of patients receiving lamotrigine in clinical trials had blurred vision, compared to 10 to 7% in controls (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007; Matsuo et al, 1993). Blurred vision was more common at higher lamotrigine doses (Matsuo et al, 1993).
    4) DIPLOPIA: Fourteen percent to 27.6% of patients have experienced diplopia in lamotrigine clinical studies (Betts et al, 1991; Jawad et al, 1989; Matsuo et al, 1993; Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007) compared to an incidence of 8% or less in controls (Matsuo et al, 1993; Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).
    3.4.6) THROAT
    A) XEROSTOMIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Dry mouth with thick secretions was reported in a 33-year-old woman following a 1500 to 2000 milligrams lamotrigine overdose (Harchelroad et al, 1994).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) ELECTROCARDIOGRAM ABNORMAL
    1) WITH THERAPEUTIC USE
    a) Premarketing studies have shown a minor incidence of increase in PR interval, not clinically significant, following therapeutic lamotrigine doses (Matsuo et al, 1993).
    b) One study reported one case who had clinically insignificant first-degree heart block following therapeutic doses of lamotrigine (Betts et al, 1991).
    c) BRUGADA-LIKE PATTERN
    1) CASE REPORT: A 22-year-old woman treated with lamotrigine 275 mg twice daily monotherapy for epilepsy presented to the emergency department (ED) after sustaining a closed head injury and losing consciousness. She spontaneously recovered full consciousness within 1 hour of arrival at the ED. Nystagmus, symmetrically diminished deep tendon reflexes in all extremities, and mild ataxia were observed on examination. She reported nausea, vertigo, alterations in consciousness, and falls over the preceding 2 months. Despite a negative family history for sudden cardiac death and no structural heart disease, ECG showed a nonspecific pattern consistent with Brugada syndrome including greater than 2 mm of downsloping, notching, and ST elevation without T-wave inversion in V2, , and a QTc measurement of 475 msec. An initial procainamide challenge was positive for Brugada type I (downsloping ST elevation of 2 mm with T-wave inversion in V2 and V3); however, her serum lamotrigine level at that time was toxic at 20.4 mcg/mL (therapeutic range, 1 to 4 mcg/mL). A repeat procainamide challenge was negative for Brugada and her baseline ECG no longer showed ST elevation when lamotrigine levels decreased to 0.2 mcg/mL. Lamotrigine was discontinued and she was free of symptoms at a 6-month follow up visit (Strimel et al, 2010).
    2) WITH POISONING/EXPOSURE
    a) No ECG abnormalities were reported in an overdose involving greater than 4000 mg lamotrigine and a plasma lamotrigine concentration of 52 mcg/mL (Prod Info Lamictal(R), lamotrigine tablets, 2000).
    b) Conduction disturbances were reported in 2 of 493 patients following lamotrigine overdose (Lofton & Klein-Schwartz, 2004).
    c) CASE REPORT: A 50-year-old woman with a history of type 2 diabetes and bipolar disorder developed coma (Glasgow Coma Scale 7) approximately 2 hours after ingesting up to 3.5 g of lamotrigine. She was hemodynamically stable and a chest X-ray was normal. An ECG revealed sinus tachycardia and arterial blood gas analysis revealed FiO2 = 0.30, pH 7.36, PaCO2 39 mm Hg, PaO2 85 mm Hg, and base deficit -3 mmol/L. At this time, serum lamotrigine concentration was 27.1 mcg/mL (reference, 2 to 16 mcg/mL). She was treated with activated charcoal, IV magnesium and sodium bicarbonate. Four hours postadmission, another ECG revealed a widening of the QRS complex with left bundle branch block. All laboratory results, including troponin-I levels, were normal. Six hours postingestion, serum lamotrigine concentration increased to 29.7 mcg/mL. Despite supportive therapy, including sodium bicarbonate, the ECG did not improve. At this time, she was treated with an intravenous bolus (1.5 mL/kg) of 20% lipid emulsion and all symptoms resolved within a few minutes. She continued to receive lipids (0.5 mL/kg/min) for another 10 hours. She was discharged 2 days postingestion (Castanares-Zapatero et al, 2011).
    B) CARDIAC ARREST
    1) WITH POISONING/EXPOSURE
    a) Severe sodium channel blockade after a massive lamotrigine overdose has been reported (Nogar et al, 2011).
    1) CASE REPORT: A 48-year-old woman, with a medical history of depression and gastric bypass surgery, as well as a remote history of seizure disorder, presented to the ED unconscious with one episode of tonic-clonic seizure after ingesting about 7.5 g of lamotrigine (lamotrigine concentration: 74.7 mcg/mL). Her vital signs included a heart rate of 131 beats/min, blood pressure of 107/68 mmHg, temperature of 99.4 degrees F, and a respiratory rate of 16 breaths/min. She was treated with supportive care and was intubated with rocuronium. An ECG revealed a narrow-complex sinus tachycardia, with a QRS duration of 106 msec. She developed a one-minute tonic-clonic seizure about 2.5 hours after presentation and received a 2 mg bolus of lorazepam. At this time, a second ECG revealed normal sinus rhythm, a heart rate of 86 beats/min, a QRS duration of 110 msec, and a small R-wave in aVR. Despite treatment with 2 boluses of sodium bicarbonate, her QRS duration did not change and she remained hemodynamically unstable. She developed status epilepticus 10 minutes later and an ECG showed a wide-complex tachycardia, and she became pulseless. She received sodium bicarbonate, 360 mL of 20% lipid emulsion, lidocaine, amiodarone, and 2 g calcium chloride during 45 minutes of resuscitation. Her pulses were reestablished with QRS narrowing with each defibrillation, but were widened again with recurrent seizure activity. Following the termination of seizures with vecuronium, she was finally stabilized. An ECG after resuscitation revealed a junctional tachycardia with a 3 mm R-wave in aVR. She was treated with fosphenytoin and propofol in the ICU, and was gradually weaned from sedation over 3 days when it was observed that the patient had extensor posturing. A MRI revealed brain edema consistent with anoxic injury and her family elected to withdraw care 4 days after presentation (Nogar et al, 2011).
    C) WIDE QRS COMPLEX
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. QRS prolongation (114 to 116 ms) developed in 2 patients (Moore et al, 2013).
    b) CASE REPORT: A 19-year-old man with bipolar disorder ingested 4 g of lamotrigine and developed tachycardia (123 beats/min), multiple seizures, charcoal aspiration, respiratory failure, prolongation of the QRS interval (214 ms), and completed heart block. Despite supportive care, he developed sepsis and multiorgan failure and died 10 days postingestion (French et al, 2011).
    c) Widening of the QRS complex (112 msec) was reported in a 26-year-old man following an overdose of 1350 mg lamotrigine. Two months later his QRS width had decreased to less than 100 ms (Buckley et al, 1993).
    d) CASE REPORT: A 50-year-old woman with a history of type 2 diabetes and bipolar disorder developed coma (Glasgow Coma Scale 7) approximately 2 hours after ingesting up to 3.5 g of lamotrigine. She was hemodynamically stable and a chest X-ray was normal. An ECG revealed sinus tachycardia and arterial blood gas analysis revealed FiO2 = 0.30, pH 7.36, PaCO2 39 mm Hg, PaO2 85 mm Hg, and base deficit -3 mmol/L. At this time, serum lamotrigine concentration was 27.1 mcg/mL (reference, 2 to 16 mcg/mL). She was treated with activated charcoal, IV magnesium and sodium bicarbonate. Four hours postadmission, another ECG revealed a widening of the QRS complex with left bundle branch block. All laboratory results, including troponin-I levels, were normal and vital signs remained stable. Six hours postingestion, serum lamotrigine concentration increased to 29.7 mcg/mL. Despite supportive therapy, including sodium bicarbonate, the ECG did not improve. At this time, she was treated with an intravenous bolus (1.5 mL/kg) of 20% lipid emulsion and her QRS complexes narrowed within a few minutes. She continued to receive lipids (0.5 mL/kg/min) for another 10 hours. She was discharged 2 days postingestion (Castanares-Zapatero et al, 2011).
    D) PROLONGED QT INTERVAL
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. QTc prolongation (463 to 586 ms) developed in 4 patients (Moore et al, 2013).
    E) COMPLETE ATRIOVENTRICULAR BLOCK
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 19-year-old man with bipolar disorder ingested 4 g of lamotrigine and developed tachycardia (123 beats/min), multiple seizures, charcoal aspiration, respiratory failure, prolongation of the QRS interval (214 ms), and completed heart block. Despite supportive care, he developed sepsis and multiorgan failure and died 10 days postingestion (French et al, 2011).
    F) HYPOTENSIVE EPISODE
    1) WITH THERAPEUTIC USE
    a) CASE REPORTS: Two children had hypotensive episodes, with blood pressure 77/45 mm Hg in one child, after addition of lamotrigine to their valproic acid regimen. Both children subsequently suffered multiorgan dysfunction, which reversed several days following discontinuation of their seizure medication (Chattergoon et al, 1997). This probably represents lamotrigine-associated anticonvulsant hypersensitivity syndrome (Schlienger et al, 1998).
    G) HYPERTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. Hypertension (BP greater than 140 mmHg) developed in 4 patients (Moore et al, 2013).
    H) TACHYCARDIA
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. Tachycardia (greater than 100 beats/min) developed in 5 patients (Moore et al, 2013).
    b) Tachycardia has been reported following overdoses (Castanares-Zapatero et al, 2011; French et al, 2011; Nwogbe et al, 2009; Lofton & Klein-Schwartz, 2004; Schwartz & Geller, 2007).
    c) Tachycardia was reported in 21 out of 493 patients (4.3%) following lamotrigine overdose (Lofton & Klein-Schwartz, 2004).
    d) CASE REPORT: A 29-year-old man presented with tachycardia (pulse 150 bpm) after an intentional ingestion of an unknown number of lamotrigine 200 mg tablets and ethanol (Schwartz & Geller, 2007).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) SUFFOCATING
    1) WITH POISONING/EXPOSURE
    a) Theoretically, aspiration can occur in a lethargic or comatose patient. Coma has rarely been reported following lamotrigine overdose (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).
    B) DECREASED RESPIRATORY FUNCTION
    1) WITH POISONING/EXPOSURE
    a) Respiratory depression has been reported rarely following lamotrigine overdose (3 of 493 patients in one series) (Lofton & Klein-Schwartz, 2004).
    C) TACHYPNEA
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. Tachypnea (greater than 24 breaths/min) developed in 4 patients (Moore et al, 2013).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) NYSTAGMUS
    1) WITH POISONING/EXPOSURE
    a) Following overdoses, nystagmus may be a prominent neurologic effect (Close & Banks, 2010; Nwogbe et al, 2009; O'Donnell & Bateman, 2000). In a series of 493 lamotrigine overdoses, 8 patients (1.6%) developed nystagmus(Lofton & Klein-Schwartz, 2004).
    b) CASE REPORT: Horizontal and vertical nystagmus was reported in a 26-year-old man one hour following ingestion of 1350 mg lamotrigine. Twenty-four hours later fine nystagmus and mild ataxia were evident (Buckley et al, 1993).
    c) CASE REPORT: Following a stated overdose of 4500 mg lamotrigine (absorbed estimated dose of 2900 mg), a 32-year-old woman presented to the emergency department 4 hours later with marked ataxia and later developed rotational nystagmus in all directions of gaze. Over the next 48 hours the nystagmus improved, and she was discharged on the third day (O'Donnell & Bateman, 2000).
    d) CASE REPORT: Two patients developed reversible downbeat nystagmus as a result of lamotrigine toxicity. Both patients had intractable epilepsy and developed oscillopsia and incoordination while taking lamotrigine in combination with other anticonvulsants. The first patient was taking lorazepam 1 milligram twice daily, lamotrigine 100 milligrams four times daily, and gabapentin 400 milligrams three times daily. She had large amplitude downbeat nystagmus in the primary gaze, which became more pronounced in both right and left gaze. Downbeat nystagmus in down gaze and minimal nystagmus in up gaze were noted. In addition, she had moderate truncal ataxia. Serum lamotrigine levels peaked at 19.9 mcg/mL (therapeutic range 1-14 mcg/mL). After lamotrigine dose was reduced, her nystagmus resolved without further sequelae (AlKawi et al, 2005).
    1) A 34-year-old woman developed intermittent diplopia and oscillopsia over a period of 2 years. She was taking lamotrigine 200 milligrams every morning, 100 milligrams at noon, 200 milligrams every evening, and valproic acid 250 milligrams five times daily which was switched to oxcarbazepine 300 milligrams three times daily the day before her evaluation. Low amplitude, downbeat oblique nystagmus in primary gaze that increased in lateral gaze was observed. The nystagmus was absent in up gaze and was of low amplitude in down gaze with frequent square wave jerks. Serum lamotrigine levels peaked at 27 mcg/mL (therapeutic range 4-18 mcg/mL using a different laboratory than the case above). The valproate dose was reduced slowly and lamotrigine serum level decreased rapidly; two days later, the serum level normalized. Valproic acid, an inhibitor of hepatic enzymes, can increase lamotrigine half-life from about 25 hours to 59 hours and increase lamotrigine levels (AlKawi et al, 2005).
    B) OCULOGYRIC CRISIS
    1) WITH THERAPEUTIC USE
    a) CASE SERIES: Three children with no history of movement disorders developed oculogyric crisis (1 to 20 episodes per day; 2 seconds to several hours duration) after taking increasing doses of lamotrigine. All patients recovered following the reduction of lamotrigine doses (Veerapandiyan et al, 2011).
    1) A 10-year-old girl who was taking lamotrigine 22 mg/kg/day (plasma concentration 6.7 mcg/mL) presented with oculogyric crisis (a few to 20 episodes/day for 3 days; duration of each episode, 2 to 3 hours; associated symptoms: tremulousness, irritability) about a week after her lamotrigine dose was increased to 25 mg/kg/day (plasma concentration 15.8 mcg/mL). MRI revealed diffuse cerebral and cerebellar atrophy. Her symptoms resolved after lamotrigine dose was decreased to 20 mg/kg/day (plasma concentration 8.5 mcg/mL).
    2) A 3-year-old boy who was taking lamotrigine 2 mg/kg/day (plasma concentration 2.1 mcg/mL) developed oculogyric crisis (1 episode/day for 1 month; duration of each episode, few seconds to 1 minute) after his lamotrigine dose was increased to 7.8 mg/kg/day. His symptoms resolved after decreasing the lamotrigine dose.
    3) An 11-year-old boy who was taking lamotrigine 5.5 mg/kg/day for absence seizures, developed recurrent episodes of oculogyric crisis (5 to 7 episodes/day for 3 months; duration of each episode, 2 to 4 seconds) after his lamotrigine dose was increased to 7 mg/kg/day (plasma concentration 9.3 mcg/mL). His symptoms resolved after his lamotrigine dose was decreased to 4 mg/kg/day.
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 25-year-old man who was taking lamotrigine 400 mg/day (6 mg/kg/day; plasma concentration 12.1 mcg/mL) for generalized tonic-clonic seizures developed oculogyric crisis (2 to 3 episodes of sustained upward deviation of both eyes) after inadvertently ingesting 1600 mg/day (23.5 mg/kg/day; plasma concentration 16.5 mcg/mL) of lamotrigine during a 24-hour period. His symptoms resolved after his lamotrigine dose was decreased to 6 mg/kg/day (plasma concentration 7 mcg/mL) (Veerapandiyan et al, 2011).
    C) ASEPTIC MENINGITIS
    1) WITH THERAPEUTIC USE
    a) The US Food and Drug Administration (FDA) has reported 40 cases of aseptic meningitis in adults and pediatric patients taking lamotrigine, with symptoms occurring 1 to 42 days (mean, 16 days) after starting lamotrigine. Hospitalization was required in 35 of 40 patients. In most cases symptoms resolved upon discontinuation of therapy. Rechallenge in 15 cases resulted in recurrence of more severe symptoms within a mean of 5 hr (range, 30 min to 24 hr). In cerebrospinal fluid analysis performed on 25 cases, there was predominance of neutrophils in most cases and a predominance of lymphocytes in one-third of cases. It is not known if the cases of aseptic meningitis are part of a hypersensitivity or a generalized drug reaction (U.S. Food and Drug Administration, 2010).
    D) DISORDER OF ACCOMMODATION
    1) WITH POISONING/EXPOSURE
    a) Absent accommodation developed 24 hours after an overdose of 1500 to 2000 mg lamotrigine (Harchelroad et al, 1994). The pupils were briskly reactive and bilaterally equal in size (4 mm).
    E) ATAXIA
    1) WITH POISONING/EXPOSURE
    a) Marked ataxia is commonly reported following overdoses in children and adults (Close & Banks, 2010; Oh et al, 2006; Nwogbe et al, 2009; Dinnerstein et al, 2007; Daana et al, 2007; O'Donnell & Bateman, 2000; Briassoulis et al, 1998; Stopforth, 1997; Reutens et al, 1993). In a series of 493 patients with lamotrigine overdose, 24 (4.9%) developed ataxia (Lofton & Klein-Schwartz, 2004).
    F) INCREASED MUSCLE TONE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: One hour after ingesting 1350 milligrams lamotrigine, a man presented with hypertonia and brisk reflexes. Twenty-four hours later the reflexes were normal and mild ataxia was present (Buckley et al, 1993).
    b) CASE REPORT: Hypertonia, muscle weakness and ataxia was reported approximately 50 minutes after an ingestion of 800 milligrams in a 2-year-old child. Symptoms resolved within 24 hours and the child was discharged after 48 hours (Briassoulis et al, 1998).
    G) HYPOREFLEXIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Twenty-four hours following an overdose of 1500 to 2000 mg lamotrigine (also taking phenytoin and clorazepate), a woman presented with zero muscle stretch reflexes in her lower extremities. Reflexes returned to normal over the next 24 hours (Harchelroad et al, 1994).
    H) HYPERREFLEXIA
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. Hyperreflexia and intermittent myoclonus developed in 5 patients; inducible clonus developed in 2 patients (Moore et al, 2013).
    b) CASE REPORTS: A 2-year-old boy experienced 2 generalized seizures 60 minutes after ingesting up to 43 mg/kg of lamotrigine. On admission, he had mild nystagmus, drowsiness, ataxia, hyperreflexia, and vomiting. Following supportive care, he recovered completely (Close & Banks, 2010).
    I) DROWSY
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Somnolence and ataxia were reported in a 45-year-old woman following a 2 week upward titration of therapeutic lamotrigine doses. No focal neurologic signs were present. Her neurological status improved over the next 2 weeks following discontinuation of lamotrigine (Schaub et al, 1994).
    2) WITH POISONING/EXPOSURE
    a) Drowsiness and obtundation have been reported following lamotrigine overdose (Close & Banks, 2010; Nwogbe et al, 2009; Dinnerstein et al, 2007; Daana et al, 2007).
    b) In a series of 493 patients with lamotrigine exposure, drowsiness and lethargy were the most common toxic effects, developing in 103 (20.9%) patients. Slurred speech developed in 9 (1.8%) patients in this series (Lofton & Klein-Schwartz, 2004).
    c) CASE REPORT: A woman presented with marked agitation, progressing over 4 hours to increasing lethargy, 4 hours after ingesting 1500 to 2000 milligrams of lamotrigine (other drugs included phenytoin and clorazepate). Dizziness and slurred speech were also reported. All symptoms resolved over 2 days with supportive treatment (Harchelroad et al, 1994).
    d) CASE REPORT: A woman who ingested greater than 4000 milligrams lamotrigine experienced dizziness, headache and somnolence. She recovered without sequelae (Prod Info Lamictal(R), lamotrigine tablets, 2000).
    e) CASE REPORT: A 17-year-old adolescent was reported to experience somnolence, dysarthria, ataxia, and lethargy following an overdose of lamotrigine (2000 milligrams) and gabapentin (12,000 milligrams). She fully recovered following supportive care (Stopforth, 1997).
    f) CASE REPORT: A 3-year-old girl developed significant somnolence, a lacy reticular blanching rash and elevated liver function tests after ingesting forty-six 25 milligram tablets (1.150 grams) of lamotrigine. Plasma lamotrigine level was 25.3 mcg/mL (elevated above adult therapeutic levels). Following supportive treatment, she recovered without further sequelae (Zidd & Hack, 2004).
    J) COMA
    1) WITH POISONING/EXPOSURE
    a) Coma has been reported rarely following lamotrigine overdose. In a series of 493 patients with lamotrigine exposure, 6 patients (1.2%) became comatose (Lofton & Klein-Schwartz, 2004).
    b) CASE REPORT: A 46-year-old man with a history of bipolar disorder developed bilateral synchronous generalized myoclonus several hours after ingesting about 6000 mg of lamotrigine in a suicide attempt. Before arrival to the ED, he experienced 3 generalized seizures and presented comatose (Glasgow-Coma score (GCS) of 5/15) with spasticity in all 4 limbs. Laboratory results revealed an elevated lamotrigine concentration of 100 mcmol/L (therapeutic, 1 to 4 mcmol/L). EEG monitoring revealed no epileptiform discharges anytime during the myoclonic jerks. Following supportive care, including treatment with IV lorazepam, phenytoin, phenobarbital, midazolam, propofol, thiopental and mechanical ventilation, his symptoms gradually resolved. On day 3, another EEG was completely normal (Algahtani et al, 2014).
    c) CASE REPORT: A 29 year-old man developed coma necessitating intubation after an intentional ingestion of an unknown amount of lamotrigine 200 milligram tablets and ethanol (190 mg/dL; normal < 10 mg/dL). The patient was extubated on the second hospital day, and made a full recovery with supportive care (Schwartz & Geller, 2007).
    d) CASE REPORT: Coma, lasting 8 to 12 hours, was reported in a woman who ingested more than 4000 milligrams lamotrigine. She recovered without sequelae (Prod Info Lamictal(R), lamotrigine tablets, 2000).
    e) CASE REPORT: A 50-year-old woman with a history of type 2 diabetes and bipolar disorder developed coma (Glasgow Coma Scale 7) approximately 2 hours after ingesting up to 3.5 g of lamotrigine. She recovered with supportive care and was discharged 2 days postingestion (Castanares-Zapatero et al, 2011).
    K) TOXIC ENCEPHALOPATHY
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Reversible encephalopathy associated with high lamotrigine blood levels (19 mg/L), with a concurrent urinary tract infection was reported in a 47-year-old woman. Concomitant medication included valproic acid, which remained at therapeutic blood levels. Symptoms included stupor, mutism, profound hemiplegia, urinary incontinence and primitive reflexes. Symptoms improved concurrent with a fall in lamotrigine levels after her lamotrigine dose was decreased from 400 milligrams/day to 75 milligrams/day (Hennessy & Wiles, 1996).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: Encephalopathy with stupor (Glasgow coma scale 6) was reported in a 55-year-old woman with elevated serum lamotrigine concentration (32 milligrams/liter) after an intentional overdose of an unknown quantity of lamotrigine. Improvement of encephalopathy correlated with declining lamotrigine serum levels. Concurrent use of valproic acid may have contributed to the toxic effects of lamotrigine (Sbei & Campellone, 2001).
    L) SEIZURE
    1) WITH THERAPEUTIC USE
    a) Seizures have been reported following high-dosage in children (Close & Banks, 2010; Guerrini et al, 1999).
    b) CASE REPORT: Myoclonic status epilepticus was reported in an 8-year-old girl with Lennox-Gastaut syndrome when her 15 milligrams/kilogram/day dose of lamotrigine was increased to 20 milligrams/kilogram. An EEG revealed continuous myoclonus of cortical origin. Following discontinuation of lamotrigine, her seizures disappeared (Guerrini et al, 1999).
    2) WITH POISONING/EXPOSURE
    a) PEDIATRIC
    1) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. Seizures developed in 3 patients (Moore et al, 2013).
    a) A 2-year-old boy (weight: 13.5 kg) who was taking lamotrigine 25 mg for a seizure disorder, presented with seizures with myoclonic jerking and agitation after ingesting about 21 lamotrigine tablets. He recovered following supportive care and was discharged 72 hours later (Moore et al, 2013).
    2) CASE REPORT: A 12-day-old infant experienced two episodes of tonic-clonic seizures 9 days apart, with no etiology determined despite extensive evaluation. Following control of the seizures, he developed stereotypical movements consisting of repeated non-purposeful thrashing movements of his arms, inability to fix or follow, irritability, loss of all social interaction and inability to feed. Urine toxicology results taken after the first seizure episode revealed elevated levels of benzodiazepines (lorazepam was administered for the patient's seizures) and lamotrigine. All previously stored serum samples were analyzed. Lamotrigine levels above 30 milligrams/liter were found, which suggested repeat poisoning. Following symptomatic treatment, he gradually returned to normal over the next 5 days and was placed in protective custody (Willis et al, 2007).
    3) CASE REPORT: Generalized tonic-clonic seizures were reported in a 2-year-old boy shortly after ingestion of sixteen 50 milligram tablets. Seizures spontaneously resolved after 15 minutes while the patient was in-route to a hospital. Shortly after arrival to the hospital he experienced another generalized seizure lasting 5 minutes and responding to 2 milligrams IV midazolam. No further seizure activity was noted, and the patient was discharged in 48 hours (Briassoulis et al, 1998).
    4) CASE REPORTS: A 2-year-old boy experienced 2 generalized seizures 60 minutes after ingesting up to 43 mg/kg of lamotrigine. On admission, he had mild nystagmus, drowsiness, ataxia, hyperreflexia, and vomiting. Following supportive care, he recovered completely (Close & Banks, 2010).
    b) ADULT
    1) Seizures have been reported following lamotrigine overdose. In a series of 493 patients with lamotrigine exposure seizures developed in 8 patients (1.6%). Six of these patients were not taking lamotrigine chronically, thus the seizures were likely due to the lamotrigine rather than an underlying seizure disorder (Lofton & Klein-Schwartz, 2004; Briassoulis et al, 1998).
    2) CASE REPORT: A 46-year-old man with a history of bipolar disorder developed bilateral synchronous generalized myoclonus several hours after ingesting about 6000 mg of lamotrigine in a suicide attempt. Before arrival to the ED, he experienced 3 generalized seizures and presented comatose (Glasgow-Coma score (GCS) of 5/15) with spasticity in all 4 limbs. Laboratory results revealed an elevated lamotrigine concentration of 100 mcmol/L (therapeutic, 1 to 4 mcmol/L). EEG monitoring revealed no epileptiform discharges anytime during the myoclonic jerks. Following supportive care, including treatment with IV lorazepam, phenytoin, phenobarbital, midazolam, propofol, thiopental and mechanical ventilation, his symptoms gradually resolved. On day 3, another EEG was completely normal (Algahtani et al, 2014).
    3) CASE REPORT: A 19-year-old man with bipolar disorder ingested 4 g of lamotrigine and developed tachycardia, multiple seizures, charcoal aspiration, respiratory failure, prolongation of the QRS interval (214 ms), and completed heart block. Despite supportive care, he developed sepsis and multiorgan failure and died 10 days postingestion (French et al, 2011).
    4) CASE REPORT: A 29-year-old man developed several generalized tonic-clonic seizures after an intentional ingestion of an unknown number of 200 milligram lamotrigine tablets along with ethanol. The seizures resolved with lorazepam. The patient required intubation for 2 days, but a full recovery was made with supportive care (Schwartz & Geller, 2007).
    5) STATUS EPILEPTICUS: Approximately 1 to 2 hours after ingesting 4.1 grams of lamotrigine, a 42-year-old woman with a history of left sided tonic seizures with preservation of consciousness experienced a series of secondarily generalized tonic-clonic seizures over 3 hours without full recovery of consciousness between seizures. Her seizures were controlled with benzodiazepines; however, she developed transient obtundation and severe ataxia. These symptoms resolved completely within 96 hours of supportive therapy. The lamotrigine concentrations were: 47.4 mcg/mL (on admission), 15.8 mcg/mL (on day 2), 3.2 mcg/mL (on day 3), and 0.8 mcg/mL (on day 4) (Dinnerstein et al, 2007).
    6) CASE REPORT: A 5-year-old epileptic girl developed ataxia, drowsiness, confusion, followed by vomiting and seizure exacerbation after ingesting 500 milligrams (25 mg/kg/day) of lamotrigine in two 250 mg doses 12 hours apart instead of the prescribed 150 mg twice daily (15 mg/kg/day). Following the discontinuation of lamotrigine and supportive therapy, she gradually improved and was discharged 30 hours after admission (Daana et al, 2007).
    7) CASE REPORT: A 48-year-old woman, with a medical history of depression and gastric bypass surgery, as well as a remote history of seizure disorder, presented to the ED unconscious with one episode of tonic-clonic seizure after ingesting about 7.5 g of lamotrigine (lamotrigine concentration: 74.7 mcg/mL). Her vital signs included a heart rate of 131 beats/min, blood pressure of 107/68 mmHg, temperature of 99.4 degrees F, and a respiratory rate of 16 breaths/min. She was treated with supportive care and was intubated with rocuronium. An ECG revealed a narrow-complex sinus tachycardia, with a QRS duration of 106 msec. She developed a one-minute tonic-clonic seizure about 2.5 hours after presentation and received a 2 mg bolus of lorazepam. At this time, a second ECG revealed normal sinus rhythm, a heart rate of 86 beats/min, a QRS duration of 110 msec, and a small R-wave in aVR. Despite treatment with 2 boluses of sodium bicarbonate, her QRS duration did not change and she remained hemodynamically unstable. She developed status epilepticus 10 minutes later and an ECG showed a wide-complex tachycardia, and she became pulseless. She received sodium bicarbonate, 360 mL of 20% lipid emulsion, lidocaine, amiodarone, and 2 g calcium chloride during 45 minutes of resuscitation. Her pulses were reestablished with QRS narrowing with each defibrillation, but were widened again with recurrent seizure activity. Following the termination of seizures with vecuronium, she was finally stabilized. An ECG after resuscitation revealed a junctional tachycardia with a 3 mm R-wave in aVR. She was treated with fosphenytoin and propofol in the ICU, and was gradually weaned from sedation over 3 days when it was observed that the patient had extensor posturing. A MRI revealed brain edema consistent with anoxic injury and her family elected to withdraw care 4 days after presentation (Nogar et al, 2011).
    M) TREMOR
    1) WITH THERAPEUTIC USE
    a) Disabling tremors with dysarthria and mild truncal ataxia have been reported in 3 patients following therapeutic doses of lamotrigine in combination with valproate sodium. Tremor resolved with reduction in dose of lamotrigine or valproate sodium (Reutens et al, 1993).
    2) WITH POISONING/EXPOSURE
    a) Tremor developed in 9 of 493 (1.8%) patients with lamotrigine overdose (Lofton & Klein-Schwartz, 2004).
    b) Tremor of the limbs was noted in a 2-year-old boy following an ingestion of sixteen 50 milligram tablets (800 milligrams) (Briassoulis et al, 1998).
    N) DYSKINESIA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Blepharospasm was reported in an adult after 4 months of therapy with lamotrigine, which was titrated up to 500 milligrams/day. The patient presented with persistent, involuntary eye blinking. Associated contractions of the platysma muscles in the cervical regions bilaterally were also noted. No other movement disorders were seen. About 4 weeks after cessation of lamotrigine, the blepharospasm resolved (Verma et al, 1999).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: Severe choreatic dyskinesia was reported in a 23-year-old man within 2 hours following an intentional ingestion of an unknown quantity of lamotrigine. Agitation and rapid, intermittent, choreatic movements of the trunk and extremities were observed, with no seizure activity. The abnormal movements continued for the next 2 hours, but subsided during the first 12 hours of admission after administration of 2 mg lorazepam IV (Miller et al, 2008) (Miller & Levsky, 2008; Miller et al, 2001).
    O) PSYCHOMOTOR AGITATION
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. Agitation developed in 5 patients (Moore et al, 2013).
    1) A 1-year-old girl (weight: 8.9 kg) developed agitation and intermittent myoclonus of her extremities after ingesting an unknown amount of lamotrigine. On presentation, she had agitation, diaphoresis, and inducible clonus, meeting the diagnostic criteria for serotonin syndrome. Following supportive care, she recovered and was discharged after 19 hours (Moore et al, 2013).
    2) A 2-year-old boy (weight: 13.5 kg) who was taking lamotrigine 25 mg for a seizure disorder, presented with seizures with myoclonic jerking and agitation after ingesting about 21 lamotrigine tablets. He recovered following supportive care and was discharged 72 hours later (Moore et al, 2013).
    3) A 19-month-old boy (weight: 11.4 kg) developed vomiting, confusion, slurred speech, and agitation after ingesting 400 mg of lamotrigine. Following supportive care, he recovered and was discharged home 24 hours after ingestion (Moore et al, 2013).
    b) CASE REPORT: An 18-year-old woman with a history of epilepsy presented with agitation, disorientation and noncooperative with a Glasgow Coma Scale (GCS) score of 12 about an hour after ingesting an unknown amount of lamotrigine 100 mg tablets and sertraline 50 mg tablets. Physical examination revealed vertical nystagmus and myoclonia. She responded to questions with dysphasia within an hour of receiving IV lipid emulsion therapy (bolus of 100 mL and 0.5 mL/kg/min infusion for 2 hours; total dose, 3100 mL). She had a GCS score of 15 and mild dysarthria 6 hours later (Eren Cevik et al, 2014).
    P) CENTRAL NERVOUS SYSTEM FINDING
    1) WITH THERAPEUTIC USE
    a) Dizziness, headache, ataxia, and weakness have been reported with therapeutic doses (Betts et al, 1991; Jawad et al, 1989; Matsuo et al, 1993).
    b) CASE REPORT: A 22-year-old woman treated with lamotrigine 275 mg twice daily monotherapy for epilepsy presented to the emergency department (ED) after sustaining a closed head injury and losing consciousness. She spontaneously recovered full consciousness within 1 hour of arrival at the ED. Nystagmus, symmetrically diminished deep tendon reflexes in all extremities, and mild ataxia were observed on examination. She reported increasing nausea, vertigo, and alterations in consciousness with falls for the preceding 2 months. Lamotrigine concentration was 20.4 mcg/mL (therapeutic 1 to 4 mcg/mL). Lamotrigine was discontinued and she was free of symptoms at a 6-month follow-up visit (Strimel et al, 2010).
    2) WITH POISONING/EXPOSURE
    a) Ataxia, drowsiness, confusion, and obtundation have been reported following lamotrigine overdose (Dinnerstein et al, 2007; Daana et al, 2007).
    b) CASE REPORT: An 18-year-old woman with a history of epilepsy presented with agitation, disorientation and noncooperative with a Glasgow Coma Scale (GCS) score of 12 about an hour after ingesting an unknown amount of lamotrigine 100 mg tablets and sertraline 50 mg tablets. Physical examination revealed vertical nystagmus and myoclonia. She responded to questions with dysphasia within an hour of receiving IV lipid emulsion therapy (bolus of 100 mL and 0.5 mL/kg/min infusion for 2 hours; total dose, 3100 mL). She had a GCS score of 15 and mild dysarthria 6 hours later (Eren Cevik et al, 2014).
    c) CASE SERIES: In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. The following CNS adverse effects were reported: seizures (n=3), depressed mental status (n=4), agitation (n=5), both agitation and depressed mental status (n=3), hyperreflexia and intermittent myoclonus (n=5), and inducible clonus (n=2). The average serum lamotrigine concentration from 7 patients was 35.4 mg/L (17 to 90 mg/L; therapeutic range: 3 to 14 mg/L) (Moore et al, 2013).
    d) CASE REPORT: A 12-day-old infant experienced two episodes of tonic-clonic seizures 9 days apart, with no etiology determined despite extensive evaluation. Following control of the seizures, he developed stereotypical movements consisting of repeated non-purposeful thrashing movements of his arms, inability to fix or follow, irritability, loss of all social interaction and inability to feed. Urine toxicology results taken after the first seizure episode revealed elevated levels of benzodiazepines (lorazepam was administered for the patient's seizures) and lamotrigine. All previously stored serum samples were analyzed. Lamotrigine levels above 30 milligrams/liter were found, which suggested repeat poisoning. Following symptomatic treatment, he gradually returned to normal over the next 5 days and was placed in protective custody (Willis et al, 2007).
    e) In a series of 493 patients with lamotrigine overdose, dizziness or vertigo developed in 22 (4.5%), confusion in 11 (2.2%), and agitation in 10 (2.0%) (Lofton & Klein-Schwartz, 2004). Headache has also been reported after overdose(Harchelroad et al, 1994; Prod Info Lamictal(R), lamotrigine tablets, 2000).
    Q) SEROTONIN SYNDROME
    1) WITH POISONING/EXPOSURE
    a) COMBINATION OVERDOSE: Features consistent with serotonin syndrome developed in a 44-year-old woman who ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g, and was found unconscious at home. After admission, a tonic-clonic seizure, rigidity, hyperreflexia, and reflex myocloni were observed. Laboratory results revealed sodium level of 129 mmol/L, creatinine kinase of 1915 Units/L, and creatine kinase-MB of 31 Units/L. Despite supportive therapy, including 8 hours of hemodialysis, her rigidity and hyperreflexia did not improve. Following treatment with an intravenous bolus (150 mL, 2.5 mL/kg) of 20% lipid emulsion, all symptoms resolved completely. On day 6, she was transferred to a psychiatric ward (Dagtekin et al, 2011).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) NAUSEA AND VOMITING
    1) WITH THERAPEUTIC USE
    a) Nausea, vomiting, anorexia and abdominal pain have been infrequently reported (Close & Banks, 2010; Oh et al, 2006; Jawad et al, 1989; Betts et al, 1991; Matsuo et al, 1993) but is likely to occur with elevated serum levels (Besag et al, 1998).
    2) WITH POISONING/EXPOSURE
    a) Nausea, vomiting, anorexia and abdominal pain are likely to occur with overdoses or elevated serum levels. In a series of 493 patients with lamotrigine overdose, 54 (11%) developed vomiting and 25 (5.1%) complained of nausea (Lofton & Klein-Schwartz, 2004; Besag et al, 1998).
    b) CASE REPORT: A 19-month-old boy (weight: 11.4 kg) developed vomiting, confusion, slurred speech, and agitation after ingesting 400 mg of lamotrigine. Following supportive care, he recovered and was discharged home 24 hours after ingestion (Moore et al, 2013).
    B) ACUTE PANCREATITIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 24-year-old woman presented with tachycardia, lethargy, dehydration, slurred speech, with cerebellar signs of nystagmus and ataxia 5 hours after ingesting 4200 mg of lamotrigine and two bottles of wine (blood ethanol 100 mg/dL). She developed upper abdominal pain and vomiting within 24 hours of admission with a serum amylase of 1043 Units/L (normal 0 to 110 Units/L). An abdominal ultrasound 2 days later revealed a thickened gall bladder wall, with some surrounding fluid and no evidence of gallstones. Following supportive care, she recovered completely and was discharged on day 5 (Nwogbe et al, 2009).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) LIVER ENZYMES ABNORMAL
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 21-year-old man developed significant elevations in liver enzymes (aspartate aminotransferase (AST) and alanine aminotransferase (ALT) serum levels of 960 units/L and 347 units/L, respectively) after taking lamotrigine 200 mg/day for 2 months for schizophrenia. At the time of admission to the inpatient psychiatry unit for paranoid delusions, home medications included aripiprazole 15 mg/day and lamotrigine 200 mg/day; however, nonadherence to aripiprazole was reported by the patient's family. Liver enzymes 2 months prior to admission were normal (ALT and AST, 19 units/L). As serology and autoimmune tests ruled out other etiologies of hepatic injury, lamotrigine was determined to be causative factor and was not restarted at admission. Instead, the patient was restarted on aripiprazole, titrating up to 25 mg/day. Five days after stopping lamotrigine, AST was normal but ALT remained slightly elevated (102 units/L). The Naranjo Probability Scale indicated a probable causative relationship of lamotrigine and acute hepatotoxicity in this patient (Moeller et al, 2008).
    b) CASE REPORT: Elevated liver enzymes (peaked at AST, 6079 International Units/L; ALT, 6900 IU/L; total bilirubin, 3.9 mg/dL; alkaline phosphatase, 149 IU/L; INR, 1.9) were reported in a 43-year-old woman after the addition of lamotrigine (50 mg/day) to oxcarbazepine 2 weeks before presentation. She presented with nausea, mild jaundice and a generalized erythematous pruritic macular rash. Following supportive care, she recovered completely and was discharged on day 3 (Su-Yin et al, 2008).
    c) CASE REPORT: Elevated lactate dehydrogenase (6830 International Units/L) and aspartate aminotransferase (1200 International Units/L) were reported in a case of multiorgan dysfunction possibly due to therapeutic doses of lamotrigine (Schaub et al, 1994). Generalized seizures did not occur, but rhabdomyolysis was present.
    d) CASE REPORT: Elevated AST (1,066 Units/L), ALT (279 Units/L), GGT (291 Units/L), and ALP (145 Units/L) serum levels were reported in an 11-year-old girl after the addition of lamotrigine to her valproic acid therapy. Multiorgan dysfunction developed, with rhabdomyolysis and no seizures. After discontinuation of her seizure medications, all abnormal serum levels returned to normal over 10 days (Chattergoon et al, 1997). This probably represents lamotrigine-associated anticonvulsant hypersensitivity syndrome.
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 3-year-old woman developed significant somnolence, a lacy reticular blanching rash and elevated liver function tests (serum AST 38 Units/L [normal range 10 to 31 Units/L]; ALP 332 Units/L [normal range 99 to 326 Units/L]) after ingesting forty-six 25 milligram tablets (1.150 grams) of lamotrigine. Plasma lamotrigine level was 25.3 mcg/mL (elevated above adult therapeutic levels). Following supportive treatment, she recovered without further sequelae (Zidd & Hack, 2004).
    B) HYPERBILIRUBINEMIA
    1) WITH THERAPEUTIC USE
    a) Slight elevations in plasma bilirubin have been reported with therapeutic lamotrigine doses (Cohen et al, 1987). The clinical significance of this is unknown.

