Propafenone Hydrochloride 150 MG Oral Tablet

DRUG INTERACTIONS

7 Inhibitors of CYP2D6, 1A2, and 3A4 increase propafenone exposure.

( 7.1 ) Propafenone may increase digoxin or warfarin levels.

( 7.2 , 7.3 ) Orlistat may reduce propafenone exposure.

Taper orlistat withdrawal.

( 7.4 ) Lidocaine may increase central nervous system side effects.

( 7.6 ) 7.1 CYP2D6 and CYP3A4 Inhibitors Drugs that inhibit CYP2D6 (such as desipramine, paroxetine, ritonavir, or sertraline) and CYP3A4 (such as ketoconazole, ritonavir, saquinavir, erythromycin, or grapefruit juice) can be expected to cause increased plasma levels of propafenone.

The combination of CYP3A4 inhibition and either CYP2D6 deficiency or CYP2D6 inhibition with administration of propafenone may increase the risk of adverse reactions, including proarrhythmia.

Therefore, simultaneous use of propafenone hydrochloride with both a CYP2D6 inhibitor and a CYP3A4 inhibitor should be avoided [see Warnings and Precautions (5.4) and Dosage and Administration (2) ].

A m iodarone: Concomitant administration of propafenone and amiodarone can affect conduction and repolarization and is not recommended.

Cimetidine: Concomitant administration of propafenone immediate release tablets and cimetidine in 12 healthy subjects resulted in a 20% increase in steady-state plasma concentrations of propafenone.

Fluoxetine: Concomitant administration of propafenone and fluoxetine in extensive metabolizers increased the S-propafenone C max and AUC by 39% and 50% and the R propafenone C max and AUC by 71% and 50%.

Quinidine: Small doses of quinidine completely inhibit the CYP2D6 hydroxylation metabolic pathway, making all patients, in effect, slow metabolizers [see Clinical Pharmacology (12) ] .

Concomitant administration of quinidine (50 mg three times daily) with 150 mg immediate release propafenone three times daily decreased the clearance of propafenone by 60% in extensive metabolizers, making them slow metabolizers.

Steady-state plasma concentrations more than doubled for propafenone, and decreased 50% for 5-OH-propafenone.

A 100 mg dose of quinidine tripled steady state concentrations of propafenone.

Avoid concomitant use of propafenone and quinidine.

Rifampin: Concomitant administration of rifampin and propafenone in extensive metabolizers decreased the plasma concentrations of propafenone by 67% with a corresponding decrease of 5-OH-propafenone by 65%.

The concentrations of norpropafenone increased by 30%.

In slow metabolizers, there was a 50% decrease in propafenone plasma concentrations and increased the AUC and C max of norpropafenone by 74% and 20%, respectively.

Urinary excretion of propafenone and its metabolites decreased significantly.

Similar results were noted in elderly patients: Both the AUC and C max propafenone decreased by 84%, with a corresponding decrease in AUC and C max of 5-OH-propafenone by 69% and 57%.

7.2 Digoxin Concomitant use of propafenone and digoxin increased steady-state serum digoxin exposure (AUC) in patients by 60% to 270%, and decreased the clearance of digoxin by 31% to 67%.

Monitor plasma digoxin levels of patients receiving propafenone and adjust digoxin dosage as needed.

7.3 Warfarin The concomitant administration of propafenone and warfarin increased warfarin plasma concentrations at steady state by 39% in healthy volunteers and prolonged the prothrombin time (PT) in patients taking warfarin.

Adjust the warfarin dose as needed by monitoring INR (international normalized ratio).

7.4 Orlistat Orlistat may limit the fraction of propafenone available for absorption.

In post marketing reports, abrupt cessation of orlistat in patients stabilized on propafenone has resulted in severe adverse events including convulsions, atrioventricular block and acute circulatory failure.

7.5 Beta-Antagonists Concomitant use of propafenone and propranolol in healthy subjects increased propranolol plasma concentrations at steady state by 113%.

In 4 patients, administration of metoprolol with propafenone increased the metoprolol plasma concentrations at steady state by 100% to 400%.

The pharmacokinetics of propafenone was not affected by the coadministration of either propranolol or metoprolol.

In clinical trials using propafenone immediate release tablets, patients who were receiving beta-blockers concurrently did not experience an increased incidence of side effects.

7.6 Lidocaine No significant effects on the pharmacokinetics of propafenone or lidocaine have been seen following their concomitant use in patients.

However, concomitant use of propafenone and lidocaine has been reported to increase the risks of central nervous system side effects of lidocaine.

OVERDOSAGE

10 The symptoms of overdosage may include hypotension, somnolence, bradycardia, intra-atrial and intraventricular conduction disturbances, and rarely convulsions and high grade ventricular arrhythmias.

Defibrillation as well as infusion of dopamine and isoproterenol have been effective in controlling abnormal rhythm and blood pressure.

Convulsions have been alleviated with intravenous diazepam.

General supportive measures such as mechanical respiratory assistance and external cardiac massage may be necessary.

The hemodialysis of propafenone in patients with an overdose is expected to be of limited value in the removal of propafenone as a result of both its high protein binding (>95%) and large volume of distribution.

DESCRIPTION

11 Propafenone hydrochloride tablets, USP are an antiarrhythmic drug supplied in scored, film-coated tablets of 150, 225 and 300 mg for oral administration.

Propafenone has some structural similarities to beta-blocking agents.

Chemically, propafenone hydrochloride (HCl) is 2’-[2-Hydroxy-3-(propylamino)- propoxy]-3-phenylpropiophenone hydrochloride, with a molecular weight of 377.92.

The molecular formula is C 21 H 27 NO 3 •HCl.

The structural formula of propafenone HCl is given below: Propafenone HCl occurs as colorless crystals or white crystalline powder with a very bitter taste.

It is slightly soluble in water (20°C), chloroform and ethanol.

The following inactive ingredients are contained in the tablet: carnauba wax, hypromellose, lactose, magnesium stearate, microcrystalline cellulose, povidone, pregelatinized corn starch, sodium starch glycolate, stearic acid, titanium dioxide and triacetin.

This is the structural formula for Propafenone HCl.

CLINICAL STUDIES

14 In two randomized, crossover, placebo-controlled, double-blind trials of 60 to 90 days duration in patients with paroxysmal supraventricular arrhythmias [paroxysmal atrial fibrillation/flutter (PAF), or paroxysmal supraventricular tachycardia (PSVT)], propafenone reduced the rate of both arrhythmias, as shown in Table 3.

Table 3: Reduction of Arrythmias in Patients with PAF or PSVT Study 1 Study 2 Propafenone Placebo Propafenone Placebo PAF n = 30 n = 30 n = 9 n = 9 Percent attack free 53% 13% 67% 22% Median time to first recurrence > 98 days 8 days 62 days 5 days PSVT n = 45 n = 45 n = 15 N = 15 Percent attack free 47% 16% 38% 7% Median time to first recurrence > 98 days 12 days 31 days 8 days The patient population in the above trials was 50% male with a mean age of 57.3 years.

Fifty percent of the patients had a diagnosis of PAF and 50% had PSVT.

Eighty percent of the patients received 600 mg/day propafenone.

No patient died in the above 2 studies.

In U.S.

long-term safety trials, 474 patients (mean age: 57.4 ± 14.5 years) with supraventricular arrhythmias [195 with PAF, 274 with PSVT and 5 with both PAF and PSVT] were treated up to 5 years (mean: 14.4 months) with propafenone.

Fourteen of the patients died.

When this mortality rate was compared to the rate in a similar patient population (n = 194 patients; mean age: 43.0 ± 16.8 years) studied in an arrhythmia clinic, there was no age-adjusted difference in mortality.

This comparison was not, however, a randomized trial and the 95% confidence interval around the comparison was large, such that neither a significant adverse or favorable effect could be ruled out.

HOW SUPPLIED

16 /STORAGE AND HANDLING Product: 63629-3869 NDC: 63629-3869-1 30 TABLET, FILM COATED in a BOTTLE

RECENT MAJOR CHANGES

Contraindications ( 4 ) 3/2013 Warnings and Precautions, Unmasking Brugada Syndrome ( 5.2 ) 3/2013

GERIATRIC USE

8.5 Geriatric Use Clinical studies of propafenone hydrochloride did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects.

Other reported clinical experience has not identified differences in responses between the elderly and younger patients.

In general, dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range, reflecting the greater frequency of decreased hepatic, renal, or cardiac function, and of concomitant disease or other drug therapy.

DOSAGE FORMS AND STRENGTHS

3 150 mg, 225 mg and 300 mg scored, round, film-coated tablets.

Tablets: 150 mg, 225 mg, 300 mg ( 3 )

MECHANISM OF ACTION

12.1 Mechanism of Action Propafenone is a Class 1C antiarrhythmic drug with local anesthetic effects, and a direct stabilizing action on myocardial membranes.