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) ACUTE RENAL FAILURE SYNDROME
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Acute renal failure, in the absence of predisposing factors, occurred in a 45-year-old woman after 14 days of increasing lamotrigine doses as an add-on therapy for complex partial seizures. Carbamazepine and clonazepam had been used previously by this patient. Serum creatinine peaked at 500 micromoles per liter. Rhabdomyolysis developed and may have contributed to the renal failure. Generalized seizures were not reported (Schaub et al, 1994).
    B) BLOOD IN URINE
    1) WITH THERAPEUTIC USE
    a) Hematuria was reported in 5% of patients receiving therapeutic doses of lamotrigine in clinical trials (Jawad et al, 1989).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) LEUKOPENIA
    1) WITH THERAPEUTIC USE
    a) Although uncommon, leukopenia has resulted from therapeutic dosages of lamotrigine.
    b) CASE REPORT: A 35-year-old woman presented with leukopenia, which progressed to sepsis following 10 days of therapy with lamotrigine. Previously she had been stabilized on valproate sodium and propranolol. On admission to the hospital she was hypoxic, hypotensive and feverish, with a total white cell count of 0.6 X 10(9)/L. Following therapy, her condition stabilized and she fully recovered (Nicholson et al, 1995).
    c) CASE REPORT: Agranulocytosis was reported in an 11-year-old girl 2 weeks after starting lamotrigine therapy (50 milligrams/day). No concomitant medications were taken. Lamotrigine was discontinued, and one week later her leucocyte count and proportion of neutrophils improved (de Camargo & Bode, 1999).
    B) DISSEMINATED INTRAVASCULAR COAGULATION
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Disseminated intravascular coagulation has been reported in a 45-year-old woman after 2 weeks of upward titration of therapeutic lamotrigine doses. She had previously been maintained on carbamazepine and clonazepam for seizures with poor control prior to lamotrigine being added to her therapy.
    1) Prothrombin and partial thromboplastin times were significantly prolonged, fibrinogen was decreased, and fibrin degradation products were extremely high (Schaub et al, 1994).
    b) CASE REPORTS: Two children developed multiorgan dysfunction with disseminated intravascular coagulation (DIC), with no concurrent seizures, within 2 weeks after adding lamotrigine to their valproic acid treatment regimen. Platelet and fibrinogen levels were markedly decreased in both patients. Both patients recovered with no sequelae several days after discontinuation of their seizure medications (Chattergoon et al, 1997). This probably represents lamotrigine-associated anticonvulsant hypersensitivity syndrome (Schlienger et al, 1998).
    C) ANEMIA
    1) WITH THERAPEUTIC USE
    a) Anemia has been reported as an uncommon adverse effect of lamotrigine. Anemias appear to be reversible after discontinuation of lamotrigine. Patients with anemias were also taking other anticonvulsants, which may have contributed to the anemia (Esfahani & Dasheiff, 1997).
    b) CASE REPORT: Lamotrigine-induced erythroblastopenic crisis was reported approximately 8 weeks after initiating lamotrigine therapy for a seizure condition in a 29-year-old woman with Diamond-Blackfan anemia, diagnosed at 4 months of age. Treatment with folinic acid resolved the erythroblastopenia while lamotrigine therapy was continued (Pulik et al, 2000).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) ERUPTION
    1) WITH THERAPEUTIC USE
    a) Maculopapular and/or erythematous rashes have been reported with therapeutic doses of lamotrigine (Su-Yin et al, 2008; Betts et al, 1991; Jawad et al, 1989; Matsuo et al, 1993; Messenheimer et al, 1998). Rashes occurred in about 10% of lamotrigine patients as compared to 5% of patients receiving a placebo (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007). The incidence of rash may be associated with increasing the lamotrigine dose and the use of combined drug therapy.
    2) WITH POISONING/EXPOSURE
    a) Rash was reported in 9 out of 493 patients (1.8%) following lamotrigine overdose (Lofton & Klein-Schwartz, 2004).
    b) CASE REPORT: A 3-year-old girl developed significant somnolence, a lacy reticular blanching rash and elevated liver function tests after ingesting forty-six 25 mg tablets (1.150 grams) of lamotrigine. Plasma lamotrigine level was 25.3 mcg/mL (elevated above adult therapeutic levels). Following supportive treatment, she recovered without further sequelae (Zidd & Hack, 2004).
    B) STEVENS-JOHNSON SYNDROME
    1) WITH THERAPEUTIC USE
    a) Serious rashes which have rarely been reported in association with lamotrigine treatment include Stevens-Johnson-Syndrome, angioedema, toxic epidermal necrolysis (TEN), and a rash complicated by hepatic, hematologic and other adverse effects(Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007; Messenheimer et al, 1998).
    b) INCIDENCE: Approximately 0.8% (16 out of 1,983) pediatric patients developed a potentially life-threatening rash in the form of Stevens-Johnson Syndrome or TEN (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007). Coadministration with valproate increases the incidence of serious rash to about 1.2% (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).
    c) Short-term therapy with lamotrigine was associated with Stevens-Johnson Syndrome (SJS) and toxic epidermal necrolysis (TEN) in a case-control study. Three cases were reported with either SJS or TEN following a range of therapy of 2 to 8 weeks. The risk is largely confined to the start of lamotrigine therapy (Rzany et al, 1999).
    d) CASE REPORT: One study reported a case of Stevens-Johnson Syndrome associated with lamotrigine therapy in a 30-year-old man (Sachs et al, 1996). The syndrome developed 5 weeks after initiation of lamotrigine which was added to valproic acid therapy (2500 milligrams/day) and was diagnosed after a positive lymphocyte transformation reaction to the drug. The patient developed a skin eruption and had complaints of influenza-like symptoms.
    C) LYELL'S TOXIC EPIDERMAL NECROLYSIS, SUBEPIDERMAL TYPE
    1) WITH THERAPEUTIC USE
    a) CASE REPORTS: One study reported 3 cases of toxic epidermal necrolysis (TEN), verified by skin biopsies, which developed within 14 days of initiating lamotrigine therapy (Wadelius et al, 1996). All were treated in burn units of hospitals. The authors speculated immune sensitization occurred; however, all patients were also receiving valproic acid. The incidence of rash with lamotrigine is especially high when combined with valproic acid, but it is unknown if the risk of developing TEN is higher (Wadelius et al, 1996).
    b) CASE REPORT: One study reported a case of TEN (confirmed by skin biopsy) in a 22-month-old man child after the addition of lamotrigine to his valproic acid therapy regimen (Vukelic et al, 1997). The seizure-free child developed a high fever and blistering rash which involved skin, conjunctivae, oral cavity and trachea. The authors suggested an interaction between valproic acid and lamotrigine may have resulted in toxic levels of lamotrigine.
    c) CASE REPORT: One study reported a case of suspected TEN in a 74-year-old man possibly secondary to lamotrigine (Chaffin & Davis, 1997). Fourteen days after beginning lamotrigine therapy, the patient developed a rash, which progressed to TEN 4 days later. Other drug therapy included carbamazepine and clindamycin. Following treatment with hydrotherapy and discontinuation of lamotrigine and clindamycin, his symptoms improved and he was discharged 26 days after the rash developed.
    d) CASE REPORT: A fatal case of TEN with multiple organ failure in a 54-year-old man, probably due to lamotrigine and possibly enhanced by valproic acid, is reported. Other contributing factors include concomitant allopurinol and captopril therapy for 4 years previously and initiation of valproic acid 3 months prior to the appearance of TEN. The patient developed TEN 4 weeks after starting lamotrigine therapy (Page et al, 1998).
    e) Administration of lamotrigine in three patients with bipolar disorder resulted in the development of generalized maculopapular rashes that quickly progressed to TEN despite administration of corticosteroids. Two of the three patients developed respiratory distress requiring mechanical ventilation. All three patients gradually recovered with supportive care (Varghese et al, 2006).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) RHABDOMYOLYSIS
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A case of myopathy with elevated creatine kinase levels (7770 International Units/L) and myoglobin levels (3600 micrograms/liter) has been reported in the absence of generalized seizures following a 2 week period of lamotrigine initialization and increasing therapeutic doses in a 45-year-old woman (Schaub et al, 1994).
    b) CASE REPORT: Rhabdomyolysis in the absence of seizures is reported in an 11-year-old woman 9 days after the addition of lamotrigine to her valproic regimen (valproic acid dose was halved). Serum creatine kinase level was reported to be 40,952 Units/L (normal <255 Units/L). Ten days after discontinuing both drugs, her creatine kinase levels returned to normal (Chattergoon et al, 1997). This probably represents lamotrigine-associated anticonvulsant hypersensitivity syndrome.
    2) WITH POISONING/EXPOSURE
    a) Rhabdomyolysis may develop in patients who develop prolonged seizures or coma after lamotrigine overdose (Schwartz & Geller, 2007).
    b) COMBINATION OVERDOSE: A 44-year-old woman ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g, and was found unconscious at home. After admission, a tonic-clonic seizure, rigidity, hyperreflexia, and reflex myocloni were observed. Laboratory results revealed sodium level of 129 mmol/L, creatinine kinase of 1915 Units/L, and creatine kinase-MB of 31 Units/L. Despite supportive therapy, including 8 hours of hemodialysis, her rigidity and hyperreflexia did not improve. Following treatment with an intravenous bolus (150 mL, 2.5 mL/kg) of 20% lipid emulsion, all symptoms resolved completely. On day 6, she was transferred to a psychiatric ward (Dagtekin et al, 2011).