The electrophysiological effect of propafenone manifests itself in a reduction of upstroke velocity (Phase 0) of the monophasic action potential.

In Purkinje fibers, and to a lesser extent myocardial fibers, propafenone reduces the fast inward current carried by sodium ions.

Diastolic excitability threshold is increased and effective refractory period prolonged.

Propafenone reduces spontaneous automaticity and depresses triggered activity.

Studies in anesthetized dogs and isolated organ preparations show that propafenone has beta-sympatholytic activity at about 1/50 the potency of propranolol.

Clinical studies employing isoproterenol challenge and exercise testing after single doses of propafenone indicate a beta- adrenergic blocking potency (per mg) about 1/40 that of propranolol in man.

In clinical trials, resting heart rate decreases of about 8% were noted at the higher end of the therapeutic plasma concentration range.

At very high concentrations in vitro , propafenone can inhibit the slow inward current carried by calcium, but this calcium antagonist effect probably does not contribute to antiarrhythmic efficacy.

Moreover, propafenone inhibits a variety of cardiac potassium currents in in vitro studies (i.e.

the transient outward, the delayed rectifier, and the inward rectifier current).

Propafenone has local anesthetic activity approximately equal to procaine.

Compared to propafenone, the main metabolite, 5-hydroxypropafenone, has similar sodium and calcium channel activity, but about 10 times less beta-blocking activity (N-depropylpropafenone has weaker sodium channel activity but equivalent affinity for beta-receptors).

INDICATIONS AND USAGE

1 Propafenone hydrochloride tablets are indicated to: prolong the time to recurrence of paroxysmal atrial fibrillation/flutter (PAF) associated with disabling symptoms in patients without structural heart disease.

prolong the time to recurrence of paroxysmal supraventricular tachycardia (PSVT) associated with disabling symptoms in patients without structural heart disease.

treat documented ventricular arrhythmias, such as sustained ventricular tachycardia that, in the judgment of the physician, are life-threatening.

Initiate treatment in the hospital.

Usage Considerations: The use of propafenone hydrochloride tablets in patients with permanent atrial fibrillation (AF) or in patients exclusively with atrial flutter or PSVT has not been evaluated.

Do not use propafenone hydrochloride tablets to control ventricular rate during AF.

Some patients with atrial flutter treated with propafenone have developed 1:1 conduction, producing an increase in ventricular rate.

Concomitant treatment with drugs that increase the functional atrioventricular (AV) nodal refractory period is recommended.

The use of propafenone hydrochloride tablets in patients with chronic atrial fibrillation has not been evaluated.

Because of the proarrhythmic effects of propafenone hydrochloride tablets, its use with lesser ventricular arrhythmias is not recommended, even if patients are symptomatic, and any use of the drug should be reserved for patients in whom, in the opinion of the physician, the potential benefits outweigh the risks.

The effect of propafenone on mortality has not been determined [see Boxed Warning ] .

Propafenone hydrochloride tablets are an antiarrhythmic indicated to: prolong the time to recurrence of symptomatic atrial fibrillation (AF) in patients with episodic (most likely paroxysmal or persistent) AF who do not have structural heart disease.

( 1 ) prolong the time to recurrence of paroxysmal supraventricular tachycardia (PSVT) associated with disabling symptoms in patients who do not have structural heart disease.

( 1 ) treat documented life-threatening ventricular arrhythmias.

( 1 ) U sage Considerations: Use in patients with permanent atrial fibrillation or with atrial flutter or PSVT has not been evaluated.

Do not use to control ventricular rate during atrial fibrillation.

( 1 ) In patients with atrial fibrillation and atrial flutter, use propafenone hydrochloride tablets with drugs that increase the atrioventricular nodal refractory period.

( 1 ) Because of proarrhythmic effects, use with lesser ventricular arrhythmias is not recommended, even if patients are symptomatic.

( 1 ) The effect of propafenone on mortality has not been determined.

( 1 )

PEDIATRIC USE

8.4 Pediatric Use The safety and effectiveness of propafenone in pediatric patients have not been established.

PREGNANCY

8.1 Pregnancy Pregnancy Category C.

There are no adequate and well-controlled studies in pregnant women.

Propafenone hydrochloride should be used during pregnancy only if the potential benefit justifies the potential risk to the fetus.

An imal Data: T e r a togenic Effects: Propafenone has been shown to be embryotoxic (decreased survival) in rabbits and rats when given in oral maternally toxic doses of 150 mg/kg day (about 3 times the maximum recommended human dose [MRHD] on a mg/m 2 basis) and 600 mg/kg/day (about 6 times the MRHD on a mg/m 2 basis), respectively.

Although maternally tolerated doses (up to 270 mg/kg/day, about 3 times the MRHD on a mg/m 2 basis) produced no evidence of embryotoxicity in rats, post-implantation loss was elevated in all rabbit treatment groups (doses as low as 15 mg/kg/day, about 1/3 the MRHD on a mg/m 2 basis).

Non-teratogenic Effects: In a study in which female rats received daily oral doses of propafenone from mid-gestation through weaning of their offspring, doses as low as 90 mg/kg/day (equivalent to the MRHD on a mg/m 2 basis) produced increases in maternal deaths.

Doses of 360 or more mg/kg/day (4 or more times the MRHD on a mg/m 2 basis) resulted in reductions in neonatal survival, body weight gain and physiological development.

NUSRING MOTHERS

8.3 Nursing Mothers Propafenone is excreted in human milk.

Because of the potential for serious adverse reactions in nursing infants from propafenone, decide whether to discontinue nursing or to discontinue the drug, taking into account the importance of the drug to the mother.

BOXED WARNING

WARNING: MORTALITY In the National Heart, Lung and Blood Institute’s Cardiac Arrhythmia Suppression Trial (CAST), a long-term, multi-center, randomized, double-blind study in patients with asymptomatic non-life-threatening ventricular arrhythmias who had a myocardial infarction more than 6 days but less than 2 years previously, an increased rate of death or reversed cardiac arrest rate (7.7%; 56/730) was seen in patients treated with encainide or flecainide (Class IC antiarrhythmics) compared with that seen in patients assigned to placebo (3.0%; 22/725).

The average duration of treatment with encainide or flecainide in this study was 10 months.

The applicability of the CAST results to other populations (e.g., those without recent myocardial infarction) or other antiarrhythmic drugs is uncertain, but at present, it is prudent to consider any IC antiarrhythmic to have a significant proarrhythmic risk in patients with structural heart disease.

Given the lack of any evidence that these drugs improve survival, antiarrhythmic agents should generally be avoided in patients with non-life-threatening ventricular arrhythmias, even if the patients are experiencing unp leasant, but not life-threatening, symptoms or signs.

W A RN I N G: M ORTALITY S e e full prescribing information for complete boxed warning.

A n increased rate of death or reversed cardiac arrest rate was seen in patients treated with encainide or flecainide (Class IC antiarrhythmics) compared with that seen in patients assigned to placebo.

At present it is prudent to consider any IC antiarrhythmic to have a significant risk of provoking proarrhythmic events in patients with structural heart disease.

Given the lack of any evidence that these drugs improve survival, antiarrhythmic agents should generally be avoided in patients with non- life-threatening ventricular arrhythmias, even if the patients are experiencing unpleasant, but not life-threatening, symptoms or signs.

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS May cause new or worsened arrhythmias.

Evaluate patients via ECG prior to and during therapy.

( 5.1 ) Propafenone hydrochloride may unmask Brugada or Brugada-like Syndrome.

( 4 , 5.2 ) Avoid use with other drugs that prolong the QT interval.

( 5.3 ) Avoid simultaneous use of propafenone with both a cytochrome P450 2D6 inhibitor and a 3A4 inhibitor.

( 5.4 ) May provoke overt heart failure.

( 5.5 ) May cause dose-related first degree AV block or other conduction disturbances.

Only use in patients with conduction disorders who have pacemakers.

( 5.6 ) May affect artificial pacemakers.

Monitor pacemaker function.

( 5.7 ) Agranulocytosis: Patients should report signs of infection.

( 5.8 ) May exacerbate myasthenia gravis.

( 5.11 ) 5.1 Proarrhythmic Effects Propafenone has caused new or worsened arrhythmias.

Such proarrhythmic effects include sudden death and life-threatening ventricular arrhythmias such as ventricular fibrillation, ventricular tachycardia, asystole and torsade de pointes.

It may also worsen premature ventricular contractions or supraventricular arrhythmias, and it may prolong the QT interval.

It is therefore essential that each patient given propafenone hydrochloride be evaluated electrocardiographically prior to and during therapy to determine whether the response to propafenone hydrochloride supports continued treatment.

Because propafenone prolongs the QRS interval in the electrocardiogram, changes in the QT interval are difficult to interpret [see Clinical Pharmacology (12.2) ].

In a U.S.

uncontrolled, open label, multicenter trial in patients with symptomatic supraventricular tachycardia (SVT), 1.9% (9/474) of these patients experienced ventricular tachycardia (VT) or ventricular fibrillation (VF) during the study.

However, in 4 of the 9 patients, the ventricular tachycardia was of atrial origin.

Six of the nine patients that developed ventricular arrhythmias did so within 14 days of onset of therapy.

About 2.3% (11/474) of all patients had a recurrence of SVT during the study which could have been a change in the patients’ arrhythmia behavior or could represent a proarrhythmic event.

Case reports in patients treated with propafenone for atrial fibrillation/flutter have included increased premature ventricular contractions (PVCs), VT, VF, torsade de pointes, asystole, and death.

Overall in clinical trials with propafenone hydrochloride (which included patients treated for ventricular arrhythmias, atrial fibrillation/flutter, and PSVT), 4.7% of all patients had new or worsened ventricular arrhythmia possibly representing a proarrhythmic event (0.7% was an increase in PVCs; 4.0% a worsening, or new appearance, of VT or VF).

Of the patients who had worsening of VT (4%), 92% had a history of VT and/or VT/VF, 71% had coronary artery disease, and 68% had a prior myocardial infarction.

The incidence of proarrhythmia in patients with less serious or benign arrhythmias, which include patients with an increase in frequency of PVCs, was 1.6%.

Although most proarrhythmic events occurred during the first week of therapy, late events also were seen and the CAST study [see Boxed Warning: Mortality ] suggests that an increased risk of proarrythmia is present throughout treatment.

In a study of sustained-release propafenone, there were too few deaths to assess the long term risk to patients.

There were 5 deaths, 3 in the pooled sustained-release propafenone group (0.8%) and 2 in the placebo group (1.6%).

In the overall sustained-release propafenone and propafenone hydrochloride immediate-release database of 8 studies, the mortality rate was 2.5% per year on propafenone and 4.0% per year on placebo.

Concurrent use of propafenone with other antiarrhythmic agents has not been well studied.

5.2 Unmasking Brugada Syndrome Brugada Syndrome may be unmasked after exposure to propafenone hydrochloride.

Perform an ECG after initiation of propafenone hydrochloride, and discontinue the drug if changes are suggestive of Brugada Syndrome [see Contraindications (4) ] .

5.3 Use with Drugs that Prolong the QT Interval and Antiarrhythmic Agents The use of propafenone hydrochloride in conjunction with other drugs that prolong the QT interval has not been extensively studied.

Such drugs may include many antiarrhythmics, some phenothiazines, tricyclic antidepressants, and oral macrolides.

Withhold Class IA and III antiarrhythmic agents for at least 5 half-lives prior to dosing with propafenone hydrochloride.

Avoid the use of propafenone with Class IA and III antiarrhythmic agents (including quinidine and amiodarone).

There is only limited experience with the concomitant use of Class IB or IC antiarrhythmics.

5.4 Drug Interactions: Simultaneous Use with Inhibitors of Cytochrome P450 Isoenzymes 2D6 and 3A4 Propafenone is metabolized by CYP2D6, CYP3A4, and CYP1A2 isoenzymes.

Approximately 6% of Caucasians in the U.S.

population are naturally deficient in CYP2D6 activity and to a somewhat lesser extent in other demographic groups.

Drugs that inhibit these CYP pathways (such as desipramine, paroxetine, ritonavir, sertraline for CYP2D6; ketoconazole, erythromycin, saquinavir, and grapefruit juice for CYP3A4; and amiodarone and tobacco smoke for CYP1A2) can be expected to cause increased plasma levels of propafenone.

Increased exposure to propafenone may lead to cardiac arrhythmias and exaggerated beta-adrenergic blocking activity.

Because of its metabolism, the combination of CYP3A4 inhibition and either CYP2D6 deficiency or CYP2D6 inhibition in users of propafenone is potentially hazardous.

Therefore, avoid simultaneous use of propafenone hydrochloride with both a CYP2D6 inhibitor and a CYP3A4 inhibitor.

5.5 Use in Patients with a History of Heart Failure Propafenone exerts a negative inotropic activity on the myocardium as well as beta blockade effects and may provoke overt heart failure.

In clinical trial experience with propafenone hydrochloride, new or worsened congestive heart failure (CHF) has been reported in 3.7% of patients with ventricular arrhythmia; of those 0.9% were considered probably or definitely related to propafenone HCl.

Of the patients with CHF probably related to propafenone, 80% had preexisting heart failure and 85% had coronary artery disease.

CHF attributable to propafenone HCl developed rarely (< 0.2%) in ventricular arrhythmia patients who had no previous history of CHF.

CHF occurred in 1.9% of patients studied with PAF or PSVT.

In a U.S.

trial of sustained-release propafenone in patients with symptomatic AF, heart failure was reported in 4 (1.0%) patients receiving sustained-release propafenone (all doses), compared to 1 (0.8%) patient receiving placebo.

5.6 Conduction Disturbances Propafenone slows atrioventricular conduction and may also cause dose-related first degree AV block.

Average PR interval prolongation and increases in QRS duration are also dose-related.

Do not give propafenone to patients with atrioventricular and intraventricular conduction defects in the absence of a pacemaker [see Contraindications (4) and Clinical Pharmacology (12.2) ].

The incidence of first degree, second degree, and third degree AV block observed in 2,127 ventricular arrhythmia patients was 2.5%, 0.6%, and 0.2%, respectively.

Development of second or third degree AV block requires a reduction in dosage or discontinuation of propafenone HCl.

Bundle branch block (1.2%) and intraventricular conduction delay (1.1%) have been reported in patients receiving propafenone.

Bradycardia has also been reported (1.5%).

Experience in patients with sick sinus node syndrome is limited and these patients should not be treated with propafenone.

In a U.S.

trial in 523 patients with a history of symptomatic AF treated with sustained-release propafenone, sinus bradycardia (rate <50 beats/min) was reported with the same frequency with sustained-release propafenone and placebo.

5.7 Effects on Pacemaker Threshold Propafenone may alter both pacing and sensing thresholds of implanted pacemakers and defibrillators.

During and after therapy, monitor and re-program these devices accordingly.

5.8 Agranulocytosis Agranulocytosis has been reported in patients receiving propafenone.

Generally, the agranulocytosis occurred within the first 2 months of propafenone therapy and upon discontinuation of therapy, the white count usually normalized by 14 days.

Unexplained fever or decrease in white cell count, particularly during the initial 3 months of therapy, warrant consideration of possible agranulocytosis or granulocytopenia.

Instruct patients to report promptly any signs of infection such as fever, sore throat, or chills.

5.9 Use in Patients with Hepatic Dysfunction Propafenone is highly metabolized by the liver.

Severe liver dysfunction increases the bioavailability of propafenone to approximately 70% compared to 3 to 40% in patients with normal liver function.

In 8 patients with moderate to severe liver disease, the mean half-life was approximately 9 hours.

Increased bioavailability of propafenone in these patients may result in excessive accumulation.

Carefully monitor patients with impaired hepatic function for excessive pharmacological effects [see Overdosage (10) ] .

5.10 Use in Patients with Renal Dysfunction Approximately 50% of propafenone metabolites are excreted in the urine following administration of propafenone hydrochloride.

In patients with impaired renal function, monitor for signs of overdosage [see Overdosage (10) ].

5.11 Use in Patients with Myasthenia Gravis Exacerbation of myasthenia gravis has been reported during propafenone therapy.

5.12 Elevated ANA Titers Positive ANA titers have been reported in patients receiving propafenone.

They have been reversible upon cessation of treatment and may disappear even in the face of continued propafenone therapy.

These laboratory findings were usually not associated with clinical symptoms, but there is one published case of drug-induced lupus erythematosis (positive rechallenge); it resolved completely upon discontinuation of therapy.

Carefully evaluate patients who develop an abnormal ANA test and, if persistent or worsening elevation of ANA titers is detected, consider discontinuing therapy.

5.13 Impaired Spermatogenesis Reversible disorders of spermatogenesis have been demonstrated in monkeys, dogs and rabbits after high dose intravenous administration of propafenone.

Evaluation of the effects of short-term propafenone hydrochloride administration on spermatogenesis in 11 normal subjects suggested that propafenone produced a reversible, short-term drop (within normal range) in sperm count.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION See FDA-approved patient labeling (Patient Information).

17.1 Information for Patients Patients should be instructed to notify their health care providers of any change in over-the-counter, prescription and supplement use.