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) CELL-MEDIATED IMMUNE REACTION
    1) WITH THERAPEUTIC USE
    a) ANTICONVULSANT HYPERSENSITIVITY SYNDROME (AHS): consisting of fever, skin eruption or lymphadenopathy, and internal organ involvement has been associated with lamotrigine therapy in 26 reported cases. Effects appear similar to AHS induced by other aromatic anticonvulsants. Patients were reported to have fever (100%), exanthematous rashes (77%), eosinophilia (19%) and lymphadenopathy (12%). Four patients (15%) were reported with disseminated intravascular coagulopathy. The most commonly reported internal organ toxicities were hematologic and liver abnormalities in 69% and 65%, respectively, followed by renal (23%) and musculoskeletal (8%). Concomitant anticonvulsant drugs were used in all 26 cases (Schlienger et al, 1998).
    b) CASE REPORTS: Two children had hypotensive episodes, with blood pressure 77/45 mmHg in one child, after addition of lamotrigine to their valproic acid regimen. Both children subsequently suffered multiorgan dysfunction (hepatotoxicity, rhabdomyolysis, disseminated intravascular coagulation) which reversed several days following discontinuation of their seizure medication (Chattergoon et al, 1997).
    c) CASE REPORT: A 27-year-old woman developed multisystem hypersensitivity reaction, with DIC, fever, rash, and hepatic dysfunction, 11 days after starting lamotrigine therapy. Adjunctive therapy included phenobarbital. After stopping lamotrigine therapy, her condition improved spontaneously with no interventions other than steroid therapy (Sarris & Wong, 1999).
    d) PSEUDOLYMPHOMA
    1) CASE REPORT: A 5-year-old girl who had a documented drug allergy to both phenytoin and carbamazepine, presented with lymphadenopathy, and a 2-week history of fever, chills, rhinorrhea, and non-productive cough after receiving lamotrigine 10 milligrams twice daily for one month. In addition, she experienced vomiting, decreased oral intake and urinary output, and increased seizure activity. During physical examination, mild throat erythema with moderate bilateral lymphadenopathy, and sand-paper-like skin rash on the back were observed. Although she was treated with antibiotics, her condition deteriorated with continued fever, decreased oral intake, increased somnolence, and enlarged tonsils with whitish membranes. CT scan of the head and neck showed a necrotic left enlarged tonsil with matted lymphadenopathy. The needle aspiration of the lymph nodes, obtained during a tonsillectomy and adenoidectomy, was negative for malignancy. The histopathology of the necrotic tonsils showed eosinophilic granulomas with no evidence of lymphoma, consistent with a hypersensitivity reaction possibly secondary to lamotrigine. Following the discontinuation of lamotrigine, her condition improved within 3 days (Marraffa & Guharoy, 2002).
    2) CASE REPORT: A 35-year-old man developed pseudolymphoma (which may develop as a hypersensitivity reaction to some drugs) after 14 weeks of lamotrigine therapy. The patient was receiving lamotrigine 225 milligrams along with valproic acid, carbamazepine, and clobazam when he developed tender cervical lymphadenopathy. The frozen section diagnosis was consistent with lymphoma. With further testing a pathologic diagnosis (leukocyte phenotyping and southern blot hybridization) of lymphoid hyperplasia was established. Lymphadenopathy resolved 1 month after lamotrigine was discontinued (Pathak & McLachlan, 1998).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT: An inadvertent overdose of lamotrigine (5 days of receiving four daily doses of 2700 milligrams) resulted in anticonvulsant hypersensitivity syndrome in a 49-year-old man. Effects included fever, maculopapular eruption, periorbital edema, leukocytosis, hepatitis and acute renal failure. Following drug discontinuation and administration of corticosteroids, symptoms and laboratory tests improved (Mylonakis et al, 1999).