The health care provider should assess the patients’ medication history including all over-the-counter, prescription and herbal/natural preparations for those that may affect the pharmacodynamics or kinetics of propafenone hydrochloride [see Warnings and Precautions (5.4) ] .

Patients should also check with their health care providers prior to taking a new over-the-counter medicine.

If patients experience symptoms that may be associated with altered electrolyte balance, such as excessive or prolonged diarrhea, sweating, vomiting, or loss of appetite or thirst, these conditions should be immediately reported to their health care provider.

Patients should be instructed NOT to double the next dose if a dose is missed.

The next dose should be taken at the usual time.

DOSAGE AND ADMINISTRATION

2 The dose of propafenone hydrochloride tablets must be individually titrated on the basis of response and tolerance.

Initiate therapy with propafenone hydrochloride tablets 150 mg given every eight hours (450 mg/day).

Dosage may be increased at a minimum of 3 to 4 day intervals to 225 mg every 8 hours (675 mg/day).

If additional therapeutic effect is needed, the dose of propafenone hydrochloride tablets may be increased to 300 mg every 8 hours (900 mg/day).

The usefulness and safety of dosages exceeding 900 mg per day have not been established.

In patients with hepatic impairment or those with significant widening of the QRS complex or second or third degree AV block, consider reducing the dose.

As with other antiarrhythmic agents, in the elderly or in ventricular arrhythmia patients with marked previous myocardial damage, the dose of propafenone hydrochloride tablets should be increased more gradually during the initial phase of treatment.

The combination of CYP3A4 inhibition and either CYP2D6 deficiency or CYP2D6 inhibition with the simultaneous administration of propafenone may significantly increase the concentration of propafenone and thereby increase the risk of proarrhythmia and other adverse events.

Therefore, avoid simultaneous use of propafenone hydrochloride tablets with both a CYP2D6 inhibitor and a CYP3A4 inhibitor [see Warnings and Precautions (5.4) and Drug Interactions (7.1) ] .

Initiate therapy with 150 mg given every 8 hours.

( 2 ) As needed, uptitrate in 3 to 4 days to 225 to 300 mg every 8 hours.

( 2 ) Consider reducing the dose in patients with hepatic impairment, significant widening of the QRS complex, or second or third degree AV block.

( 2 )

cefdinir 50 MG/ML Oral Suspension

Generic Name: CEFDINIR
Brand Name: Cefdinir
  • Substance Name(s):
  • CEFDINIR

WARNINGS

BEFORE THERAPY WITH CEFDINIR IS INSTITUTED, CAREFUL INQUIRY SHOULD BE MADE TO DETERMINE WHETHER THE PATIENT HAS HAD PREVIOUS HYPERSENSITIVITY REACTIONS TO CEFDINIR, OTHER CEPHALOSPORINS, PENICILLINS, OR OTHER DRUGS.

IF CEFDINIR IS TO BE GIVEN TO PENICILLIN-SENSITIVE PATIENTS, CAUTION SHOULD BE EXERCISED BECAUSE CROSS-HYPERSENSITIVITY AMONG β-LACTAM ANTIBIOTICS HAS BEEN CLEARLY DOCUMENTED AND MAY OCCUR IN UP TO 10% OF PATIENTS WITH A HISTORY OF PENICILLIN ALLERGY.

IF AN ALLERGIC REACTION TO CEFDINIR OCCURS, THE DRUG SHOULD BE DISCONTINUED.

SERIOUS ACUTE HYPERSENSITIVITY REACTIONS MAY REQUIRE TREATMENT WITH EPINEPHRINE AND OTHER EMERGENCY MEASURES, INCLUDING OXYGEN, INTRAVENOUS FLUIDS, INTRAVENOUS ANTIHISTAMINES, CORTICOSTEROIDS, PRESSOR AMINES, AND AIRWAY MANAGEMENT, AS CLINICALLY INDICATED.

Clostridium difficile associated diarrhea (CDAD) has been reported with use of nearly all antibacterial agents, including cefdinir, and may range in severity from mild diarrhea to fatal colitis.

Treatment with antibacterial agents alters the normal flora of the colon leading to overgrowth of C.

difficile.

C.

difficile produces toxins A and B which contribute to the development of CDAD.

Hypertoxin producing strains of C.

difficile cause increased morbidity and mortality, as these infections can be refractory to antimicrobial therapy and may require colectomy.

CDAD must be considered in all patients who present with diarrhea following antibacterial use.

Careful medical history is necessary since CDAD has been reported to occur over two months after the administration of antibacterial agents.

If CDAD is suspected or confirmed, ongoing antibacterial use not directed against C.

difficile may need to be discontinued.

Appropriate fluid and electrolyte management, protein supplementation, antibacterial treatment of C.

difficile , and surgical evaluation should be instituted as clinically indicated.

DRUG INTERACTIONS

Drug Interactions Antacids: ( aluminum- or magnesium-containing ) Concomitant administration of 300 mg cefdinir capsules with 30 mL Maalox ® TC suspension reduces the rate (C max ) and extent (AUC) of absorption by approximately 40%.

Time to reach C max is also prolonged by 1 hour.

There are no significant effects on cefdinir pharmacokinetics if the antacid is administered 2 hours before or 2 hours after cefdinir.

If antacids are required during cefdinir therapy, cefdinir should be taken at least 2 hours before or after the antacid.

Probenecid As with other β-lactam antibiotics, probenecid inhibits the renal excretion of cefdinir, resulting in an approximate doubling in AUC, a 54% increase in peak cefdinir plasma levels, and a 50% prolongation in the apparent elimination t ½ .

Iron Supplements and Foods Fortified With Iron Concomitant administration of cefdinir with a therapeutic iron supplement containing 60 mg of elemental iron (as FeSO 4 ) or vitamins supplemented with 10 mg of elemental iron reduced extent of absorption by 80% and 31%, respectively.

If iron supplements are required during cefdinir therapy, cefdinir should be taken at least 2 hours before or after the supplement.

The effect of foods highly fortified with elemental iron (primarily iron-fortified breakfast cereals) on cefdinir absorption has not been studied.

Concomitantly administered iron-fortified infant formula (2.2 mg elemental iron/6 oz) has no significant effect on cefdinir pharmacokinetics.

Therefore, cefdinir for oral suspension can be administered with iron-fortified infant formula.

There have been reports of reddish stools in patients receiving cefdinir.

In many cases, patients were also receiving iron-containing products.

The reddish color is due to the formation of a nonabsorbable complex between cefdinir or its breakdown products and iron in the gastrointestinal tract.

OVERDOSAGE

Information on cefdinir overdosage in humans is not available.

In acute rodent toxicity studies, a single oral 5600 mg/kg dose produced no adverse effects.

Toxic signs and symptoms following overdosage with other β-lactam antibiotics have included nausea, vomiting, epigastric distress, diarrhea, and convulsions.

Hemodialysis removes cefdinir from the body.

This may be useful in the event of a serious toxic reaction from overdosage, particularly if renal function is compromised.

DESCRIPTION

Cefdinir for oral suspension, USP contains the active ingredient cefdinir USP, an extended-spectrum, semisynthetic cephalosporin, for oral administration.

Chemically, cefdinir is [6R-[6α,7β(Z)]]-7-[[(2-amino-4-thiazolyl)(hydroxyimino)acetyl]amino]-3-ethenyl-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid.

Cefdinir USP is a white to slightly brownish-yellow solid.

It is slightly soluble in dilute hydrochloric acid and sparingly soluble in 0.1 M pH 7.0 phosphate buffer.

The molecular formula is C 14 H 13 N 5 O 5 S 2 and the molecular weight is 395.42.

Cefdinir has the structural formula shown below: Cefdinir for oral suspension, USP after reconstitution, contains 125 mg cefdinir USP per 5 mL or 250 mg cefdinir USP per 5 mL and the following inactive ingredients: sucrose, sodium benzoate, colloidal silicone dioxide, xanthan gum, guar gum, citric acid (anhydrous), sodium citrate (dihydrate), strawberry flavour, fresh cream flavour and magnesium stearate.

Chemical Structure

CLINICAL STUDIES

Community-Acquired Bacterial Pneumonia In a controlled, double-blind study in adults and adolescents conducted in the U.S., cefdinir BID was compared with cefaclor 500 mg TID.

Using strict evaluability and microbiologic/clinical response criteria 6 to 14 days posttherapy, the following clinical cure rates, presumptive microbiologic eradication rates, and statistical outcomes were obtained: U.S.