Reproductive

    3.20.1) SUMMARY
    A) Lamotrigine is classified as FDA pregnancy category C. Preliminary data have shown an association with cleft palate and/or cleft lip in infants exposed to lamotrigine during the first trimester of pregnancy. There is insufficient information concerning teratogenic effects of lamotrigine in the literature to recommend its use during pregnancy. Lamotrigine has been shown to be excreted in breast milk at low concentrations. Monitor breastfed infants closely for signs or symptoms of lamotrigine toxicity, including apnea, drowsiness, or poor sucking. Because the effects on nursing infants exposed to lamotrigine from the mother's breast milk are not known, administer lamotrigine cautiously to breastfeeding mothers.
    3.20.2) TERATOGENICITY
    A) OROFACIAL CLEFT PALATE
    1) A preliminary data collected by the North American Antiepileptic Drug pregnancy registry in 2006 revealed an unexpectedly high prevalence of isolated, nonsyndromic, cleft palate and/or cleft lip in infants of women exposed to lamotrigine monotherapy during the first trimester of pregnancy. Five cases of oral cleft (2 isolated cleft lip, 3 isolated cleft palate) occurred among 564 women who received lamotrigine monotherapy during the first trimester, resulting in a total prevalence of 8.9 per 1000 (US Food and Drug Administration, 2006).
    B) CONGENITAL ANOMALY
    1) In an analysis of data collected by the Australian Pregnancy Register from 1999 through 2010 (n=1317), the incidence of fetal malformations that occurred with prenatal exposure to antiepileptic (AED) drug therapy during the first trimester was similar among women who used new AEDs (lamotrigine, levetiracetam, or topiramate), those with epilepsy untreated with AEDs, and those who used traditional AEDs (carbamazepine, clonazepam, or phenytoin) with the exception of valproic acid. The incidence of fetal malformations was 12/231 (5.2%), 0/22 (0%), and 1/31 (3.2%) among patients treated with lamotrigine, levetiracetam, and topiramate monotherapy, respectively, compared with 19/301 (6.3%), 0/24 (0%), 1/35 (2.9%), and 35/215 (16.3%) among patients treated with carbamazepine, clonazepam, phenytoin, or valproate monotherapy, respectively. Fetal malformations were reported in 6/139 (5.2%) of patients who were not treated with AEDs for at least the first trimester (Vajda et al, 2012).
    2) A May 2011 report from the Lamotrigine Pregnancy Registry, established by the manufacturer to collect data on pregnant women exposed to the drug, identified 1699 instances of mothers treated with lamotrigine monotherapy during the first trimester of pregnancy. Among the 1699 mothers, 141 pregnancies resulted in fetal death, elective termination, or spontaneous abortion without a reported malformation and were excluded from the analysis. There were 35 live births in which major congenital malformations (MCM) were observed (2.2%). The MCMs ranged in type and severity, with transposition of the great vessels, ventricular septal defects, hydronephrosis, and clubfoot being the most common. Among outcomes in pregnant women treated with lamotrigine/valproate combination therapy during the first trimester, 16 of 150 infants presented with MCMs (10.7%) with clubfoot and cleft palate being the most common. Among outcomes in pregnant women treated with lamotrigine plus 1 or more other anticonvulsants (other than valproate) during the first trimester, 12 of 430 infants presented with MCMs (2.8%) with esophageal defects being the most common MCM. There was no evidence to suggest that an increase in the lamotrigine dose increased the proportion of infants born with a MCM (1) (Cunnington et al, 2011).
    3) A July 2005 report from the Lamotrigine Pregnancy Registry, established by the manufacturer to collect data on pregnant women exposed to the drug, identified 648 instances of mothers treated with lamotrigine monotherapy during the first trimester of pregnancy. Sixteen live births and 1 induced abortion with congenital abnormalities were noted in this group. In mothers treated with lamotrigine plus 1 or more other anticonvulsants during the first trimester, 17 of 337 infants presented with anomalies. However, there was no consistent pattern of anomalies among the birth defects reported to the registry (Anon, July 2005).
    C) LACK OF EFFECT
    1) There was not an increased risk of isolated orofacial cleft (OC) relative to other malformations in neonates who were exposed to maternal lamotrigine compared with those who were not exposed to any antiepileptic drugs in a population-based case-control study (n=85,563). The study included 19 registries comprising 5511 OC cases and 80,052 non-OC controls. For isolated OC in lamotrigine-exposed neonates, the risk was not increased relative to other malformations (odds ratio adjusted for maternal age [adjOR] = 0.8). There were also no increases in risk relative to other malformations for any of the other 3 OC categories: isolated and multiply malformed OC (adjOR = 0.67), isolated cleft palate (CP; adjOR = 1.01), and isolated and multiply malformed CP (adjOR = 0.79) with maternal lamotrigine exposure. There were 72 lamotrigine mono- or polytherapy-exposed registrations, 40 of which were lamotrigine monotherapy. For lamotrigine monotherapy, the 40 total cases corresponded to a prevalence of 0.47 cases of OC per 1000 registrations (Dolk et al, 2008).
    2) A series of observational cohort studies suggested that lamotrigine does not cause an increased rate of congenital anomalies when used during pregnancy. Of 68 pregnant women who took the drug, three discontinued the drug before the last menstrual period; 59 were exposed during the first trimester versus 6 during the second or third trimester. Of the 59 exposed during the first trimester, there were 39 births (31 without congenital anomalies), ten spontaneous abortions, 1 missed abortion, and 9 pregnancies intentionally terminated. Three infants were delivered full term with congenital anomalies including congenital respiratory stridor (mother also exposed to phenytoin), palatal cleft, hypospadias, and undescended testes (mother also exposed to carbamazepine, valproic acid, and phenytoin), and ventricular septal defect (mother also exposed to phenobarbitone and valproic acid). One infant was delivered prematurely with abdominal distension and possible abdominal intestinal obstruction. The mother had been exposed to labetalol and experienced preeclamptic toxemia (Wilton et al, 1998).
    3) CASE SERIES: A case series described the effects of lamotrigine use during pregnancy for the treatment of bipolar disorder in 3 different women. In the first case, a 29-year-old woman presented with depression, irritability, and panic attacks at 26 weeks of gestation. The patient was previously treated with fluoxetine 60 mg/day until her 8th week of pregnancy and lamotrigine 300 mg/day until 2 weeks before conception. At week 32, the patient was restarted on lamotrigine 25 mg/day, a dose that was increased to 50 mg/day after 2 weeks and continued until delivery. The infant had no signs of respiratory distress upon delivery but was transferred to the neonatal intensive care unit 3 days later due to decreased oxygen saturation. The infant was intubated and treated with antibiotics for 7 days for possible infection. The infant was discharged 7 days later. The second case, a 34-year-old woman receiving lamotrigine 200 mg/day during pregnancy that was increased to 250 mg/day at week 23 due to decreased sleep and irritability, delivered a healthy male infant with no noted abnormalities. Finally, a 33-year-old woman was administered lamotrigine 25 mg/day that was titrated to 200 mg/day during pregnancy. At week 26, the patient complained of increased anxiety and lability of mood, for which the dose was increased to 250 mg/day and clonazepam 0.5 mg twice daily was initiated. The patient delivered a healthy female infant. After 18 months, all 3 infants displayed normal growth and development (Wakil et al, 2009).
    4) CASE REPORT: No adverse effects or signs of embryopathy were reported in a newborn infant whose 24-year-old mother had been maintained on lamotrigine for seizure control during pregnancy (Rambeck et al, 1997a).
    5) There was no clear evidence of lamotrigine-induced teratogenicity in a study that analyzed 42 pregnancies to evaluate the safety of lamotrigine (Richens, 1994).
    D) ANIMAL STUDIES
    1) Female mice were given intraperitoneal lamotrigine at various doses on days 7 and 8 of gestation. At necropsy, fetuses given low-dose lamotrigine (50 to 100 mg/kg) exhibited maxillary-mandibular hypoplasia, exencephaly, cleft palate, median facial cleft, urogenital anomalies, and various degrees of caudal regression. Fetuses given high-dose lamotrigine (200 mg/kg) developed significant intrauterine growth retardation. Although fetuses in the single- and multiple dose groups both developed skeletal malformations and developmental delay of the skeleton, the effects appeared dose-dependent. The saline and untreated groups had a low incidence of resorption and exencephaly but no other anomalies (Padmanabhan et al, 2003).
    2) There was no evidence of teratogenicity in mice, rats, or rabbits at maternal lamotrigine doses up to 1.2, 0.5, and 1.1 times, respectively, the highest usual human dose (500 mg/day) when given orally during organogenesis. There was no effect on fertility, teratogenesis, or postnatal development in rats at maternal doses equivalent to 0.4 times the highest usual human dose on a mg/m(2) basis given prior to and during mating and throughout gestation and lactation (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).
    3) The administration of lamotrigine up to 125, 25, and 30 mg/kg to pregnant mice, rats, and rabbits, respectively, during organogenesis resulted in decreased fetal body weight and increased fetal skeletal variations in mice and rabbits at maternally toxic doses. Lamotrigine doses in mice, rats, and rabbits of 75, 6.25, and 30 mg/kg, respectively, were determined to be no effect doses for embryofetal developmental toxicity and are similar to or less than the recommended human dose of 400 mg/day based on body surface area (mg/m(2)) (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011a).
    4) The administration of oral lamotrigine 5 or 25 mg/kg to pregnant rats during organogenesis resulted in behavioral abnormalities in exposed offspring at both doses and maternal toxicity at the 25 mg/kg dose. The lowest effect dose for neurotoxicity was less than the recommended human dose of 400 mg/day based on body surface area (mg/m(2)) (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011a).
    5) Although lamotrigine did not result in teratogenicity in rat studies, lamotrigine does decrease fetal folate concentrations in rats, an effect associated with teratogenesis (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011a; Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) Lamotrigine is classified by the manufacturer as FDA Pregnancy Category C (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011a).
    B) PREGNANCY REGISTRY
    1) The North American Antiepileptic Drug Pregnancy Registry maintains a pregnancy registry for women exposed to lamotrigine during pregnancy. Patients are encouraged to enroll by contacting the registry at 1-888-233-2334 or http://www.aedpregnancyregistry.org (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011a).
    C) COGNITIVE ABILITY
    1) In an interim analysis of the prospective observational Neurodevelopmental Effects of Antiepileptic Drugs study, IQ testing at ages 2 and 3 years (n=310) and age 4.5 years (n=209) following prenatal exposure to the antiepileptic drugs (AEDs) valproate, carbamazepine, lamotrigine, or phenytoin revealed pediatric cognitive abilities that correlated with maternal IQ for all AEDs, but valproate and remained consistent over time. Dose-dependent effects were observed for valproate but not for each of the other AEDs. Using the Bayley Scale of Infant Development at age 2 and the Differential Ability Scale at ages 3 and 4.5 to assess cognitive development, adjusted mean IQ at age 4.5 in the carbamazepine group was 107 (n=54); 106 in the lamotrigine group (n=73); 106 in the phenytoin group (n=43), and 96 in the valproate group (n=39). IQ improved from age 3 to 4.5 years in the carbamazepine, lamotrigine, and phenytoin groups, while no improvement occurred in the valproate group from age 3 to 4.5 years. The incidence of marked cognitive impairment (IQ less than 70) at age 4.5 years was highest in the valproate group (10%) compared with the other AEDs (0% to 4%). Children in all 4 AED groups had impaired verbal abilities compared to nonverbal skills (Meador et al, 2012).
    D) LAMOTRIGINE CONCENTRATION IN AMNIOTIC FLUID AND CORD BLOOD
    1) In a study of 6 mother-infant pairs, there was a strong correlation between maternal serum lamotrigine levels and lamotrigine levels in amniotic fluid and cord blood. Mothers were diagnosed with seizures of localized onset and received a mean daily lamotrigine dose of 458.3 mg (range, 200 to 650 mg) divided into 2 daily doses. The mean maternal lamotrigine serum, amniotic fluid, and cord blood levels were 4 mcg/mL (range, 1.1 to 6.5 mcg/mL), 2.3 mcg/mL (range, 0.8 to 3.8 mcg/mL), and 3.1 mcg/mL (range, 1.4 to 6.2 mcg/mL), respectively. The level-by-dose ratios for maternal serum, amniotic fluid, and cord blood were 0.9, 0.51, and 0.71, respectively. The mean penetration ratio into amniotic fluid was 0.58 (range, 0.31 to 0.75) and into fetal circulation (calculated using umbilical cord blood levels) was 0.81 (range, 0.48 to 1.27). Blood samples were taken at the time of delivery under steady-state conditions and were not trough levels. There was no significant correlation between lamotrigine dosage and maternal serum levels (Paulzen et al, 2015).
    E) LACK OF EFFECT
    1) CASE REPORT: A 24-year-old woman was maintained on lamotrigine during pregnancy with good seizure control. No adverse effects in the fetus were observed. Serum lamotrigine concentrations in the newborn (up to 2.8 mcg/mL) were reported (Rambeck et al, 1997).
    2) CASE SERIES: A case series described the effects of lamotrigine use during pregnancy for the treatment of bipolar disorder in 3 different women. In the first case, a 29-year-old woman presented with depression, irritability, and panic attacks at 26 weeks of gestation. The patient was previously treated with fluoxetine 60 mg/day until her 8th week of pregnancy and lamotrigine 300 mg/day until 2 weeks before conception. At week 32, the patient was restarted on lamotrigine 25 mg/day, a dose that was increased to 50 mg/day after 2 weeks and continued until delivery. The infant had no signs of respiratory distress upon delivery but was transferred to the neonatal intensive care unit 3 days later due to decreased oxygen saturation. The infant was intubated and treated with antibiotics for 7 days for possible infection. The infant was discharged 7 days later. The second case, a 34-year-old woman receiving lamotrigine 200 mg/day during pregnancy that was increased to 250 mg/day at week 23 due to decreased sleep and irritability, delivered a healthy male infant with no noted abnormalities. Finally, a 33-year-old woman was administered lamotrigine 25 mg/day that was titrated to 200 mg/day during pregnancy. At week 26, the patient complained of increased anxiety and lability of mood, for which the dose was increased to 250 mg/day and clonazepam 0.5 mg twice daily was initiated. The patient delivered a healthy female infant. After 18 months, all 3 infants displayed normal growth and development (Wakil et al, 2009).
    F) ANIMAL STUDIES
    1) Maternal toxicity and secondary fetal toxicity resulting in reduced fetal weight and/or delayed ossification were observed in mice and rats at maternal lamotrigine doses up to 1.2 and 0.5 times, respectively, the highest usual human dose (500 mg/day) administered orally during organogenesis (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).
    2) The administration of oral lamotrigine 5, 10, or 20 mg/kg during late gestation resulted in an increased incidence of offspring mortality, including stillbirths, as well as maternal toxicity at the 10 and 20 mg/kg doses. The lowest effect dose for peri- or postnatal developmental toxicity was less than the recommended human dose of 400 mg/day based on body surface area (mg/m(2)) (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011a).
    3) The incidence of intrauterine death without signs of teratogenicity was increased in rats at maternal lamotrigine intravenous doses 0.6 times the highest usual human dose. A behavioral teratology study demonstrated a significantly longer latent period for open field exploration and a lower frequency of offspring rearing at day 21 postpartum following maternal lamotrigine doses of 5 mg/kg/day or higher (0.1 times the clinical dose on a mg/m(2) basis). Time to completion for a swimming maze test was increased in rats at a maternal lamotrigine dose of 25 mg/kg/day (0.5 times the clinical dose) on days 39 to 44 postpartum. Maternal toxicity and fetal death were observed in rats at maternal lamotrigine doses of 0.1, 0.14, or 0.3 times the highest human maintenance dose when given orally during days 15 to 20 of gestation. In mothers, food consumption and weight gain were lower and the gestation period was slightly longer compared with the control group (22.6 vs 22 days). Stillbirths were reported at all 3 doses. Postnatal death was also observed in the 2 highest doses and occurred between day 1 and 20. Some of these deaths are most likely drug-related and not due to maternal toxicity. A no-observed-effect level determination could not be made for this study (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) Monitor breastfed infants closely for signs or symptoms of lamotrigine toxicity, including apnea, drowsiness, or poor sucking. Because the effects on nursing infants exposed to lamotrigine from the mother's breast milk are not known, administer lamotrigine cautiously to breastfeeding mothers (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011a).
    2) In multiple small studies, lamotrigine plasma concentrations of breastfed infants were as high as 50% of the maternal serum concentrations. If lamotrigine doses are not reduced postpartum, infants and neonates may be at increased risk of raised serum levels. In addition, lamotrigine exposure may be increased due to the immaturity of the infant glucuronidation capacity required for drug clearance (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011a).
    3) In a prospective study that investigated lamotrigine pharmacokinetics in 9 epileptic pregnant women, the median lamotrigine concentration ratio of breast milk to maternal blood was 0.59 mcg/mL (range, 0.35 to 0.86 mcg/mL). The lamotrigine concentration in the breast milk decreased over time postpartum (Fotopoulou et al, 2009).
    4) Lamotrigine milk to plasma (M/P) ratio was 41.3% and the infant/maternal ratio of total lamotrigine concentration was 18.3% in a prospective observational study of 30 lamotrigine-treated nursing mothers (mean dose, 386.5 mg) and their infants. The M/P ratio calculated using each participant's mean breast milk concentration ranged from 5.7% to 147.1%. Using minimum and maximum breast milk concentrations for each participant, M/P ratios were 26.5% and 63.1%, respectively. Infant/maternal ratio of free lamotrigine concentration was 30.9%, 1.7 times higher than the total. Infants had a 1.8 times higher ratio of free lamotrigine/total lamotrigine compared with their mothers (53.5% vs 29.5%). The theoretical infant dose and relative infant dose were 0.51 mg/kg/d and 9.2%, respectively. Univariate Pearson correlation coefficients (r) demonstrated significant positive correlations of lamotrigine concentration in breast milk with maternal daily lamotrigine dose (r=0.63), total lamotrigine in the maternal plasma (r=0.37), and free lamotrigine in the maternal plasma (r=0.51). Maternal dose (F(1147)=25.62) and free lamotrigine concentration in the maternal plasma (F(1147)=17.31) were significant predictors of lamotrigine breast milk concentration in a multiple regression analysis; the interaction of these 2 predictors was also significant (F(1147)=6.44). The final regression model accounted for 45.6% of variability in lamotrigine breast milk concentrations (F(3147)=41.11) (Newport et al, 2008).
    5) The evaluation of six infants who were breastfed by mothers treated with lamotrigine (mean dose, 400 mg/day) did not reveal any adverse effects. The mean infant dose of lamotrigine received through breast milk was 0.45 mg/kg/day. The mean infant plasma concentration as a percent of the mother's plasma concentration was 18% (Page-Sharp et al, 2006). This report confirms the results of previously published data (Rambeck et al, 1997a; Tomson & Ohman, 1997; Ohman & Vitols, 2000; Gentile, 2005).
    6) Lamotrigine levels were measured on day 10 of life in 4 full-term nursing infants born to epileptic mothers treated with lamotrigine monotherapy. Serum levels ranged from less than 1 to 2 mcg/mL and were a mean of 30% (range, 20% to 43%) of the maternal lamotrigine levels. No significant decline was detected in 2 infants with repeated levels at 2 months. Both infants were nursing with supplemental formula 2 to 3 times a day. The authors suggest that the higher than expected drug levels in the neonate were a result of immature enzyme systems in the infants, specifically hepatic glucuronidation. No short-term adverse effects were observed in the infants (Liporace et al, 2004).
    7) Serum lamotrigine levels in 3 women and their nursed infants were measured and the infants' intake of lamotrigine was estimated to be 0.5 to 1 mg/kg/day. None of the infants demonstrated signs of adverse effects (Ohman & Vitols, 2000).
    8) A 16-day-old infant developed severe apnea and required resuscitation following exposure to lamotrigine during breastfeeding. The mother was taking lamotrigine in increasing doses throughout pregnancy and during breastfeeding (dose, 850 mg). The infant's lamotrigine serum concentrations 12.5 hours after birth and at the time of admission were 7.71 mcg/mL and 4.87 mcg/mL, respectively. Breastfeeding was discontinued on day 17 postpartum and the infant recovered completely (Nordmo et al, 2009).
    3.20.5) FERTILITY
    A) LACK OF INFORMATION
    1) The effect of lamotrigine on human fertility is not known (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).
    B) ANIMAL STUDIES
    1) RATS: There was no evidence of impaired fertility in rats given oral lamotrigine doses of up to 20 mg/kg/day. It should be noted that all doses were less than the recommended human dose of 400 mg/day based on body surface area (mg/m(2)) (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011).
    2) RATS: No adverse effect on fertility was seen in rats given oral lamotrigine at doses up to 2.4 times the highest usual maintenance dose (8.33 mg/kg/day) or 0.4 times the human dose on a mg/m(2) basis (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007).

Carcinogenicity

    3.21.2) SUMMARY/HUMAN
    A) At the time of this review, the manufacturer does not report any carcinogenic potential of lamotrigine in humans.
    3.21.4) ANIMAL STUDIES
    A) LACK OF EFFECT
    1) Lamotrigine was administered orally to mice and rats at doses of 30 mg/kg/day and 10 to 15 mg/kg/day, respectively, for 2 years with no evidence of carcinogenicity at the highest dose tested (less than the human dose of 400 mg/day on a mg/m(2) basis) (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011).