Community-Acquired Pneumonia Study Cefdinir vs Cefaclor Cefdinir BID Cefaclor TID Outcome Clinical Cure Rates 150/187 (80%) 147/186 (79%) Cefdinir equivalent to control Eradication Rates Overall 177/195 (91%) 184/200 (92%) Cefdinir equivalent to control S.

pneumoniae 31/31 (100%) 35/35 (100%) H.

influenzae 55/65 (85%) 60/72 (83%) M.

catarrhalis 10/10 (100%) 11/11 (100%) H.

parainfluenzae 81/89 (91%) 78/82 (95%) In a second controlled, investigator-blind study in adults and adolescents conducted primarily in Europe, cefdinir BID was compared with amoxicillin/clavulanate 500/125 mg TID.

Using strict evaluability and clinical response criteria 6 to 14 days posttherapy, the following clinical cure rates, presumptive microbiologic eradication rates, and statistical outcomes were obtained: European Community-Acquired Pneumonia Study Cefdinir vs Amoxicillin/Clavulanate Cefdinir BID Amoxicillin/ Clavulanate TID Outcome Clinical Cure Rates 83/104 (80%) 86/97(89%) Cefdinir not equivalent to control Eradication Rates Overall 85/96 (89%) 84/90 (93%) Cefdinir equivalent to control S.

pneumoniae 42/44 (95%) 43/44 (98%) H.

influenzae 26/35 (74%) 21/26 (81%) M.

catarrhalis 6/6 (100%) 8/8 (100%) H.

parainfluenzae 11/11 (100%) 12/12 (100%) Streptococcal Pharyngitis/Tonsillitis In four controlled studies conducted in the United States, cefdinir was compared with 10 days of penicillin in adult, adolescent, and pediatric patients.

Two studies (one in adults and adolescents, the other in pediatric patients) compared 10 days of cefdinir QD or BID to penicillin 250 mg or 10 mg/kg QID.

Using strict evaluability and microbiologic/clinical response criteria 5 to 10 days posttherapy, the following clinical cure rates, microbiologic eradication rates, and statistical outcomes were obtained: Pharyngitis/Tonsillitis Studies Cefdinir (10 days) vs Penicillin (10 days) Study Efficacy Parameter Cefdinir QD Cefdinir BID Penicillin QID Outcome Adults/ Adolescents Eradication of S.

pyogenes 192/210 (91%) 199/217 (92%) 181/217 (83%) Cefdinir superior to control Clinical Cure Rates 199/210 (95%) 209/217 (96%) 193/217 (89%) Cefdinir superior to control Pediatric Patients Eradication of S.

pyogenes 215/228 (94%) 214/227 (94%) 159/227 (70%) Cefdinir superior to control Clinical Cure Rates 222/228 (97%) 218/227 (96%) 196/227 (86%) Cefdinir superior to control Two studies (one in adults and adolescents, the other in pediatric patients) compared 5 days of cefdinir BID to 10 days of penicillin 250 mg or 10 mg/kg QID.

Using strict evaluability and microbiologic/clinical response criteria 4 to 10 days posttherapy, the following clinical cure rates, microbiologic eradication rates, and statistical outcomes were obtained: Pharyngitis/Tonsillitis Studies Cefdinir (5 days) vs Penicillin (10 days) Study Efficacy Parameter Cefdinir BID Penicillin QID Outcome Adults/ Adolescents Eradication of S.

pyogenes 193/218 (89%) 176/214 (82%) Cefdinir equivalent to control Clinical Cure Rates 194/218 (89%) 181/214 (85%) Cefdinir equivalent to control Pediatric Patients Eradication of S.

pyogenes 176/196 (90%) 135/193 (70%) Cefdinir superior to control Clinical Cure Rates 179/196 (91%) 173/193 (90%) Cefdinir equivalent to control

HOW SUPPLIED

Cefdinir for Oral Suspension, USP 125 mg/5 mL is a off-white to yellowish — white colored granular powder, on constitution with water, forming an off-white to yellowish-white colored suspension with strawberry and cream flavors.

60 mL Bottle NDC 65862-218-60 100 mL Bottle NDC 65862-218-01 Cefdinir for Oral Suspension, USP 250 mg/5 mL is a off-white to yellowish — white colored granular powder, on constitution with water, forming an off-white to yellowish-white colored suspension with strawberry and cream flavors.

60 mL Bottle NDC 65862-219-60 100 mL Bottle NDC 65862-219-01 Store dry powder at 20° to 25°C (68° to 77°F); excursions permitted to 15° to 30°C (59° to 86°F) [see USP Controlled Room Temperature].

Once reconstituted, the oral suspension can be stored at controlled room temperature for 10 days.

GERIATRIC USE

Geriatric Use Efficacy is comparable in geriatric patients and younger adults.

While cefdinir has been well-tolerated in all age groups, in clinical trials geriatric patients experienced a lower rate of adverse events, including diarrhea, than younger adults.

Dose adjustment in elderly patients is not necessary unless renal function is markedly compromised (see DOSAGE AND ADMINISTRATION ).

INDICATIONS AND USAGE

To reduce the development of drug-resistant bacteria and maintain the effectiveness of cefdinir and other antibacterial drugs, cefdinir should be used only to treat or prevent infections that are proven or strongly suspected to be caused by susceptible bacteria.

When culture and susceptibility information are available, they should be considered in selecting or modifying antibacterial therapy.

In the absence of such data, local epidemiology and susceptibility patterns may contribute to the empiric selection of therapy.

Cefdinir for oral suspension is indicated for the treatment of patients with mild to moderate infections caused by susceptible strains of the designated microorganisms in the conditions listed below.

Adults and Adolescents Community-Acquired Pneumonia caused by Haemophilus influenzae (including β-lactamase producing strains), Haemophilus parainfluenzae (including β-lactamase producing strains), Streptococcus pneumoniae (penicillin-susceptible strains only), and Moraxella catarrhalis (including β-lactamase producing strains) (see CLINICAL STUDIES ).

Acute Exacerbations of Chronic Bronchitis caused by Haemophilus influenzae (including β-lactamase producing strains), Haemophilus parainfluenzae (including β-lactamase producing strains), Streptococcus pneumoniae (penicillin-susceptible strains only), and Moraxella catarrhalis (including β-lactamase producing strains).

Acute Maxillary Sinusitis caused by Haemophilus influenzae (including β-lactamase producing strains), Streptococcus pneumoniae (penicillin-susceptible strains only), and Moraxella catarrhalis (including β-lactamase producing strains).

NOTE: For information on use in pediatric patients, see PRECAUTIONS, Pediatric Use and DOSAGE AND ADMINISTRATION .

Pharyngitis/Tonsillitis caused by Streptococcus pyogenes (see CLINICAL STUDIES ).

NOTE: Cefdinir is effective in the eradication of S.

pyogenes from the oropharynx.

Cefdinir has not, however, been studied for the prevention of rheumatic fever following S.

pyogenes pharyngitis/tonsillitis.

Only intramuscular penicillin has been demonstrated to be effective for the prevention of rheumatic fever.

Uncomplicated Skin and Skin Structure Infections caused by Staphylococcus aureus (including β-lactamase producing strains) and Streptococcus pyogenes .

Pediatric Patients Acute Bacterial Otitis Media caused by Haemophilus influenzae (including β-lactamase producing strains), Streptococcus pneumoniae (penicillin-susceptible strains only), and Moraxella catarrhalis (including β-lactamase producing strains).

Pharyngitis/Tonsillitis caused by Streptococcus pyogenes (see CLINICAL STUDIES ).

NOTE: Cefdinir is effective in the eradication of S.

pyogenes from the oropharynx.

Cefdinir has not, however, been studied for the prevention of rheumatic fever following S.

pyogenes pharyngitis/tonsillitis.

Only intramuscular penicillin has been demonstrated to be effective for the prevention of rheumatic fever.

Uncomplicated Skin and Skin Structure Infections caused by Staphylococcus aureus (including β-lactamase producing strains) and Streptococcus pyogenes .

PEDIATRIC USE

Pediatric Use Safety and efficacy in neonates and infants less than 6 months of age have not been established.

Use of cefdinir for the treatment of acute maxillary sinusitis in pediatric patients (age 6 months through 12 years) is supported by evidence from adequate and well-controlled studies in adults and adolescents, the similar pathophysiology of acute sinusitis in adult and pediatric patients, and comparative pharmacokinetic data in the pediatric population.

PREGNANCY

Pregnancy Teratogenic Effects Pregnancy Category B Cefdinir was not teratogenic in rats at oral doses up to 1000 mg/kg/day (70 times the human dose based on mg/kg/day, 11 times based on mg/m 2 /day) or in rabbits at oral doses up to 10 mg/kg/day (0.7 times the human dose based on mg/kg/day, 0.23 times based on mg/m 2 /day).

Maternal toxicity (decreased body weight gain) was observed in rabbits at the maximum tolerated dose of 10 mg/kg/day without adverse effects on offspring.

Decreased body weight occurred in rat fetuses at ≥100 mg/kg/day, and in rat offspring at ≥32 mg/kg/day.