Genotoxicity

    A) There was no evidence of genotoxicity or clastogenicity in the following tests: in vitro gene mutation with Ames and mouse lymphoma assays, in vitro human lymphocyte assay, and in vivo rat bone marrow assay. (Prod Info LAMICTAL(R) XR(TM) oral extended-release tablets, 2011).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) The value of plasma lamotrigine monitoring has not been established.
    B) Monitor vital signs, serial CBC with differential, serum electrolytes, liver enzymes, and renal function in symptomatic patients.
    C) Obtain an ECG and institute continuous cardiac monitoring following significant exposures. Overdoses have resulted in ECG abnormalities, including widening QRS complex and PR prolongation.
    D) Monitor for CNS and respiratory depression.
    E) Monitor CPK levels in patients with prolonged coma or seizures.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Although therapeutic serum levels have not been established, it may be advisable to monitor serum levels, if available at the hospital laboratory (Cohen et al, 1987).
    2) Monitor serum electrolytes. Hypokalemia has been reported in overdoses (Buckley et al, 1993; Harchelroad et al, 1994).
    4.1.3) URINE
    A) LABORATORY INTERFERENCE
    1) In 2 case reports, lamotrigine produced false positive results for phencyclidine (PCP) on Bio-Rad Tox rapid urine tests (Geraci et al, 2010).
    4.1.4) OTHER
    A) OTHER
    1) ECG
    a) Obtain an ECG and institute cardiac monitoring. Overdoses have resulted in ECG abnormalities, including widening QRS complex and PR prolongation (Buckley et al, 1993).

Radiographic Studies

    A) CHEST RADIOGRAPH
    1) Obtain a chest x-ray in patients with severe respiratory or CNS depression.

Methods

    A) CHROMATOGRAPHY
    1) A quantitative high-performance liquid chromatographic assay method for lamotrigine measurements in serum is described by Fraser et al (1995). The only potentially interfering drug in this assay was carbamazepine, which elutes at 2.5 times longer than lamotrigine. One study described a HPLC method for simultaneous determination of lamotrigine and carbamazepine in plasma. The lowest limit of quantitation (LOC) of plasma lamotrigine and carbamazepine concentrations was 0.1 mcg/mL for each drug. The authors also described a high-resolution gas chromatography method, which was sensitive enough to work with microsamples (LOC, 0.5 and 0.25 mcg/mL for lamotrigine and carbamazepine, respectively), but was not as sensitive as HPLC (Queiroz et al, 2001).
    2) One study described a method of isolation and characterization from urine of the major metabolite, lamotrigine N-glucuronide, by means of XAD-2 column chromatography and semi-preparative HPLC (Sinz & Remmel, 1991).
    3) One study used a high-pressure liquid chromatographic method to measure lamotrigine levels in umbilical cord serum, serum samples of a mother and child, and mother's milk during the first five postpartum months (Rambeck et al, 1997).
    B) IMMUNOASSAY
    1) One study described an immunofluorometric assay method for lamotrigine in human plasma. Intraassay and interassay accuracy and precision were greater than 90% and 95% respectively (Sailstad & Findlay, 1991).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients with a deliberate ingestions demonstrating cardiotoxicity, or other persistent neurotoxicity should be admitted.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Call a Poison Center for assistance in managing patients with severe toxicity or in whom the diagnosis is unclear.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) All patients with deliberate self-harm ingestions should be evaluated in a healthcare facility and monitored until symptoms resolve. Children with unintentional ingestions who are symptomatic should be observed in a healthcare facility.

Monitoring

    A) The value of plasma lamotrigine monitoring has not been established.
    B) Monitor vital signs, serial CBC with differential, serum electrolytes, liver enzymes, and renal function in symptomatic patients.
    C) Obtain an ECG and institute continuous cardiac monitoring following significant exposures. Overdoses have resulted in ECG abnormalities, including widening QRS complex and PR prolongation.
    D) Monitor for CNS and respiratory depression.
    E) Monitor CPK levels in patients with prolonged coma or seizures.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) PREHOSPITAL: Prehospital gastrointestinal decontamination is not recommended because of the potential for CNS depression and subsequent aspiration.
    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) In one study, oral activated charcoal significantly decreased the gastrointestinal absorption and accelerated the elimination of lamotrigine. In 6 subjects who were given lamotrigine 100 mg orally, a single dose (50 g) of oral activated charcoal administered 30 minutes after the dose, decreased the AUC of lamotrigine to 58% of the respective variable without oral activated charcoal. Repeated oral activated charcoal decreased the AUC (from 6 hours to infinity) and half-life of lamotrigine to 39% and 44%, respectively (Keranen et al, 2010).
    6.5.3) TREATMENT
    A) MONITORING OF PATIENT
    1) The value of plasma lamotrigine monitoring has not been established.
    2) Monitor vital signs, serial CBC with differential, serum electrolytes, liver enzymes, and renal function in symptomatic patients.
    3) Obtain an ECG and institute continuous cardiac monitoring following significant exposures. Overdoses have resulted in ECG abnormalities, including widening QRS complex and PR prolongation.
    4) Monitor for CNS and respiratory depression.
    5) Monitor CPK levels in patients with prolonged coma or seizures.
    B) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    2) DOPAMINE
    a) DOSE: Begin at 5 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). If hypotension persists, dopamine may need to be discontinued and a more potent vasoconstrictor (eg, norepinephrine) should be considered (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    b) CAUTION: If ventricular dysrhythmias occur, decrease rate of administration (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    3) NOREPINEPHRINE
    a) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    b) DOSE
    1) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    C) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    D) HYPOKALEMIA
    1) Administer potassium chloride (oral or IV) to patients with hypokalemia. Do not exceed 10 to 15 milliequivalents per hour IV in adults or 0.25 to 0.5 milliequivalents per kilogram per hour in children (Olson, 1994).
    2) Monitor ECG and serum potassium levels.
    E) FAT EMULSION
    1) Intravenous lipid emulsion (ILE) has been effective in reversing severe cardiovascular toxicity from local anesthetic overdose in animal studies and human case reports. Several animal studies and human case reports have also evaluated the use of ILE for patients following exposure to other drugs. Although the results of these studies are mixed, there is increasing evidence that it can rapidly reverse cardiovascular toxicity and improve mental function for a wide variety of lipid soluble drugs. It may be reasonable to consider ILE in patients with severe symptoms who are failing standard resuscitative measures (Lavonas et al, 2015).
    2) The American College of Medical Toxicology has issued the following guidelines for lipid resuscitation therapy (LRT) in the management of overdose in cases involving a highly lipid soluble xenobiotic where the patient is hemodynamically unstable, unresponsive to standard resuscitation measures (ie, fluid replacement, inotropes and pressors). The decision to use LRT is based on the judgement of the treating physician. When possible, it is recommended these therapies be administered with the consultation of a medical toxicologist (American College of Medical Toxicology, 2016; American College of Medical Toxicology, 2011):
    a) Initial intravenous bolus of 1.5 mL/kg 20% lipid emulsion (eg, Intralipid) over 2 to 3 minutes. Asystolic patients or patients with pulseless electrical activity may have a repeat dose, if there is no response to the initial bolus.
    b) Follow with an intravenous infusion of 0.25 mL/kg/min of 20% lipid emulsion (eg, Intralipid). Evaluate the patient's response after 3 minutes at this infusion rate. The infusion rate may be decreased to 0.025 mL/kg/min (ie, 1/10 the initial rate) in patients with a significant response. This recommendation has been proposed because of possible adverse effects from very high cumulative rates of lipid infusion. Monitor blood pressure, heart rate, and other hemodynamic parameters every 15 minutes during the infusion.
    c) If there is an initial response to the bolus followed by the re-emergence of hemodynamic instability during the lowest-dose infusion, the infusion rate may be increased back to 0.25 mL/kg/min or, in severe cases, the bolus could be repeated. A maximum dose of 10 mL/kg has been recommended by some sources.
    d) Where possible, LRT should be terminated after 1 hour or less, if the patient's clinical status permits. In cases where the patient's stability is dependent on continued lipid infusion, longer treatment may be appropriate.
    3) CASE REPORT: A 44-year-old woman ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g, and was found unconscious at home. After admission, a tonic-clonic seizure, rigidity, hyperreflexia, and reflex myocloni were observed. Laboratory results revealed sodium level of 129 mmol/L, creatinine kinase of 1915 Units/L, and creatine kinase-MB of 31 Units/L. Despite supportive therapy, including 8 hours of hemodialysis, her rigidity and hyperreflexia did not improve. Following treatment with an intravenous bolus (150 mL, 2.5 mL/kg) of 20% lipid emulsion, all symptoms resolved completely. On day 6, she was transferred to a psychiatric ward. Before hemodialysis, plasma concentrations of lamotrigine, diazepam, and venlafaxine were 42.4 mg/L (therapeutic range, 1 to 4 mg/L), 560 mcg/L (therapeutic range, 200 to 500 mcg/L), and 1254 mcg/L (therapeutic range, 30 to 150 mcg/L), respectively (Dagtekin et al, 2011).
    4) CASE REPORT: A 50-year-old woman with a history of type 2 diabetes and bipolar disorder developed coma (Glasgow Coma Scale 7) approximately 2 hours after ingesting up to 3.5 g of lamotrigine. She was hemodynamically stable and a chest X-ray was normal. An ECG revealed sinus tachycardia and arterial blood gas analysis revealed FiO2 = 0.30, pH 7.36, PaCO2 39 mm Hg, PaO2 85 mm Hg, and base deficit -3 mmol/L. At this time, serum lamotrigine concentration was 27.1 mcg/mL (reference, 2 to 16 mcg/mL). She was treated with activated charcoal, IV magnesium and sodium bicarbonate. Four hours postadmission, another ECG revealed a widening of the QRS complex with left bundle branch block. All laboratory results, including troponin-I levels, were normal and vital signs were stable. Six hours postingestion, serum lamotrigine concentration increased to 29.7 mcg/mL. Despite supportive therapy, including sodium bicarbonate, the ECG did not improve. At this time, she was treated with an intravenous bolus (1.5 mL/kg) of 20% lipid emulsion and QRS narrowed within a few minutes. She continued to receive lipids (0.5 mL/kg/min) for another 10 hours. She was discharged 2 days postingestion (Castanares-Zapatero et al, 2011).
    5) CASE REPORT: An 18-year-old woman with a history of epilepsy presented with agitation, disorientation and noncooperative with a Glasgow Coma Scale (GCS) score of 12 about an hour after ingesting an unknown amount of lamotrigine 100 mg tablets and sertraline 50 mg tablets. Physical examination revealed vertical nystagmus and myoclonia. She responded to questions with dysphasia within an hour of receiving IV lipid emulsion therapy (bolus of 100 mL and 0.5 mL/kg/min infusion for 2 hours; total dose, 3100 mL). She had a GCS score of 15 and mild dysarthria 6 hours later (Eren Cevik et al, 2014).
    F) VENTRICULAR ARRHYTHMIA
    1) Obtain an ECG, institute continuous cardiac monitoring and administer oxygen. Evaluate for hypoxia, acidosis, and electrolyte disorders (particularly hypokalemia, hypocalcemia, and hypomagnesemia). Sodium bicarbonate is generally first line therapy for QRS widening and ventricular dysrhythmias. In patients unresponsive to bicarbonate, consider lidocaine.
    2) SERUM ALKALINIZATION
    a) Administer sodium bicarbonate. A reasonable starting dose is 1 to 2 mEq/kg by intravenous bolus, repeated as needed. Maintain arterial pH between 7.45 and 7.55. Monitor serial ECGs and arterial blood gases frequently.
    3) LIDOCAINE
    a) LIDOCAINE/INDICATIONS
    1) Ventricular tachycardia or ventricular fibrillation (Prod Info Lidocaine HCl intravenous injection solution, 2006; Neumar et al, 2010; Vanden Hoek et al, 2010).
    b) LIDOCAINE/DOSE
    1) ADULT: 1 to 1.5 milligrams/kilogram via intravenous push. For refractory VT/VF an additional bolus of 0.5 to 0.75 milligram/kilogram can be given at 5 to 10 minute intervals to a maximum dose of 3 milligrams/kilogram (Neumar et al, 2010). Only bolus therapy is recommended during cardiac arrest.
    a) Once circulation has been restored begin a maintenance infusion of 1 to 4 milligrams per minute. If dysrhythmias recur during infusion repeat 0.5 milligram/kilogram bolus and increase the infusion rate incrementally (maximal infusion rate is 4 milligrams/minute) (Neumar et al, 2010).
    2) CHILD: 1 milligram/kilogram initial bolus IV/IO; followed by a continuous infusion of 20 to 50 micrograms/kilogram/minute (de Caen et al, 2015).
    c) LIDOCAINE/MAJOR ADVERSE REACTIONS
    1) Paresthesias; muscle twitching; confusion; slurred speech; seizures; respiratory depression or arrest; bradycardia; coma. May cause significant AV block or worsen pre-existing block. Prophylactic pacemaker may be required in the face of bifascicular, second degree, or third degree heart block (Prod Info Lidocaine HCl intravenous injection solution, 2006; Neumar et al, 2010).
    d) LIDOCAINE/MONITORING PARAMETERS
    1) Monitor ECG continuously; plasma concentrations as indicated (Prod Info Lidocaine HCl intravenous injection solution, 2006).
    4) Severe sodium channel blockade after a massive lamotrigine overdose has been reported (Nogar et al, 2011).
    a) CASE REPORT: A 48-year-old woman, with a medical history of depression and gastric bypass surgery, as well as a remote history of seizure disorder, presented to the ED unconscious with one episode of tonic-clonic seizure after ingesting about 7.5 g of lamotrigine (lamotrigine concentration: 74.7 mcg/mL). Her vital signs included a heart rate of 131 beats/min, blood pressure of 107/68 mmHg, temperature of 99.4 degrees F, and a respiratory rate of 16 breaths/min. She was treated with supportive care and was intubated with rocuronium. An ECG revealed a narrow-complex sinus tachycardia, with a QRS duration of 106 msec. She developed a one-minute tonic-clonic seizure about 2.5 hours after presentation and received a 2 mg bolus of lorazepam. At this time, a second ECG revealed normal sinus rhythm, a heart rate of 86 beats/min, a QRS duration of 110 msec, and a small R-wave in aVR. Despite treatment with 2 boluses of sodium bicarbonate, her QRS duration did not change and she remained hemodynamically unstable. She developed status epilepticus 10 minutes later and an ECG showed a wide-complex tachycardia, and she became pulseless. She received sodium bicarbonate, 360 mL of 20% lipid emulsion, lidocaine, amiodarone, and 2 g calcium chloride during 45 minutes of resuscitation. Her pulses were reestablished with QRS narrowing with each defibrillation, but were widened again with recurrent seizure activity. Following the termination of seizures with vecuronium, she was finally stabilized. An ECG after resuscitation revealed a junctional tachycardia with a 3 mm R-wave in aVR. She was treated with fosphenytoin and propofol in the ICU, and was gradually weaned from sedation over 3 days when it was observed that the patient had extensor posturing. A MRI revealed brain edema consistent with anoxic injury and her family elected to withdraw care 4 days after presentation (Nogar et al, 2011).
    G) RHABDOMYOLYSIS
    1) SUMMARY: Early aggressive fluid replacement is the mainstay of therapy and may help prevent renal insufficiency. Diuretics such as mannitol or furosemide may be added if necessary to maintain urine output but only after volume status has been restored as hypovolemia will increase renal tubular damage. Urinary alkalinization is NOT routinely recommended.
    2) Initial treatment should be directed towards controlling acute metabolic disturbances such as hyperkalemia, hyperthermia, and hypovolemia. Control seizures, agitation, and muscle contractions (Erdman & Dart, 2004).
    3) FLUID REPLACEMENT: Early and aggressive fluid replacement is the mainstay of therapy to prevent renal failure. Vigorous fluid replacement with 0.9% saline (10 to 15 mL/kg/hour) is necessary even if there is no evidence of dehydration. Several liters of fluid may be needed within the first 24 hours (Walter & Catenacci, 2008; Camp, 2009; Huerta-Alardin et al, 2005; Criddle, 2003; Polderman, 2004). Hypovolemia, increased insensible losses, and third spacing of fluid commonly increase fluid requirements. Strive to maintain a urine output of at least 1 to 2 mL/kg/hour (or greater than 150 to 300 mL/hour) (Walter & Catenacci, 2008; Camp, 2009; Erdman & Dart, 2004; Criddle, 2003). To maintain a urine output this high, 500 to 1000 mL of fluid per hour may be required (Criddle, 2003). Monitor fluid input and urine output, plus insensible losses. Monitor for evidence of fluid overload and compartment syndrome; monitor serum electrolytes, CK, and renal function tests.
    4) DIURETICS: Diuretics (eg, mannitol or furosemide) may be needed to ensure adequate urine output and to prevent acute renal failure when used in combination with aggressive fluid therapy. Loop diuretics increase tubular flow and decrease deposition of myoglobin. These agents should be used only after volume status has been restored, as hypovolemia will increase renal tubular damage. If the patient is maintaining adequate urine output, loop diuretics are not necessary (Vanholder et al, 2000).
    5) URINARY ALKALINIZATION: Alkalinization of the urine is not routinely recommended, as it has never been documented to reduce nephrotoxicity, and may cause complications such as hypocalcemia and hypokalemia (Walter & Catenacci, 2008; Huerta-Alardin et al, 2005; Brown et al, 2004; Polderman, 2004). Retrospective studies have failed to demonstrate any clinical benefit from the use of urinary alkalinization (Brown et al, 2004; Polderman, 2004; Homsi et al, 1997).
    6) MANNITOL/INDICATIONS
    a) Osmotic diuretic used in the management of rhabdomyolysis and myoglobinuria (Zimmerman & Shen, 2013).
    7) RHABDOMYOLYSIS/MYOGLOBINURIA
    a) ADULT: TEST DOSE: (for patients with marked oliguria or those with inadequate renal function) 0.2 g/kg IV as a 15% to 25% solution infused over 3 to 5 minutes to produce a urine flow of at least 30 to 50 mL/hr; a second test dose may be given if urine flow does not increase within 2 to 3 hours. The patient should be reevaluated if there is inadequate response following the second test dose (Prod Info MANNITOL intravenous injection, 2009). TREATMENT DOSE: 50 to 100 g IV as a 15% to 25% solution may be used. The rate should be adjusted to maintain urinary output at 30 to 50 mL/hour (Prod Info mannitol IV injection, urologic irrigation, 2006) OR 300 to 400 mg/kg or up to 100 g IV administered as a single dose (Prod Info MANNITOL intravenous injection, 2009).
    b) PEDIATRIC: Dosing has not been established in patients less than 12 years of age(Prod Info Mannitol intravenous injection, 2009). TEST DOSE (for patients with marked oliguria or those with inadequate renal function): 0.2 g/kg or 6 g/m(2) body surface area IV as a 15% to 25% solution infused over 3 to 5 minutes to produce a urine flow of at least 30 to 50 mL/hr; a second test dose may be given if urine flow does not increase; TREATMENT DOSE: 0.25 to 2 g/kg or 60 g/m(2) body surface area IV as a 15% to 20% solution over 2 to 6 hours; do not repeat dose for persistent oliguria (Prod Info MANNITOL intravenous injection, 2009).
    8) ADVERSE EFFECTS
    a) Fluid and electrolyte imbalance, in particular sodium and potassium; expansion of the extracellular fluid volume leading to pulmonary edema or CHF exacerbations(Prod Info MANNITOL intravenous injection, 2009).
    9) PRECAUTION
    a) Contraindicated in well-established anuria or impaired renal function not responding to a test dose, pulmonary edema, CHF, severe dehydration; caution in progressive oliguria and azotemia; do not add to whole blood for transfusions(Prod Info Mannitol intravenous injection, 2009); enhanced neuromuscular blockade observed with tubocurarine(Miller et al, 1976).
    10) MONITORING PARAMETERS
    a) Renal function, urine output, fluid balance, serum potassium, serum sodium, and serum osmolality (Prod Info Mannitol intravenous injection, 2009).
    11) MAJOR ADVERSE REACTIONS: Congestive heart failure, hypernatremia, hyponatremia, hyperkalemia, renal failure, pulmonary edema, and allergic reactions.
    12) PRECAUTIONS: Contraindicated in well-established anuria or impaired renal function not responding to a test dose, pulmonary edema, CHF, severe dehydration; caution in progressive oliguria and azotemia.
    13) MONITORING PARAMETERS: Renal function, urine output, fluid balance, serum potassium levels, serum osmolarity, and CVP.

Enhanced Elimination

    A) HEMODIALYSIS
    1) Hemodialysis has not been established as an effective means of removing lamotrigine from the blood. In 6 renal failure patients approximately 17% of the amount of lamotrigine in the body was removed during 4 hours of hemodialysis. The elimination half-life of lamotrigine between dialysis treatments averaged approximately 59.6 hours, and during dialysis approximately 15.5 hours (Fillastre et al, 1993a).
    2) Approximately 20% (range, 5.6% to 35.1%) of the amount of lamotrigine present in the body was eliminated by hemodialysis during a 4-hour session (Prod Info LAMICTAL chewable dispersible oral tablets, oral tablets, orally disintegrating tablets, 2009).
    B) CASE REPORT
    1) ACETAMINOPHEN may increase the clearance of lamotrigine (Depot et al, 1990). The use of acetaminophen in the treatment of lamotrigine overdose has not been established. In one case, a patient with lamotrigine overdose was treated with oral acetaminophen. The acetaminophen dosage and effect on the clinical outcome were not provided in this brief abstract (Harchelroad et al, 1994).

Case Reports

    A) ADULT
    1) Buckley et al (1993) report a case of intentional overdose of 1350 milligrams in a 26-year-old man (singular ingestion of lamotrigine) (Buckley et al, 1993). He presented to the emergency department one hour following the ingestion alert and oriented with pulse of 96, blood pressure 154/90 mm Hg and temperature of 36 degrees Celsius. He exhibited horizontal and vertical nystagmus and was hypertonic with brisk reflexes.
    a) Blood chemistries on admission were normal with the exception of hypokalemia (potassium of 3.3 mmol/L). The ECG showed QRS widening (112 ms), with minimal change the following day. Liver and renal function tests were normal. Serum lamotrigine levels were 17.4 and 6.4 mcg/mL at 3 and 17 hours postingestion, respectively. Two months later the QRS width had decreased to less than 100 ms.
    2) A 33-year-old woman was agitated, non-verbal, tachycardic (120 BPM) and hypokalemic (2.8 mmol/L) 4 hours after ingesting 15 to 20 lamotrigine 100 milligram tablets(Harchelroad et al, 1994). Eight hours after ingestion she became lethargic and 24 hours after ingestion she had a dry mouth with thick secretions, slurred speech, pupils that were 4 mm and reacted to light but not accommodation and absent deep tendon reflexes in her lower extremities. All effects resolved by 48 hours after ingestion.
    B) PEDIATRIC
    1) One study reported seizures, hypertonia, ataxia, tremor of the limbs, and muscle weakness in a 2-year-old man within an hour of ingesting sixteen 50 milligram tablets (800 milligrams). EEG, laboratory tests, and vital signs were normal. Treatment with 2 milligrams of intravenous midazolam resolved generalized seizure activity. Gastric lavage followed by activated charcoal, saline cathartic infusion and fluid replacement were also administered. Symptoms resolved after 24 hours and the patient was discharged in 48 hours (Briassoulis et al, 1998).

Summary

    A) TOXICITY: Adults have survived overdoses of greater than 4000 mg with supportive therapy. A man developed generalized myoclonus status epilepticus and coma several hours after ingesting 6 g of lamotrigine. He recovered following supportive care. A 3-year-old girl survived an overdose of 1.15 g of lamotrigine. Another child, a 2-year-old, also survived an overdose of 800 mg. Fatalities have been reported following overdoses of up to 15 g.
    B) THERAPEUTIC: Adult maintenance doses of lamotrigine for bipolar disorder can be up to 400 mg/day and up to 500 mg/day for anti-seizure therapy. For children 2 to 12 years of age, the maintenance dose for the anti-seizure therapy can be up to 15 mg/kg/day (max 400 mg/day). For children over the age of 12, the maintenance dose for the anti-seizure therapy can be up to 500 mg/day.

Therapeutic Dose

    7.2.1) ADULT
    A) Dosing varies by indication. Lamotrigine is used alone or in combination with other medicines. Doses may require adjustment based on clinical response and/or concomitant antiepileptic drugs (Prod Info LAMICTAL ODT(R) oral disintegrating tablets, 2014; Prod Info LAMICTAL(R) oral tablets, oral chewable dispersible tablets, 2014).
    B) ORAL CHEWABLE DISPERSAL, DISINTEGRATING, AND IMMEDIATE-RELEASE TABLETS: 25 mg orally daily or every other day or 50 mg orally daily; gradually increased to a maintenance dose up to 400 mg daily (in divided doses) in patients with bipolar disorder and 500 mg/day (in divided doses) for seizures (Prod Info LAMICTAL ODT(R) oral disintegrating tablets, 2014; Prod Info LAMICTAL(R) oral tablets, oral chewable dispersible tablets, 2014).
    C) ORAL EXTENDED-RELEASE TABLETS: 25 mg orally every other day or daily 50 mg orally daily; gradually increased to a maintenance dose up to 600 mg daily (Prod Info LAMICTAL(R) XR(TM) oral extended release tablets, 2013).
    7.2.2) PEDIATRIC
    A) Lamotrigine is used alone or in combination with other medicines. Doses may require adjustment based on clinical response and/or concomitant antiepileptic drugs (Prod Info LAMICTAL ODT(R) oral disintegrating tablets, 2014; Prod Info LAMICTAL(R) oral tablets, oral chewable dispersible tablets, 2014).
    B) BIPOLAR DISORDER
    1) Safety and efficacy in pediatric patients have not been established (Prod Info LAMICTAL ODT(R) oral disintegrating tablets, 2014; Prod Info LAMICTAL(R) oral tablets, oral chewable dispersible tablets, 2014).
    C) EPILEPSY
    1) UNDER 2 YEARS
    a) Safety and efficacy in patients under 2 years were not established (Prod Info LAMICTAL ODT(R) oral disintegrating tablets, 2014; Prod Info LAMICTAL(R) oral tablets, oral chewable dispersible tablets, 2014).
    2) 2 TO 12 YEARS
    a) ORAL CHEWABLE DISPERSAL, DISINTEGRATING, AND IMMEDIATE-RELEASE TABLETS: 0.15 mg/kg to 0.6 mg/kg orally daily in divided doses; gradually increased to a maintenance dose up to 15 mg/kg daily; MAX: 400 mg/day in 2 divided doses (Prod Info LAMICTAL ODT(R) oral disintegrating tablets, 2014; Prod Info LAMICTAL(R) oral tablets, oral chewable dispersible tablets, 2014).
    b) ADMINISTRATION: ONLY use whole tablets; round down to the nearest whole tablet (Prod Info LAMICTAL ODT(R) oral disintegrating tablets, 2014; Prod Info LAMICTAL(R) oral tablets, oral chewable dispersible tablets, 2014).
    3) 13 YEARS AND OLDER
    a) ORAL CHEWABLE DISPERSAL, DISINTEGRATING, AND IMMEDIATE-RELEASE TABLETS: 25 mg orally daily or every other day or 50 mg orally daily; gradually increased to a maintenance dose up to 500 mg/day (in divided doses) (Prod Info LAMICTAL ODT(R) oral disintegrating tablets, 2014; Prod Info LAMICTAL(R) oral tablets, oral chewable dispersible tablets, 2014).
    b) ORAL EXTENDED-RELEASE TABLETS: 25 mg orally every other day or daily or 50 mg orally daily; gradually increased to a maintenance dose up to 600 mg daily (Prod Info LAMICTAL(R) XR(TM) oral extended release tablets, 2013).

Minimum Lethal Exposure

    A) ADULT
    1) Overdoses of up to 15 g have resulted in some fatalities (Prod Info Lamictal(R), lamotrigine tablets, 2000).
    2) CASE REPORT: A 19-year-old man with bipolar disorder ingested 4 g of lamotrigine (blood concentration, 35.7 mcg/mL, 19 hours post-ingestion) and developed tachycardia (123 beats/min), multiple seizures, charcoal aspiration, respiratory failure, prolongation of the QRS interval (214 ms), and completed heart block. Despite supportive care, he developed sepsis and multiorgan failure and died 10 days post-ingestion (French et al, 2011).
    3) CASE REPORT: A 48-year-old woman developed cardiac arrest with significant conduction disturbances and status epilepticus after ingesting about 7.5 g of lamotrigine (lamotrigine concentration: 74.7 mcg/mL; therapeutic range: 3 to 14 mcg/mL). Despite intensive supportive care, an MRI 3 days after admission revealed brain edema consistent with anoxic injury and her family elected to withdraw care 4 days after presentation (Nogar et al, 2011).

Maximum Tolerated Exposure

    A) ADULT
    1) In a retrospective, case series from 2003 to 2012, 57 cases of possible lamotrigine overdose, including 9 patients (6 adults and 3 children [ages: 1 year, 19 months, and 2 years]) with lamotrigine-only ingestions (median amount ingested: 2 g; range 0.5 to 13.5 g), were identified. The following adverse effects were reported: seizures (n=3), hypertension (n=4), tachycardia (n=5), tachypnea (n=4), depressed mental status (n=4), agitation (n=5), both agitation and depressed mental status (n=3), hyperreflexia and intermittent myoclonus (n=5), inducible clonus (n=2), QRS prolongation (114 to 116 ms; n=2), QTc prolongation (463 to 586 ms; n=4), and hyperthermia (n=1). Average serum lamotrigine concentration from 7 patients was 35.4 mg/L (17 to 90 mg/L; therapeutic range: 3 to 14 mg/L) (Moore et al, 2013).
    2) CASE REPORT: A 46-year-old man with a history of bipolar disorder developed bilateral synchronous generalized myoclonus several hours after ingesting about 6000 mg of lamotrigine in a suicide attempt. Before arrival to the ED, he experienced 3 generalized seizures and presented comatose (Glasgow-Coma score (GCS) of 5/15) with spasticity in all 4 limbs. Laboratory results revealed an elevated lamotrigine concentration of 100 mcmol/L (therapeutic, 1 to 4 mcmol/L). EEG monitoring revealed no epileptiform discharges anytime during the myoclonic jerks. Following supportive care, including treatment with IV lorazepam, phenytoin, phenobarbital, midazolam, propofol, thiopental and mechanical ventilation, his symptoms gradually resolved. On day 3, another EEG was completely normal (Algahtani et al, 2014).
    3) CASE REPORT: A 25-year-old man who was taking lamotrigine 400 mg/day (6 mg/kg/day; plasma concentration 12.1 mcg/mL) for generalized tonic-clonic seizures developed oculogyric crisis (2 to 3 episodes of sustained upward deviation of both eyes) after inadvertently ingesting 1600 mg/day (23.5 mg/kg/day; plasma concentration 16.5 mcg/mL) of lamotrigine during a 24-hour period. His symptoms resolved after his lamotrigine dose was decreased to 6 mg/kg/day (plasma concentration 7 mcg/mL) (Veerapandiyan et al, 2011).
    4) CASE REPORT: A 50-year-old woman with a history of type 2 diabetes and bipolar disorder developed coma and widening of QRS with left bundle branch block after ingesting up to 3.5 g of lamotrigine. Her ECG did not improve with IV sodium bicarbonate. Following treatment with an intravenous bolus (1.5 mL/kg) of 20% lipid emulsion, her QRS complexes narrowed within a few minutes. She continued to receive lipids (0.5 mL/kg/min) for another 10 hours. She was discharged 2 days postingestion (Castanares-Zapatero et al, 2011).
    5) CASE REPORT: A 24-year-old woman developed acute pancreatitis after ingesting 4200 mg of lamotrigine and two bottles of wine. Following supportive care, she recovered completely and was discharged on day 5 (Nwogbe et al, 2009).
    6) CASE REPORT: A 26-year-old man survived, following treatment, an acute lamotrigine overdose of 1350 mg with ECG signs of slight QRS widening, hypokalemia, and nystagmus (Buckley et al, 1993).
    7) CASE REPORT: An adult with a history of focal seizures developed secondarily generalized status epilepticus after ingesting 4.1 grams of lamotrigine. She recovered with supportive care (Dinnerstein et al, 2007).
    8) CASE REPORT: A 33-year-old woman presented to the ED following an acute ingestion of 1500 to 2000 mg lamotrigine in addition to unknown amounts of phenytoin and clorazepate. CNS depression, mild tachycardia and hypokalemia were present. Full recovery ensued following supportive care and treatment with acetaminophen (Harchelroad et al, 1994).
    9) CASE REPORT: Two adult women who each ingested greater than 4000 mg lamotrigine recovered with medical treatment. One woman was comatose for 8 to 12 hours and the other woman presented with somnolence, dizziness, and headache. No electrolyte abnormalities were present in either woman (Prod Info Lamictal(R), lamotrigine tablets, 2000).
    10) CASE REPORT: A 17-year-old girl recovered uneventfully following an overdose of lamotrigine (2000 mg) and gabapentin (12,000 mg). Toxic effects included somnolence, dysarthria, ataxia and lethargy (Stopforth, 1997).
    11) CASE REPORT: A 32-year-old woman survived a stated overdose of 4500 mg (absorbed estimated dose 2900 mg) with neurologic manifestations of marked ataxia and rotary nystagmus. No remarkable ECG findings were reported (O'Donnell & Bateman, 2000).
    B) PEDIATRIC
    1) OCULOGYRIC CRISIS: Three children with no history of movement disorders developed oculogyric crisis (1 to 20 episodes per day; 2 seconds to several hours duration) after taking increasing doses of lamotrigine. All patients recovered following the reduction of lamotrigine doses (Veerapandiyan et al, 2011).
    a) A 10-year-old girl who was taking lamotrigine 22 mg/kg/day (plasma concentration 6.7 mcg/mL) presented with oculogyric crisis (a few to 20 episodes/day for 3 days; duration of each episode, 2 to 3 hours; associated symptoms: tremulousness, irritability) about a week after her lamotrigine dose was increased to 25 mg/kg/day (plasma concentration 15.8 mcg/mL). MRI revealed diffuse cerebral and cerebellar atrophy. Her symptoms resolved after lamotrigine dose was decreased to 20 mg/kg/day (plasma concentration 8.5 mcg/mL).
    b) A 3-year-old boy who was taking lamotrigine 2 mg/kg/day (plasma concentration 2.1 mcg/mL) developed oculogyric crisis (1 episode/day for 1 month; duration of each episode, few seconds to 1 minute) after his lamotrigine dose was increased to 7.8 mg/kg/day. His symptoms resolved after decreasing the lamotrigine dose.
    c) An 11-year-old boy who was taking lamotrigine 5.5 mg/kg/day for absence seizures, developed recurrent episodes of oculogyric crisis (5 to 7 episodes/day for 3 months; duration of each episode, 2 to 4 seconds) after his lamotrigine dose was increased to 7 mg/kg/day (plasma concentration 9.3 mcg/mL). His symptoms resolved after his lamotrigine dose was decreased to 4 mg/kg/day.
    2) CASE REPORT: A 2-year-old boy (weight: 13.5 kg) who was taking lamotrigine 25 mg for a seizure disorder developed agitation and myoclonic jerking after ingesting about 21 lamotrigine tablets. He recovered following supportive care and was discharged 72 hours later (Moore et al, 2013).
    3) CASE REPORT: A 19-month-old boy (weight: 11.4 kg) developed vomiting, confusion, slurred speech, and agitation after ingesting 400 mg of lamotrigine. Following supportive care, he recovered and was discharged home 24 hours after ingestion (Moore et al, 2013).
    4) CASE REPORT: A 3-year-old girl developed significant somnolence, a lacy reticular blanching rash and elevated liver function tests after ingesting 46 25-mg tablets (1.15 g) of lamotrigine. Plasma lamotrigine level was 25.3 mcg/mL (elevated above adult therapeutic levels). Following supportive treatment, she recovered without further sequelae (Zidd & Hack, 2004).
    5) CASE REPORT: A 5-year-old epileptic girl developed ataxia, drowsiness, confusion, followed by vomiting and seizure exacerbation after ingesting 500 mg (25 mg/kg/day) of lamotrigine in 2 250-mg doses 12 hours apart instead of the prescribed 150 mg twice daily (15 mg/kg/day). Following the discontinuation of lamotrigine and supportive therapy, she gradually improved and was discharged 30 hours after admission (Daana et al, 2007).
    6) CASE REPORT: A 2-year-old boy recovered uneventfully following an overdose of 16 50-mg tablets (800 mg). Within an hour of the ingestion, the boy was reported to have generalized tonic-clonic seizures, ataxia, muscle weakness, and hypertonia. Vital signs, EEG, and laboratory tests were all reported within normal limits. Symptoms resolved within 24 hours following therapy with 2 mg midazolam, gastric lavage, activated charcoal, saline cathartic and fluid replacement (Briassoulis et al, 1998).
    7) CASE REPORT: A 2-year-old boy experienced 2 generalized seizures 60 minutes after ingesting up to 43 mg/kg of lamotrigine. On admission, he had mild nystagmus, drowsiness, ataxia, hyperreflexia, and vomiting. Following supportive care, he recovered completely (Close & Banks, 2010).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) A target plasma therapeutic range has not been established for lamotrigine by the product manufacturer (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007). By comparison with therapeutic plasma concentrations of standard anticonvulsants in animal models, putative therapeutic plasma levels are estimated to be in the range of 1 to 3 mcg/mL. These concentrations have been shown to inhibit photoconvulsive responses and interictal EEG changes in epileptic patients (Cohen et al, 1987).
    2) Serum lamotrigine concentrations in a target range of 1 to 13 mcg/mL have been proposed when lamotrigine is used as an add on-drug. Serum concentrations above 13 to 14 mcg/mL have been associated with a steep increase in adverse effects (Froscher et al, 2002).
    3) PEDIATRIC: Lamotrigine plasma levels equal to or greater than 8 mcg/mL have been well tolerated by pediatric patients according to 1 study. Therapeutic lamotrigine plasma concentrations ranged from approximately 4 mcg/mL to 5 mcg/mL in this study (Uvebrant & Bauziene, 1994).
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) ADULT
    a) A 48-year-old woman developed cardiac arrest with significant conduction disturbances and status epilepticus after ingesting about 7.5 g of lamotrigine (lamotrigine concentration: 74.7 mcg/mL; therapeutic range: 3 to 14 mcg/mL). Despite intensive supportive care, a MRI obtained 3 days after admission revealed brain edema consistent with anoxic injury and her family elected to withdraw care 4 days after presentation (Nogar et al, 2011).
    b) A 19-year-old man with bipolar disorder ingested 4 g of lamotrigine (blood concentration, 35.7 mcg/mL, 19 hours post-ingestion) and developed tachycardia (123 beats/min), multiple seizures, charcoal aspiration, respiratory failure, prolongation of the QRS interval (214 ms), and completed heart block. Despite supportive care, he developed sepsis and multiorgan failure and died 10 days post-ingestion (French et al, 2011).
    c) A 50-year-old woman with a history of type 2 diabetes and bipolar disorder developed coma and widening of QRS with left bundle branch block after ingesting up to 3.5 g of lamotrigine. Despite supportive therapy, including activated charcoal, IV magnesium, and sodium bicarbonate, the ECG did not improve. Serum lamotrigine concentrations were 27.1 mcg/mL (reference, 2 to 16 mcg/mL) and 29.7 mcg/mL, 2 and 6 hours postingestion, respectively. Following treatment with an intravenous bolus (1.5 mL/kg) of 20% lipid emulsion, all symptoms resolved within a few minutes. She continued to receive lipids (0.5 mL/kg/min) for another 10 hours. She was discharged 2 days postingestion (Castanares-Zapatero et al, 2011).
    d) COMBINATION OVERDOSE: A woman who ingested diazepam 200 mg, lamotrigine 20 g, and venlafaxine 4.5 g developed coma, seizures, rigidity, and hyperreflexia. Plasma concentrations of lamotrigine, diazepam, and venlafaxine were 42.4 mg/L (therapeutic range, 1 to 4 mg/L), 560 mcg/L (therapeutic range, 200 to 500 mcg/L), and 1254 mcg/L (therapeutic range, 30 to 150 mcg/L), respectively. Following treatment with lipid emulsion, all symptoms resolved completely (Dagtekin et al, 2011).
    e) Serum lamotrigine concentrations above 13 mcg/mL have been associated with a steep increase in adverse effects, such as tremor (Froscher et al, 2002).
    f) A 25-year-old man who was taking lamotrigine 400 mg/day (6 mg/kg/day; plasma concentration 12.1 mcg/mL) for generalized tonic-clonic seizures developed oculogyric crisis (2 to 3 episodes of sustained upward deviation of both eyes) after inadvertently ingesting 1600 mg/day (23.5 mg/kg/day; plasma concentration 16.5 mcg/mL) of lamotrigine during a 24-hour period. His symptoms resolved after his lamotrigine dose was decreased to 6 mg/kg/day (plasma concentration 7 mcg/mL) (Veerapandiyan et al, 2011).
    g) A serum level of 25.2 mcg/mL was reported on initial presentation to the emergency department of a 29-year-old man who intentionally ingested an unknown number of 200 milligram lamotrigine tablets and ethanol (191 mg/dL; normal less than 10 mg/dL). This value most likely does not represent the peak lamotrigine concentration since it is uncertain when the ingestion occurred; it is estimated they it may have been up to 12 hours prior to presentation (Schwartz & Geller, 2007).
    h) Serum level of 52 micrograms/milliliter (a ten-fold increase over normal therapeutic values) was reported four hours following an acute ingestion of greater than 4000 milligrams lamotrigine in an adult woman (Prod Info Lamictal(R), lamotrigine tablets, 2000).
    i) STATUS EPILEPTICUS- Approximately 1 to 2 hours after ingesting 4.1 grams of lamotrigine, a 42-year-old woman with a history of focal seizures developed secondarily generalized status epilepticus. The lamotrigine concentrations were: 47.4 micrograms/milliliter (on admission), 15.8 micrograms/milliliter (on day 2), 3.2 micrograms/milliliter (on day 3), and 0.8 micrograms/milliliter (on day 4) (Dinnerstein et al, 2007).
    j) Harchelroad et al (1994) reported serum lamotrigine levels of 52.5 micrograms/milliliter at 28 hours post-ingestion in a 33-year-old woman who ingested 1500 to 2000 milligrams (Harchelroad et al, 1994).
    k) Lamotrigine serum levels at 3 and 17 hours post-ingestion were 17.4 and 6.4 micrograms/milliliter respectively following an acute overdose of 1350 milligrams in a 26-year-old man (Buckley et al, 1993).
    l) Serum lamotrigine level of 19.3 micrograms/milliliter was associated with marked sedation, fatigue, and decreased cognition in a 17-year-old girl (Kaufman & Gerner, 1998).
    m) Peak serum lamotrigine concentration of 35.8 milligrams/liter was reported following a stated overdose of 4500 milligrams (absorbed estimated dose of 2900 milligrams) in a 32-year-old woman. The patient survived, with clinical manifestations of rotary nystagmus and marked ataxia which improved over 2 days (O'Donnell & Bateman, 2000).
    n) Following ingestion of an unknown quantity of lamotrigine, a serum level of 62.4 micrograms/milliliter was reported at 24 hours after admission in a 23-year-old man. The overdose was associated with severe choreatic dyskinesia which improved following an injection of lorazepam (Miller et al, 2001).
    o) A lamotrigine serum level of 32 milligrams/liter was reported following deliberate ingestion of an unknown quantity of lamotrigine and valproic acid. Encephalopathy resulted, with improvement correlating with declining lamotrigine serum levels. It was suggested that valproic acid may have potentiated toxicity by decreasing hepatic clearance of lamotrigine (Sbei & Campellone, 2001).
    p) Two patients developed reversible downbeat nystagmus as a result of lamotrigine toxicity. Both patients had intractable epilepsy and developed oscillopsia and incoordination while taking lamotrigine (patient one took 100 milligrams four times daily; patient two took 200 milligrams every morning, 100 milligrams at noon, 200 milligrams every evening) in combination with other anticonvulsants (eg; gabapentin or valproic acid). Serum lamotrigine levels peaked at 19.9 micrograms/milliliter (therapeutic range 1-14 micrograms/milliliter) in patient one and peaked at 27 micrograms/milliliter (therapeutic range 4-18 micrograms/milliliter using a different laboratory) in patient two (AlKawi et al, 2005).
    2) PEDIATRIC
    a) OCULOGYRIC CRISIS: Three children with no history of movement disorders developed oculogyric crisis (1 to 20 episodes per day; 2 seconds to several hours duration) after taking increasing doses of lamotrigine. All patients recovered following the reduction of lamotrigine doses (Veerapandiyan et al, 2011).
    1) A 10-year-old girl who was taking lamotrigine 22 mg/kg/day (plasma concentration 6.7 mcg/mL) presented with oculogyric crisis (a few to 20 episodes/day for 3 days; duration of each episode, 2 to 3 hours; associated symptoms: tremulousness, irritability) about a week after her lamotrigine dose was increased to 25 mg/kg/day (plasma concentration 15.8 mcg/mL). MRI revealed diffuse cerebral and cerebellar atrophy. Her symptoms resolved after lamotrigine dose was decreased to 20 mg/kg/day (plasma concentration 8.5 mcg/mL).
    2) A 3-year-old boy who was taking lamotrigine 2 mg/kg/day (plasma concentration 2.1 mcg/mL) developed oculogyric crisis (1 episode/day for 1 month; duration of each episode, few seconds to 1 minute) after his lamotrigine dose was increased to 7.8 mg/kg/day. His symptoms resolved after decreasing the lamotrigine dose.
    3) An 11-year-old boy who was taking lamotrigine 5.5 mg/kg/day for absence seizures, developed recurrent episodes of oculogyric crisis (5 to 7 episodes/day for 3 months; duration of each episode, 2 to 4 seconds) after his lamotrigine dose was increased to 7 mg/kg/day (plasma concentration 9.3 mcg/mL). His symptoms resolved after his lamotrigine dose was decreased to 4 mg/kg/day.
    b) A plasma lamotrigine level of 35 milligrams/liter (above 30 milligrams/liter in several previously stored samples) was reported in a 12-day-old infant after receiving an unknown amount of lamotrigine. He experienced tonic-clonic seizures, and other neurological symptoms. Following symptomatic treatment, he gradually returned to normal over the next 5 days (Willis et al, 2007).
    c) One study reported that plasma levels equal to or greater than 8 micrograms/milliliter have been well tolerated by pediatric patients (Uvebrant & Bauziene, 1994).
    d) Plasma lamotrigine level of 25.3 micrograms/milliliter (elevated above adult therapeutic levels) was reported in a 3-year-old after ingesting forty-six 25 milligram tablets (1.150 grams) of lamotrigine. Following supportive treatment, she recovered without further sequelae (Zidd & Hack, 2004).
    e) Peak plasma level of 3.8 milligrams/liter was reported in a 2-year-old boy after an ingestion of 800 milligrams (Briassoulis et al, 1998).