No effects were observed on maternal reproductive parameters or offspring survival, development, behavior, or reproductive function.

There are, however, no adequate and well-controlled studies in pregnant women.

Because animal reproduction studies are not always predictive of human response, this drug should be used during pregnancy only if clearly needed.

NUSRING MOTHERS

Nursing Mothers Following administration of single 600 mg doses, cefdinir was not detected in human breast milk.

INFORMATION FOR PATIENTS

Information for Patients Patients should be counseled that antibacterial drugs including cefdinir should only be used to treat bacterial infections.

They do not treat viral infections (e.g., the common cold).

When cefdinir is prescribed to treat a bacterial infection, patients should be told that although it is common to feel better early in the course of therapy, the medication should be taken exactly as directed.

Skipping doses or not completing the full course of therapy may (1) decrease the effectiveness of the immediate treatment and (2) increase the likelihood that bacteria will develop resistance and will not be treatable by cefdinir or other antibacterial drugs in the future.

Antacids containing magnesium or aluminum interfere with the absorption of cefdinir.

If this type of antacid is required during cefdinir therapy, cefdinir should be taken at least 2 hours before or after the antacid.

Iron supplements, including multivitamins that contain iron, interfere with the absorption of cefdinir.

If iron supplements are required during cefdinir therapy, cefdinir should be taken at least 2 hours before or after the supplement.

Iron-fortified infant formula does not significantly interfere with the absorption of cefdinir.

Therefore, cefdinir for oral suspension can be administered with iron-fortified infant formula.

Diabetic patients and caregivers should be aware that the 125 mg/5 mL oral suspension contains 2.94 g of sucrose per teaspoon and the 250 mg/5 mL oral suspension contains 2.82 g of sucrose for teaspoon.

Diarrhea is a common problem caused by antibiotics which usually ends when the antibiotic is discontinued.

Sometimes after starting treatment with antibiotics, patients can develop watery and bloody stools (with or without stomach cramps and fever) even as late as two or more months after having taken the last dose of the antibiotic.

If this occurs, patients should contact their physician as soon as possible.

DOSAGE AND ADMINISTRATION

(see INDICATIONS AND USAGE for Indicated Pathogens) The recommended dosage and duration of treatment for infections in pediatric patients are described in the following chart; the total daily dose for all infections is 14 mg/kg, up to a maximum dose of 600 mg per day.

Once-daily dosing for 10 days is as effective as BID dosing.

Once-daily dosing has not been studied in skin infections; therefore, cefdinir for oral suspension should be administered twice daily in this infection.

Cefdinir for oral suspension may be administered without regard to meals.

Pediatric Patients (Age 6 Months Through 12 Years) Type of Infection Dosage Duration Acute Bacterial Otitis Media 7 mg/kg q12h or 14 mg/kg q24h 5 to 10 days 10 days Acute Maxillary Sinusitis 7 mg/kg q12h or 14 mg/kg q24h 10 days 10 days Pharyngitis/Tonsillitis 7 mg/kg q12h or 14 mg/kg q24h 5 to 10 days 10 days Uncomplicated Skin and Skin Structure Infections 7 mg/kg q12h 10 days CEFDINIR FOR ORAL SUSPENSION PEDIATRIC DOSAGE CHART a Pediatric patients who weigh ≥ 43 kg should receive the maximum daily dose of 600 mg.

Weight 125 mg/5 mL 250 mg/5 mL 9 kg/20 lbs 2.5 mL q12h or 5 mL q24h Use 125 mg/5 mL product 18 kg/40 lbs 5 mL q12h or 10 mL q24h 2.5 mL q12h or 5 mL q24h 27 kg/60 lbs 7.5 mL q12h or 15 mL q24h 3.75 mL q12h or 7.5 mL q24h 36 kg/80 lbs 10 mL q12h or 20 mL q24h 5 mL q12h or 10 mL q24h ≥ 43 kg a /95 lbs 12 mL q12h or 24 mL q24h 6 mL q12h or 12 mL q24h Patients With Renal Insufficiency For adult patients with creatinine clearance <30 mL/min, the dose of cefdinir should be 300 mg given once daily.

Creatinine clearance is difficult to measure in outpatients.

However, the following formula may be used to estimate creatinine clearance (CL cr ) in adult patients.

For estimates to be valid, serum creatinine levels should reflect steady-state levels of renal function.

Males: CL cr = (weight) (140 – age) (72) (serum creatinine) Females: CL cr = 0.85 x above value where creatinine clearance is in mL/min, age is in years, weight is in kilograms, and serum creatinine is in mg/dL.

1 The following formula may be used to estimate creatinine clearance in pediatric patients: CL cr = K x body length or height serum creatinine where K=0.55 for pediatric patients older than 1 year 2 and 0.45 for infants (up to 1 year) 3 .

In the above equation, creatinine clearance is in mL/min/1.73 m 2 , body length or height is in centimeters, and serum creatinine is in mg/dL.

For pediatric patients with a creatinine clearance of <30 mL/min/1.73 m 2 , the dose of cefdinir should be 7 mg/kg (up to 300 mg) given once daily.

Patients on Hemodialysis Hemodialysis removes cefdinir from the body.

In patients maintained on chronic hemodialysis, the recommended initial dosage regimen is a 300 mg or 7 mg/kg dose every other day.

At the conclusion of each hemodialysis session, 300 mg (or 7 mg/kg) should be given.

Subsequent doses (300 mg or 7 mg/kg) are then administered every other day.

Directions for Mixing Cefdinir for Oral Suspension Final Concentration Final Volume (mL) Amount of Water Directions 125 mg/5 mL 60 100 38 mL 63 mL Tap bottle to loosen powder, then add water in 2 portions.

Shake well after each aliquot.

250 mg/5 mL 60 100 38 mL 63 mL Tap bottle to loosen powder, then add water in 2 portions.

Shake well after each aliquot.

After mixing, the suspension can be stored at room temperature (25°C/77°F).

The container should be kept tightly closed, and the suspension should be shaken well before each administration.

The suspension may be used for 10 days, after which any unused portion must be discarded.

methylene blue 100 MG in 10 ML (1 % ) Injection

WARNINGS

Methylene Blue should not be given by subcutaneous or intrathecal injection.

Methylene blue is a potent monoamine oxidase inhibitor: Methylene blue has been demonstrated to be a potent monoamine oxidase inhibitor (MAOI) and may cause potentially fatal serotonin toxicity (serotonin syndrome) when combined with serotonin reputake inhibitors (SRIs).

(4) (See DRUG INTERACTIONS.) Serotonin toxicity is characterized by development of neuromuscular hyperactivity (tremor, clonus, myoclonus and hyperreflexia, and, in the advanced stage, pyramidal rigidity); autonomic hyperactivity (diaphoresis, fever, tachycardia, tachypnoea, and mydraisis); and altered mental status (agitation, excitement, and in the advanced stage, confusion).

If methylene blue is judged to be indicated, SRIs must be ceased, prior to treatment/procedure/surgery.

DESCRIPTION

Methylene Blue Injection, USP is a sterile solution of Phenothiazin_5_ium, 3, 7-bis (dimethylamino)- chloride, trihydrate.

Each mL contains Methylene Blue, USP, 10 mg in Water for Injection q.s.

pH adjusted with Hydrochloric Acid and /or Sodium Hydroxide when necessary.

The structural formula is: Formula1.jpg

HOW SUPPLIED

REFERENCES: (1) DiSanto AR, Wagner JG.

Pharmacokinetics of highly ionized drugs II: methylene blue-absorption, metabolism, and excretion in man and dog after oral administration.

J Pharm Sci.

1972;61:1086-1090 (2) Cragan JD.

Teratogen update: methylene blue.

Teratology.

1999;60:42-48.

(3) Kidd SA, Lancaster PA, Anderson JC, Boogert A, Fisher CC, Robertson R et al.

Fetal death after exposure to methylene bluedye during mid-trimester amniocentesis in twin pregnancy.

Prenat Diagn.

1996;16:39-47.

(4) Ramsay RR, Dunford C, Gillman PK.

Methylene blue and serotonin toxicity: inhibition of monoamine oxidase A (MAOA) confirms a theoretical prediction.

Br J Pharmacol.

2007;152:946-51.

(5) Beutler E.

G6PD Deficiency.

Blood.

1994;84:3613-3636 (6) Youngster I.

Arcavi L.

Schechmaster R.

Akayzen Y, Popliski H, Shimonov J, Beig S, Berkovitch M.

Medications and glucose-6-phosphate dehydrogenase deficiency: an evidence-based review.

Drug Saf.

2010;33:713-726 IN0372 Rev.

3/11 MG# 14080 AMERICAN REGENT, INC Shirley, NY 11967 Image1.jpg

INDICATIONS AND USAGE

Drug-induced methemoglobinemia.