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) ANIMAL DATA
    1) LD50- (ORAL)MOUSE:
    a) 245 mg/kg (RTECS, 2001)
    2) LD50- (ORAL)RAT:
    a) 205 mg/kg (RTECS, 2001)

Pharmacologic Mechanism

    A) Lamotrigine is an anticonvulsant whose mechanisms of action are not known. One proposed mechanism is that lamotrigine inhibits sodium channels, resulting in neuronal membrane stabilization and control of excitatory neurotransmitter release (Prod Info LAMICTAL(R) oral tablets, chewable dispersible oral tablets, 2007). Lamotrigine also inhibits folate metabolism, an action shared by some other antiepileptics but not confirmed as a mechanism of anticonvulsant effects (Yuen, 1991).
    B) Lamotrigine appears to block voltage-dependent sodium channels, which stabilizes the presynaptic membrane and prevents release of glutamate, an excitatory neurotransmitter, and shows anticonvulsant activity in a wide range of animal models (Cohen et al, 1987; O'Donohoe, 1991; Porter, 1989).
    1) In vitro animal studies have shown lamotrigine to inhibit veratrine-induced glutamate and aspartate release in brain tissue, with no effect on potassium-induced amino acid release. This suggests that the drug acts at voltage-sensitive sodium channels to stabilize neuronal membranes and inhibit neurotransmitter release, namely glutamate (Leach et al, 1986).
    2) In animal models, plasma concentrations of approximately 3 micrograms/milliliter are of similar protective efficacy as therapeutic concentrations of phenytoin and carbamazepine in the maximal electroshock and maximal pentylenetetrazol tests. It also reduces or eliminates the afterdischarge induced by focal stimulation of the cortex or hippocampus in animal models (Wheatley & Miller, 1989). While it does not block or reduce the rate of development of kindling, it does decrease the number of kindled responses and the duration of kindled seizures (O'Donnell & Miller, 1991). Lamotrigine is not effective in threshold tests (Jawad et al, 1989; Leach et al, 1991; Peck, 1991a).
    3) Further evidence that lamotrigine inhibits glutamate release is shown in the rat model, in which kainic acid neurotoxicity, mediated by glutamate release, is inhibited, whereas quinolinic acid and ibotenic acid neurotoxicity, mediated by N-methyl-d-aspartate receptor excitation, is not (McGeer & Zhu, 1990).
    C) Single doses of lamotrigine cause an acute reduction in or abolition of photosensitivity in patients with epilepsy, and reduces the incidence of interictal EEG spikes, hallmarks of epileptic activity (Binnie et al, 1986; Jawad et al, 1986).

Physical Characteristics

    A) Lamotrigine is a white to pale cream-colored powder that is very slightly soluble in water (0.17 mg/mL at 25 degrees C) and slightly soluble in 0.1 molar hydrochloride (4.1 mg/mL at 25 degrees C) (Prod Info LAMICTAL XR oral extended-release tablets, 2009).
    B) Lamotrigine

Molecular Weight

    A) 256.09 (Prod Info LAMICTAL XR oral extended-release tablets, 2009)