PREGNANCY

USE IN Pregnancy Category X: Epidemiologic evidence exists that Methylene blue is a teratogen.

An association exists between the use of methylene blue in amniocentesis and atresia of the ileum and jejunum, ileal occlusions and other adverse effects in the neonate.

(2,3) Methylene blue Injection should not be administered to pregnant women during amniocentesis due to the risk of teratogenicity and other newborn adverse effects (see CONTRAINDICATIONS).

DOSAGE AND ADMINISTRATION

0.1 to 0.2 mL per kg body weight (0.045 to 0.09 mL per pound body weight).

Inject Methylene Blue intravenously very slowly over a period of several minutes.

Methylene blue must be injected intravenously very slowly over a period of several minutes to prevent local high concentration of the compound from producing additional methemoglobin.

Do not exceed recommended dosage.

Parenteral drug products should be inspected visually for particulate matter and discoloration, whenever solution and container permit.

Store at 20 degrees-25 degrees C (68 degrees-77 degrees F); excursions permitted to 15 degrees-30 degrees C (59 degrees-86 degrees F) (See USP Controlled Room Temperature).

MALARONE 250 MG / 100 MG Oral Tablet

DRUG INTERACTIONS

7 Administration with rifampin or rifabutin is known to reduce atovaquone concentrations; concomitant use with MALARONE is not recommended.

( ) • 7.1 Proguanil may potentiate anticoagulant effect of warfarin and other coumarin-based anticoagulants.

Caution advised when initiating or withdrawing MALARONE in patients on anticoagulants; coagulation tests should be closely monitored.

( ) • 7.2 Tetracycline may reduce atovaquone concentrations; parasitemia should be closely monitored.

( ) • 7.3 7.1 Rifampin/Rifabutin Concomitant administration of rifampin or rifabutin is known to reduce atovaquone concentrations .

The concomitant administration of MALARONE and rifampin or rifabutin is not recommended.

[see Clinical Pharmacology (12.3)] 7.2 Anticoagulants Proguanil may potentiate the anticoagulant effect of warfarin and other coumarin-based anticoagulants.

The mechanism of this potential drug interaction has not been established.

Caution is advised when initiating or withdrawing malaria prophylaxis or treatment with MALARONE in patients on continuous treatment with coumarin-based anticoagulants.

When these products are administered concomitantly, coagulation tests should be closely monitored.

7.3 Tetracycline Concomitant treatment with tetracycline has been associated with a reduction in plasma concentrations of atovaquone .

Parasitemia should be closely monitored in patients receiving tetracycline.

[see Clinical Pharmacology (12.3)] 7.4 Metoclopramide While antiemetics may be indicated for patients receiving MALARONE, metoclopramide may reduce the bioavailability of atovaquone and should be used only if other antiemetics are not available .

[see Clinical Pharmacology (12.3)] 7.5 Indinavir Concomitant administration of atovaquone and indinavir did not result in any change in the steady‑state AUC and C of indinavir but resulted in a decrease in the C of indinavir .

Caution should be exercised when prescribing atovaquone with indinavir due to the decrease in trough concentrations of indinavir.

max trough [see Clinical Pharmacology (12.3)]

OVERDOSAGE

10 There is no information on overdoses of MALARONE substantially higher than the doses recommended for treatment.

There is no known antidote for atovaquone, and it is currently unknown if atovaquone is dialyzable.

Overdoses up to 31,500 mg of atovaquone have been reported.

In one such patient who also took an unspecified dose of dapsone, methemoglobinemia occurred.

Rash has also been reported after overdose.

Overdoses of proguanil hydrochloride as large as 1,500 mg have been followed by complete recovery, and doses as high as 700 mg twice daily have been taken for over 2 weeks without serious toxicity.

Adverse experiences occasionally associated with proguanil hydrochloride doses of 100 to 200 mg/day, such as epigastric discomfort and vomiting, would be likely to occur with overdose.

There are also reports of reversible hair loss and scaling of the skin on the palms and/or soles, reversible aphthous ulceration, and hematologic side effects.

DESCRIPTION

11 MALARONE (atovaquone and proguanil hydrochloride) Tablets (adult strength) and MALARONE (atovaquone and proguanil hydrochloride) Pediatric Tablets, for oral administration, contain a fixed‑dose combination of the antimalarial agents atovaquone and proguanil hydrochloride.

The chemical name of atovaquone is -2-[4-(4-chlorophenyl)cyclohexyl]-3-hydroxy-1,4-naphthalenedione.

Atovaquone is a yellow crystalline solid that is practically insoluble in water.

It has a molecular weight of 366.84 and the molecular formula C H ClO .

The compound has the following structural formula: trans 22 19 3 The chemical name of proguanil hydrochloride is 1-(4-chlorophenyl)-5-isopropyl-biguanide hydrochloride.

Proguanil hydrochloride is a white crystalline solid that is sparingly soluble in water.

It has a molecular weight of 290.22 and the molecular formula C H ClN •HCl.

The compound has the following structural formula: 11 16 5 Each MALARONE Tablet (adult strength) contains 250 mg of atovaquone and 100 mg of proguanil hydrochloride and each MALARONE Pediatric Tablet contains 62.5 mg of atovaquone and 25 mg of proguanil hydrochloride.

The inactive ingredients in both tablets are low‑substituted hydroxypropyl cellulose, magnesium stearate, microcrystalline cellulose, poloxamer 188, povidone K30, and sodium starch glycolate.

The tablet coating contains hypromellose, polyethylene glycol 400, polyethylene glycol 8000, red iron oxide, and titanium dioxide.

atovaquone molecular structure proguanil hydrochloride molecular structure

CLINICAL STUDIES

14 14.1 Prevention of Malaria P.

falciparum MALARONE was evaluated for prophylaxis of malaria in 5 clinical trials in malaria‑endemic areas and in 3 active‑controlled trials in non‑immune travelers to malaria‑endemic areas.

P.

falciparum Three placebo‑controlled trials of 10 to 12 weeks’ duration were conducted among residents of malaria‑endemic areas in Kenya, Zambia, and Gabon.

The mean age of subjects was 30 (range 17‑55), 32 (range 16‑64), and 10 (range 5‑16) years, respectively.

Of a total of 669 randomized patients (including 264 pediatric patients 5 to 16 years of age), 103 were withdrawn for reasons other than falciparum malaria or drug‑related adverse events (55% of these were lost to follow‑up and 45% were withdrawn for protocol violations).

The results are listed in Table 6.

Table 6.

Prevention of Parasitemia in Placebo Controlled Clinical Trials of MALARONE for Prophylaxis of P.

falciparum Malaria in Residents of Malaria Endemic Areas a MALARONE Placebo Total number of patients randomized 326 343 Failed to complete study 57 46 Developed parasitemia ( ) P.

falciparum 2 92 Free of parasitemia during the 10 to 12-week period of prophylactic therapy.

a In another study, 330 Gabonese pediatric patients (weighing 13 to 40 kg, and aged 4 to 14 years) who had received successful open‑label radical cure treatment with artesunate, were randomized to receive either MALARONE (dosage based on body weight) or placebo in a double‑blind fashion for 12 weeks.

Blood smears were obtained weekly and any time malaria was suspected.

Nineteen of the 165 children given MALARONE and 18 of 165 patients given placebo withdrew from the study for reasons other than parasitemia (primary reason was lost to follow-up).

One out of 150 evaluable patients (<1%) who received MALARONE developed parasitemia while receiving prophylaxis with MALARONE compared with 31 (22%) of the 144 evaluable placebo recipients.

P.

falciparum In a 10‑week study in 175 South African subjects who moved into malaria‑endemic areas and were given prophylaxis with 1 MALARONE Tablet daily, parasitemia developed in 1 subject who missed several doses of medication.

Since no placebo control was included, the incidence of malaria in this study was not known.

Two active-controlled trials were conducted in non‑immune travelers who visited a malaria‑endemic area.

The mean duration of travel was 18 days (range 2 to 38 days).

Of a total of 1,998 randomized patients who received MALARONE or controlled drug, 24 discontinued from the study before follow-up evaluation 60 days after leaving the endemic area.

Nine of these were lost to follow-up, 2 withdrew because of an adverse experience, and 13 were discontinued for other reasons.

These trials were not large enough to allow for statements of comparative efficacy.

In addition, the true exposure rate to malaria in both trials is unknown.

The results are listed in Table 7.

P.

falciparum Table 7.