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) AlKawi A, Kattah JC, & Wyman K: Downbeat nystagmus as a result of lamotrigine toxicity. Epilepsy Research 2005; 63:85-88.
    3) Algahtani HA, Aldarmahi AA, Al-Rabia MW, et al: Generalized myoclonus and spasticity induced by lamotrigine toxicity: a case report and literature review. Clin Neuropharmacol 2014; 37(2):52-54.
    4) American College of Medical Toxicology : ACMT Position Statement: Interim Guidance for the Use of Lipid Resuscitation Therapy. J Med Toxicol 2011; 7(1):81-82.
    5) American College of Medical Toxicology: ACMT position statement: guidance for the use of intravenous lipid emulsion. J Med Toxicol 2016; Epub:Epub-.
    6) Anon: Lamotrigine pregnancy registry interim report 1September 1992 through March 2005. GlaxoSmithKline, Charles River Clinical Services, Wilmington, NC. 800-336-2176, July 2005.
    7) Besag FMC, Berry DJ, & Pool F: Carbamazepine toxicity with lamotrigine: Pharmacokinetic or pharmacodynamic interaction?. Epilepsia 1998; 39:183-187.
    8) Besag FMC, Berry DJ, & Pool F: Carbamazepine toxicity with lamotrigine: pharmacokinetic or pharmacodynamic interaction?. Epilepsia 1998a; 39:183-187.
    9) Betts T, Goodwin G, & Withers RM: Human safety of lamotrigine. Epilepsia 1991; 32(Suppl 2):S17-21.
    10) Binnie CD, van Emde Boas W, & Kasteleijn-Nolste-Trenite DG: Acute effects of lamotrigine (BW430C) in persons with epilepsy. Epilepsia 1986; 27:248-254.
    11) Birnbaum AK, Kriel RL, & Im Y: Relative bioavailability of lamotrigine chewable dispersible tablets administered rectally. Pharmacother 2001; 21:158-162.
    12) Briassoulis G, Kalabalikis P, & Tamiolaki M: Lamotrigine childhood overdose. Pediatr Neurol 1998; 19:239-242.
    13) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    14) Brown CV, Rhee P, Chan L, et al: Preventing renal failure in patients with rhabdomyolysis: do bicarbonate and mannitol make a difference?. J Trauma 2004; 56(6):1191-1196.
    15) Buckley NA, Whyte IM, & Dawson AH: Self-poisoning with lamotrigine. Lancet 1993; 342:1552-1553.
    16) Camp NE: Drug- and toxin-induced Rhabdomyolysis. J Emerg Nurs 2009; 35(5):481-482.
    17) Castanares-Zapatero D, Wittebole X, Huberlant V, et al: Lipid Emulsion as Rescue Therapy in Lamotrigine Overdose. J Emerg Med 2011; Epub:Epub.
    18) Chaffin JJ & Davis SM: Suspected lamotrigine-induced toxic epidermal necrolysis. Ann Pharmacother 1997; 31:720-723.
    19) 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.
    20) Chattergoon DS, McGuigan MA, & Koren G: Multiorgan dysfunction and disseminated intravascular coagulation in children receiving lamotrigine and valproic acid. Neurology 1997; 49:1442-1444.
    21) 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.
    22) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    23) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    24) Close BR & Banks CJ: Seizures secondary to lamotrigine toxicity in a two-year-old. Ann Pharmacother 2010; 44(6):1112-1115.
    25) Cohen AF, Land GS, & Breimer DD: Lamotrigine, a new anticonvulsant: pharmacokinetics in normal humans. Clin Pharmacol Ther 1987; 42:535-541.
    26) Criddle LM: Rhabdomyolysis. Pathophysiology, recognition, and management. Crit Care Nurse 2003; 23(6):14-22, 24-26, 28.
    27) Cunnington MC, Weil JG, Messenheimer JA, et al: Final results from 18 years of the International Lamotrigine Pregnancy Registry. Neurology 2011; 76(21):1817-1823.
    28) Daana M, Nevo Y, Tenenbaum A, et al: Lamotrigine overdose in a child. J Child Neurol 2007; 22(5):642-644.
    29) Dagtekin O, Marcus H, Muller C, et al: Lipid therapy for serotonin syndrome after intoxication with venlafaxine, lamotrigine and diazepam. Minerva Anestesiol 2011; 77(1):93-95.
    30) Depot M, Powell JR, & Messenheimer JA: Kinetic effects of multiple oral doses of acetaminophen on a single oral dose of lamotrigine. Clin Pharmacol Ther 1990; 48:346-355.
    31) Dinnerstein E, Jobst BC, & Williamson PD: Lamotrigine intoxication provoking status epilepticus in an adult with localization-related epilepsy. Arch Neurol 2007; 64(9):1344-1346.
    32) Dolk H, Jentink J, Loane M, et al: Does lamotrigine use in pregnancy increase orofacial cleft risk relative to other malformations?. Neurology 2008; 71(10):714-722.
    33) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    34) Erdman AR & Dart RC: Rhabdomyolysis. In: Dart RC, Caravati EM, McGuigan MA, et al, eds. Medical Toxicology, 3rd ed. Lippincott Williams & Wilkins, Philadelphia, PA, 2004, pp 123-127.
    35) Eren Cevik S, Tasyurek T, & Guneysel O: Intralipid emulsion treatment as an antidote in lipophilic drug intoxications. Am J Emerg Med 2014; 32(9):1103-1108.
    36) Esfahani FE & Dasheiff RM: Anemia associated with lamotrigine. Neurology 1997; 49:306-307.
    37) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    38) Fillastre JP, Taburet AM, & Fialaire A: Pharmacokinetics of lamotrigine in patients with renal impairment: influence of haemodialysis. Drugs Exptl Clin Res 1993a; 19(1):25-32.
    39) Fillastre JP, Taburet AM, Fialaire A, et al: Pharmacokinetics of lamotrigine in patients with renal impairment: Influence of haemodialysis.. Drugs Exp Clin Res 1993; XIX(1):25-32.
    40) Fotopoulou C, Kretz R, Bauer S, et al: Prospectively assessed changes in lamotrigine-concentration in women with epilepsy during pregnancy, lactation and the neonatal period. Epilepsy Res 2009; 85(1):60-64.
    41) French LK, McKeown NJ, Hendrickson RG, et al: Complete heart block and death following lamotrigine overdose. Clinical Toxicol (Phila) 2011; 49(4):330-333.
    42) Froscher W, Keller F, & Vogt H: Prospective study on concentration-efficacy and concentration-toxicity: correlations with lamotrigine serum levels. Epileptic Disord 2002; 1:49-55.
    43) Gentile S: Lamotrigine in pregnancy and lactation. Arch Womens Ment Health 2005; 8(1):57-58.
    44) Geraci MJ, Peele J, McCoy SL, et al: Phencyclidine false positive induced by lamotrigine (Lamictal(R)) on a rapid urine toxicology screen. Int J Emerg Med 2010; 3(4):327-331.
    45) Goa KL, Ross SR, & Chrisp P: Lamotrigine: A review of its pharmacological properties and clinical efficacy in epilepsy. Drugs 1993; 46(1):152-176.
    46) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    47) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    48) Guerrini R, Belmonte A, & Parmeggiani L: Myoclonic status epilepticus following high-dosage lamotrigine therapy. Brain Develop 1999; 21:420-424.
    49) Harchelroad F, Lang D, & Valeriano J: Lamotrigine overdose (Abstract). Vet Hum Tox 1994; 36(4):372.
    50) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    51) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    52) Hennessy MJ & Wiles CM: Lamotrigine encephalopathy (letter). Lancet 1996; 347:974-975.
    53) Homsi E, Barreiro MF, Orlando JM, et al: Prophylaxis of acute renal failure in patients with rhabdomyolysis. Ren Fail 1997; 19(2):283-288.
    54) Huerta-Alardin AL, Varon J, & Marik PE: Bench-to-bedside review: Rhabdomyolysis -- an overview for clinicians. Crit Care 2005; 9(2):158-169.
    55) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    56) Ishioka M, Yasui-Furukori N, Hashimoto K, et al: Neuroleptic malignant syndrome induced by lamotrigine. Clin Neuropharmacol 2013; 36(4):131-132.
    57) Jawad S, Oxley J, & Yuen WC: The effect of lamotrigine, a novel anticonvulsant, on interictal spikes in patients with epilepsy. Br J Clin Pharmacol 1986; 22:191-193.
    58) Jawad S, Richens A, & Goodwin G: Controlled trial of lamotrigine (Lamictal) for refractory partial seizures. Epilepsia 1989; 30:356-363.
    59) Kanner AM & Frey M: Adding valproate to lamotrigine: a study of their pharmacokinetic interaction. Neurol 2000; 55:588-591.
    60) Kaufman KR & Gerner R: Lamotrigine toxicity secondary to sertraline. Seizure 1998; 7:163-165.
    61) Kaufman KR & Gerner R: Lamotrigine toxicity secondary to sertraline. Seizure 1998a; 7:163-165.
    62) Keranen T , Sorri A , Moilanen E , et al: Effects of charcoal on the absorption and elimination of the antiepileptic drugs lamotrigine and oxcarbazepine. Arzneimittelforschung 2010; 60(7):421-426.
    63) 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.
    64) Lavonas EJ, Drennan IR, Gabrielli A, et al: Part 10: Special Circumstances of Resuscitation: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S501-S518.
    65) Leach MJ, Baxter MG, & Critchley MAE: Neurochemical and behavioral aspects of lamotrigine. Epilepsia 1991; 32(Suppl 2):S4-S8.
    66) Leach MJ, Marden CM, & Miller AA: Pharmacological studies on lamotrigine, a novel potential antiepileptic drug: II. Neurochemical studies on the mechanism of action. Epilepsia 1986; 27(5):490-497.
    67) Liporace J, Kao A, & D'Abreu A: Concerns regarding lamotrigine and breast-feeding. Epilepsy Behav 2004; 5(1):102-105.
    68) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    69) Lofton AL & Klein-Schwartz W: Evaluation of lamotrigine toxicity reported to poison centers. Ann Pharmacother 2004; 38:1811-1815.
    70) Loiseau P: Treatment of concomitant illneses in patients receiving anticonvulsants. Drug interactions of clinical significance. Drug Safety 1998; 19:495-510.
    71) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    72) Marraffa JM & Guharoy R: Lymphadenopathy secondary to lamotrigine (Lamictal). Vet Hum Toxicol 2002; 44(5):276-277.
    73) Matsuo F, Bergen D, & Faught E: Placebo-controlled study of the efficacy and safety of lamotrigine in patients with partial seizures. Neurology 1993; 43(11):2284-2291.
    74) McGeer EG & Zhu SG: Lamotrigine protects against kainate but not ibotenate lesions in rat striatum. Neuroscience Letters 1990; 112:348-351.
    75) Meador KJ, Baker GA, Browning N, et al: Effects of fetal antiepileptic drug exposure: outcomes at age 4.5 years. Neurology 2012; 78(16):1207-1214.
    76) Messenheimer J, Mullens EL, & Giorgi L: Safety review of adult clinical trial experience with lamotrigine. Drug Safety 1998; 18:281-296.
    77) Mewasingh L, Aylett S, & Kirkham F: Hyponatraemia associated with lamotrigine in cranial diabetes insipidus. Lancet 2000; 356:656.
    78) Mikati MA, Schachter SC, Schomer DL, et al: Long-term tolerability, pharmacokinetic and preliminary efficacy study of lamotrigine in patients with resistant partial seizures. Clin Neuropharmacol 1989; 12:312-321.
    79) Miller M, Patel M, & Tai W: Choreatic dyskinesia following lamotrigine overdose (abstract). J Toxicol Clin Toxicol 2001; 39:499.
    80) Miller MA & Levsky ME: Choreiform dyskinesia following isolated lamotrigine overdose. J Child Neurol 2008; 23(2):243-243.
    81) Miller RD, Sohn YJ, & Matteo RS: Enhancement of d-tubocurarine neuromuscular blockade by diuretics in man. Anesthesiology 1976; 45:442-445.
    82) Moeller KE, Wei L, Jewell AD, et al: Acute hepatotoxicity associated with lamotrigine. Am J Psychiatry 2008; 165(4):539-540.
    83) Moore PW, Donovan JW, Burkhart KK, et al: A case series of patients with lamotrigine toxicity at one center from 2003 to 2012. Clin Toxicol (Phila) 2013; 51(7):545-549.
    84) Mylonakis E, Vittorio CC, & Hollik DA: Lamotrigine overdose presenting as anticonvulsant hypersensitivity syndrome. Ann Pharmacother 1999; 33:557-559.
    85) 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.
    86) Newport DJ, Pennell PB, Calamaras MR, et al: Lamotrigine in breast milk and nursing infants: determination of exposure. Pediatrics 2008; 122(1):e223-e231.
    87) Nicholson RJ, Kelly KP, & Grant IS: Leukopenia associated with lamotrigine. Br Med J 1995; 310:504.
    88) Nogar JN, Minns AB, Savaser DJ, et al: Severe sodium channel blockade and cardiovascular collapse due to a massive lamotrigine overdose. Clin Toxicol (Phila) 2011; 49(9):854-857.
    89) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    90) Nordmo E, Aronsen L, Wasland K, et al: Severe apnea in an infant exposed to lamotrigine in breast milk. Ann Pharmacother 2009; 43(11):1893-1897.
    91) Nwogbe B, Ferie J, Smith H, et al: Significant lamotrigine overdose associated with acute pancreatitis. J R Soc Med 2009; 102(3):118-119.
    92) O'Donnell J & Bateman DN: Lamotrigine overdose in an adult. Clin Toxicol 2000; 38:659-660.
    93) O'Donnell RA & Miller AA: The effect of lamotrigine upon development of cortical kindled seizures in the rat. Neuropharmacology 1991; 30:253-258.
    94) O'Donohoe NV: Use of antiepileptic drugs in childhood epilepsy. Arch Dis Child 1991; 66:1173-1179.
    95) Oh SY , Kim JS , Lee YH , et al: Downbeat, positional, and perverted head-shaking nystagmus associated with lamotrigine toxicity. J Clin Neurol 2006; 2(4):283-285.
    96) Ohman I & Vitols S: Lamotrigine in pregnancy: pharmacokinetics during delivery, in the neonate, and during lactation. Epilepsia 2000; 41(6):709-713.
    97) Olson KR: Comprehensive Evaluation and Treatment of Poisoning and Overdose. In: Olson KR (Ed): Poisoning and Drug Overdose, 2nd ed, Appleton & Lange, Norwalk, CT, 1994.
    98) Padmanabhan R, Abdulrazzaq YM, Bastaki SMA, et al: Experimental studies on reproductive toxicologic effects of lamotrigine in mice. Birth Defects Res 2003; 68 (part B):428-438.
    99) Page RL, O'Neil MG, & Yarbrough DR: Fatal toxic epidermal necrolysis related to lamotrigine administration. Pharmacother 1998; 18:392-398.
    100) Page-Sharp M, Kristensen JH, Hackett LP, et al: Transfer of lamotrigine into breast milk. Ann Pharmacother 2006; 40(7-8):1470-1471.
    101) Pathak P & McLachlan RS: Drug-induced pseudolymphoma secondary to lamotrigine. Neurology 1998; 50:1509-1510.
    102) Paulzen M, Lammertz SE, Veselinovic T, et al: Lamotrigine in pregnancy - therapeutic drug monitoring in maternal blood, amniotic fluid, and cord blood. Int Clin Psychopharmacol 2015; 30(5):249-254.
    103) 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.
    104) Peck AW: Clinical pharmacology of lamotrigine. Epilepsia 1991; 32(suppl 2):S9-S12.
    105) Peck AW: Clinical pharmacology of lamotrigine. Epilepsia 1991a; 32(Suppl2):S9-S12.
    106) Polderman KH: Acute renal failure and rhabdomyolysis. Int J Artif Organs 2004; 27(12):1030-1033.
    107) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    108) Porter RJ: Mechanisms of action of new antiepileptic drugs. Epilepsia 1989; 30(Suppl 1):S29-S34.
    109) Posner J, Cohen AF, & Land G: The pharmacokinetics of lamotrigine (BW430C) in healthy subjects with unconjugated hyperbilirubinaemia (Gilbert's Syndrome). Br J Clin Pharmacol 1989; 28:117-120.
    110) Pricone MG, King CV, & Drummer OH: Postmortem investigation of lamotrigine concentrations. J Forensic Sci 2000; 45:11-15.
    111) Product Information: LAMICTAL ODT(R) oral disintegrating tablets, lamotrigine oral disintegrating tablets. GlaxoSmithKline (per manufacturer), Research Triangle Park, NC, 2014.
    112) Product Information: LAMICTAL XR oral extended-release tablets, lamotrigine oral extended-release tablets. GlaxoSmithKline, Research Triangle Park, NC, 2009.
    113) Product Information: LAMICTAL chewable dispersible oral tablets, oral tablets, orally disintegrating tablets, lamotrigine chewable dispersible oral tablets, oral tablets, orally disintegrating tablets. GlaxoSmithKline, Research Triangle Park, NC, 2009.
    114) Product Information: LAMICTAL(R) XR(TM) oral extended release tablets, lamotrigine oral extended release tablets. GlaxoSmithKline (per FDA), Research Triangle Park, NC, 2013.
    115) Product Information: LAMICTAL(R) XR(TM) oral extended-release tablets, lamotrigine oral extended-release tablets. GlaxoSmithKline (per Manufacturer), Research Triangle Park, NC, 2011a.
    116) Product Information: LAMICTAL(R) XR(TM) oral extended-release tablets, lamotrigine oral extended-release tablets. GlaxoSmithKline, Research Triangle Park, NC, 2011.
    117) Product Information: LAMICTAL(R) oral tablets, chewable dispersible oral tablets, lamotrigine oral tablets chewable dispersible oral tablets. GlaxoSmithKline, Research Triangle Park, NC, 2007.
    118) Product Information: LAMICTAL(R) oral tablets, oral chewable dispersible tablets, lamotrigine oral tablets, oral chewable dispersible tablets. GlaxoSmithKline (per manufacturer), Research Triangle Park, NC, 2014.
    119) Product Information: Lamictal(R), lamotrigine tablets. Glaxo Wellcome Co, Research Triangle Park, NC, 2000.
    120) Product Information: Lidocaine HCl intravenous injection solution, lidocaine HCl intravenous injection solution. Hospira (per manufacturer), Lake Forest, IL, 2006.
    121) Product Information: MANNITOL intravenous injection, mannitol intravenous injection. Hospira, Inc. (per DailyMed), Lake Forest, IL, 2009.
    122) Product Information: Mannitol intravenous injection, mannitol intravenous injection. American Regent, Inc. (per DailyMed), Shirley, NY, 2009.
    123) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    124) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    125) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    126) Product Information: mannitol IV injection, urologic irrigation, mannitol IV injection, urologic irrigation. Abraxis Pharmaceutical Products, Schaumburg, IL, 2006.
    127) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    128) Pulik M, Lionnet F, & Genet P: Successful treatment of lamotrigine-induced erythroblastopenic crisis with folinic acid. Neurol 2000; 55:1235-1236.
    129) Queiroz MEC, Carrilho E, & Carvalho D: Comparison of high-resolution gas chromatography and high-performance liquid chromatography for simultaneous determination of lamotrigine and carbamazepine in plasma. Chromatographia 2001; 53:485-489.
    130) 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.
    131) Rambeck B, Kurlemann G, & Stodieck SRG: Concentrations of lamotrigine in a mother on lamotrigine treatment and her newborn child. Eur J Clin Pharmacol 1997; 51:481-484.
    132) Rambeck B, Kurlemann G, Stodieck SRG, et al: Concentrations of lamotrigine in a mother on lamotrigine treatment and her newborn child. Eur J Clin Pharmacol 1997a; 51:481-484.
    133) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    134) Reutens DC, Duncan JS, & Patsalos PN: Disabling tremor after lamotrigine with sodium valproate. Lancet 1993; 342:185-186.
    135) Richens A: Safety of lamotrigine.. Epilepsia 1994; 35 Suppl 5:S37-S40.
    136) 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.
    137) Sachs B, Ronnau AC, & Ruzicka T: Lamotrigine and toxic epidermal necrolysis (letter). Lancet 1996; 348:1597.
    138) Sailstad JM & Findlay JWA: Immunofluorometric assay for lamotrigine in human plasma. Ther Drug Monit 1991; 13:433-442.
    139) Saito S, Shioda K, & Nisijima K: Anger with murderous impulse induced by lamotrigine. Gen Hosp Psychiatry 2014; 36(4):451.e1-451.e2.
    140) Sarris BM & Wong JG: Multisystem hypersensitivity reaction to lamotrigine. Neurology 1999; 53:1367.
    141) Sbei M & Campellone JV: Stupor from lamotrigine toxicity. Epilepsia 2001; 42:1082-1083.
    142) Schaub JEM, Williamson PJ, & Barnes EW: Multisystem adverse reaction to lamotrigine. Lancet 1994; 344:481.
    143) Schlienger RG, Knowles SR, & Shear NH: Lamotrigine-associated anticonvulsant hypersensitivity syndrome. Neurology 1998; 51:1172-1175.
    144) Schwartz MD & Geller RJ: Seizures and altered mental status after lamotrigine overdose. Ther Drug Monit 2007; 29(6):843-844.
    145) 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.
    146) Sinz MW & Remmel RP: Isolation and characterization of a novel quaternary ammonium-linked glucuronide of lamotrigine. Drug Metab Dispos 1991; 19:149-153.
    147) 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.
    148) Stopforth J: Overdose with gabapentin and lamotrigine (letter). S Afr Med J 1997; 87:1388.
    149) Strimel WJ , Woodruff A , Cheung P , et al: Brugada-like electrocardiographic pattern induced by lamotrigine toxicity. Clin Neuropharmacol 2010; 33(5):265-267.
    150) Su-Yin AN, Tai WW, & Olson KR: Lamotrigine-associated reversible severe hepatitis: a case report. J Med Toxicol 2008; 4(4):258-260.
    151) Tomson T & Ohman I: Lamotrigine in pregnancy and lactation: a case report. Epilepsia 1997; 38:1039-1041.
    152) U.S. Food and Drug Administration: FDA Safety Communication: Aseptic meningitis associated with the use of Lamictal (lamotrigine). U.S. Food and Drug Administration. Rockville, MD. 2010. Available from URL: http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm221847.htm. As accessed 2010-08-12.
    153) US Food and Drug Administration: Information for Healthcare Professionals: lamotrigine (marketed as LAMICTAL(R)). US Food and Drug Administration. Rockville, MD. 2006. Available from URL: http://www.fda.gov/cder/drug/InfoSheets/HCP/lamotrigineHCP.htm.
    154) Uvebrant P & Bauziene R: Intractable epilepsy in children. The efficacy of lamotrigine treatment, including non-seizure-related benefits. Neuroped 1994; 25:284-289.
    155) Vajda FJ, Graham J, Roten A, et al: Teratogenicity of the newer antiepileptic drugs--the Australian experience. J Clin Neurosci 2012; 19(1):57-59.
    156) Vanden Hoek TL, Morrison LJ, Shuster M, et al: Part 12: cardiac arrest in special situations: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2010; 122(18 Suppl 3):S829-S861.
    157) Vanholder R, Sever MS, Erek E, et al: Rhabdomyolysis. J Am Soc Nephrol 2000; 11(8):1553-1561.
    158) Varghese SP, Haith LR, Patton ML, et al: Lamotrigine-induced toxic epidermal necrolysis in three patients treated for bipolar disorder. Pharmacotherapy 2006; 26(5):699-704.
    159) Veerapandiyan A, Gallentine WB, Winchester SA, et al: Oculogyric crises secondary to lamotrigine overdosage. Epilepsia 2011; 52(3):e4-e6.
    160) Verma A, Miller P, & Carwile ST: Lamotrigine-induced blepharospasm. Pharmacother 1999; 19:877-880.
    161) Vukelic D, Bozinovic D, & Tesovic G: Lamotrigine and toxic epidermal necrolysis (letter). Dermatology 1997; 195:307.
    162) Wadelius M, Karlsson T, & Wadelius C: Lamotrigine and toxic epidermal necrolysis. Lancet 1996; 348:1041.
    163) Wakil L, Epperson CN, Gonzalez J, et al: Neonatal outcomes with the use of lamotrigine for bipolar disorder in pregnancy and breastfeeding: a case series and review of the literature. Psychopharmacol Bull 2009; 42(3):91-98.
    164) Walter LA & Catenacci MH: Rhabdomyolysis. Hosp Physician 2008; 44(1):25-31.
    165) Wheatley PL & Miller AA: Effects of lamotrigine on electrically induced afterdischarge duration in anaesthetised rat, dog, and marmoset. Epilepsia 1989; 30(1):34-40.
    166) Willis T, Roper H, & Rabb L: Lamotrigine poisoning presenting as seizures: A case of deliberate poisoning. Child Abuse Negl 2007; 31(1):85-88.
    167) Wilton LV, Pearce GL, Martin RM, et al: The outcomes of pregnancy in women exposed to newly marketed drugs in general practice in England. Br J Obstet Gynaecol 1998; 105:882-889.
    168) Wootton R, Soul-Lawton J, & Rolan PE: Comparison of the pharmacokinetics of lamotrigine in patients with chronic renal failure and healthy volunteers. Br J Clin Pharmacol 1997; 43:23-27.
    169) Yuen AWC & Peck AW: Lamotrigine pharmacokinetics: oral and i.v. infusion in man. Abstract. Br J Clin Pharm 1988; 26:242P.
    170) Yuen AWC: Lamotrigine. New Antiepileptic Drugs (Epilepsy Res. Suppl 3) , Elsevier Science Publishers B.V, New York, NY, 1991, pp 115-123.
    171) Zidd AG & Hack JB: Pediatric ingestion of lamotrigine. Pediatr Neurol 2004; 31(1):71-72.
    172) Zimmerman JL & Shen MC: Rhabdomyolysis. Chest 2013; 144(3):1058-1065.
    173) 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.
    174) de Camargo OAK & Bode H: Agranulocytosis associated with lamotrigine. Br Med J 1999; 318:1179.