Prevention of Parasitemia in Active-Controlled Clinical Trials of MALARONE for Prophylaxis of P.

falciparum Malaria in Non-Immune Travelers a MALARONE Mefloquine Chloroquine plus Proguanil Total number of randomized patients who received study drug 1,004 483 511 Failed to complete study 14 6 4 Developed parasitemia ( ) P.

falciparum 0 0 3 Free of parasitemia during the period of prophylactic therapy.

a A third randomized, open‑label study was conducted which included 221 otherwise healthy pediatric patients (weighing ≥11 kg and 2 to 17 years of age) who were at risk of contracting malaria by traveling to an endemic area.

The mean duration of travel was 15 days (range 1 to 30 days).

Prophylaxis with MALARONE (n = 110, dosage based on body weight) began 1 or 2 days before entering the endemic area and lasted until 7 days after leaving the area.

A control group (n = 111) received prophylaxis with chloroquine/proguanil dosed according to WHO guidelines.

No cases of malaria occurred in either group of children.

However, the study was not large enough to allow for statements of comparative efficacy.

In addition, the true exposure rate to malaria in this study is unknown.

P.

falciparum In separate trials with small numbers of volunteers, atovaquone and proguanil hydrochloride were independently shown to have causal prophylactic activity directed against liver‑stage parasites of .

Six patients given a single dose of atovaquone 250 mg 24 hours prior to malaria challenge were protected from developing malaria, whereas all 4 placebo‑treated patients developed malaria.

Causal Prophylaxis: P.

falciparum During the 4 weeks following cessation of prophylaxis in clinical trial participants who remained in malaria‑endemic areas and were available for evaluation, malaria developed in 24 of 211 (11.4%) subjects who took placebo and 9 of 328 (2.7%) who took MALARONE.

While new infections could not be distinguished from recrudescent infections, all but 1 of the infections in patients treated with MALARONE occurred more than 15 days after stopping therapy.

The single case occurring on day 8 following cessation of therapy with MALARONE probably represents a failure of prophylaxis with MALARONE.

The possibility that delayed cases of malaria may occur some time after stopping prophylaxis with MALARONE cannot be ruled out.

Hence, returning travelers developing febrile illnesses should be investigated for malaria.

P.

falciparum 14.2 Treatment of Acute, Uncomplicated Malaria Infections P.

falciparum In 3 phase II clinical trials, atovaquone alone, proguanil hydrochloride alone, and the combination of atovaquone and proguanil hydrochloride were evaluated for the treatment of acute, uncomplicated malaria caused by .

Among 156 evaluable patients, the parasitological cure rate (elimination of parasitemia with no recurrent parasitemia during follow‑up for 28 days) was 59/89 (66%) with atovaquone alone, 1/17 (6%) with proguanil hydrochloride alone, and 50/50 (100%) with the combination of atovaquone and proguanil hydrochloride.

P.

falciparum MALARONE was evaluated for treatment of acute, uncomplicated malaria caused by in 8 phase III randomized, open-label, controlled clinical trials (N = 1,030 enrolled in both treatment groups).

The mean age of subjects was 27 years and 16% were children ≤12 years of age; 74% of subjects were male.

Evaluable patients included those whose outcome at 28 days was known.

Among 471 evaluable patients treated with the equivalent of 4 MALARONE Tablets once daily for 3 days, 464 had a sensitive response (elimination of parasitemia with no recurrent parasitemia during follow‑up for 28 days) (Table 8).

Seven patients had a response of RI resistance (elimination of parasitemia but with recurrent parasitemia between 7 and 28 days after starting treatment).

In these trials, the response to treatment with MALARONE was similar to treatment with the comparator drug in 4 trials.

P.

falciparum Table 8.

Parasitological Response in 8 Clinical Trials of MALARONE for Treatment of P.

falciparum Malaria Study Site MALARONE a Comparator Evaluable Patients (n) % Sensitive Response b Drug(s) Evaluable Patients (n) % Sensitive Response b Brazil 74 98.6% Quinine and tetracycline 76 100.0% Thailand 79 100.0% Mefloquine 79 86.1% France c 21 100.0% Halofantrine 18 100.0% Kenya c,d 81 93.8% Halofantrine 83 90.4% Zambia 80 100.0% Pyrimethamine/ sulfadoxine (P/S) 80 98.8% Gabon c 63 98.4% Amodiaquine 63 81.0% Philippines 54 100.0% Chloroquine (Cq) Cq and P/S 23 32 30.4% 87.5% Peru 19 100.0% Chloroquine P/S 13 7 7.7% 100.0% MALARONE = 1,000 mg atovaquone and 400 mg proguanil hydrochloride (or equivalent based on body weight for patients weighing ≤40 kg) once daily for 3 days.

a Elimination of parasitemia with no recurrent parasitemia during follow‑up for 28 days.

b Patients hospitalized only for acute care.

Follow‑up conducted in outpatients.

c Study in pediatric patients 3 to 12 years of age.

d When these 8 trials were pooled and 2 additional trials evaluating MALARONE alone (without a comparator arm) were added to the analysis, the overall efficacy (elimination of parasitemia with no recurrent parasitemia during follow‑up for 28 days) in 521 evaluable patients was 98.7%.

The efficacy of MALARONE in the treatment of the erythrocytic phase of nonfalciparum malaria was assessed in a small number of patients.

Of the 23 patients in Thailand infected with and treated with atovaquone/proguanil hydrochloride 1,000 mg/400 mg daily for 3 days, parasitemia cleared in 21 (91.3%) at 7 days.

Parasite relapse occurred commonly when malaria was treated with MALARONE alone.

Relapsing malarias including and require additional treatment to prevent relapse.

P.

vivax P.

vivax P.

vivax P.

ovale The efficacy of MALARONE in treating acute uncomplicated malaria in children weighing ≥5 and <11 kg was examined in an open‑label, randomized trial conducted in Gabon.

Patients received either MALARONE (2 or 3 MALARONE Pediatric Tablets once daily depending upon body weight) for 3 days (n = 100) or amodiaquine (10 mg/kg/day) for 3 days (n = 100).

In this study, the MALARONE Tablets were crushed and mixed with condensed milk just prior to administration.

An adequate clinical response (elimination of parasitemia with no recurrent parasitemia during follow‑up for 28 days) was obtained in 95% (87/92) of the evaluable pediatric patients who received MALARONE and in 53% (41/78) of those evaluable who received amodiaquine.

A response of RI resistance (elimination of parasitemia but with recurrent parasitemia between 7 and 28 days after starting treatment) was noted in 3% and 40% of the patients, respectively.

Two cases of RIII resistance (rising parasite count despite therapy) were reported in the patients receiving MALARONE.

There were 4 cases of RIII in the amodiaquine arm.

P.

falciparum

HOW SUPPLIED

16 /STORAGE AND HANDLING NDC:54569-5762-0 in a BOTTLE of 12 TABLET, FILM COATEDS

GERIATRIC USE

8.5 Geriatric Use Clinical trials of MALARONE did not include sufficient numbers of subjects aged 65 years and older to determine whether they respond differently from younger subjects.

In general, dose selection for an elderly patient should be cautious, reflecting the greater frequency of decreased hepatic, renal, or cardiac function, the higher systemic exposure to cycloguanil, and the greater frequency of concomitant disease or other drug therapy.

[See Clinical Pharmacology (12.3).]

DOSAGE FORMS AND STRENGTHS

3 Each MALARONE Tablet (adult strength) contains 250 mg atovaquone and 100 mg proguanil hydrochloride.

MALARONE Tablets are pink, film‑coated, round, biconvex tablets engraved with “GX CM3” on one side.

Each MALARONE Pediatric Tablet contains 62.5 mg atovaquone and 25 mg proguanil hydrochloride.

MALARONE Pediatric Tablets are pink, film‑coated, round, biconvex tablets engraved with “GX CG7” on one side.

Tablets (adult strength): 250 mg atovaquone and 100 mg proguanil hydrochloride.

( ) • 3 Pediatric Tablets: 62.5 mg atovaquone and 25 mg proguanil hydrochloride.

( ) • 3

MECHANISM OF ACTION

12.1 Mechanism of Action The constituents of MALARONE, atovaquone and proguanil hydrochloride, interfere with 2 different pathways involved in the biosynthesis of pyrimidines required for nucleic acid replication.

Atovaquone is a selective inhibitor of parasite mitochondrial electron transport.

Proguanil hydrochloride primarily exerts its effect by means of the metabolite cycloguanil, a dihydrofolate reductase inhibitor.

Inhibition of dihydrofolate reductase in the malaria parasite disrupts deoxythymidylate synthesis.

INDICATIONS AND USAGE

1 MALARONE is an antimalarial indicated for: prophylaxis of malaria, including in areas where chloroquine resistance has been reported.

( ) • Plasmodium falciparum 1.1 treatment of acute, uncomplicated malaria.

( ) • P.

falciparum 1.2 1.2 Treatment of Malaria MALARONE is indicated for the treatment of acute, uncomplicated malaria.

MALARONE has been shown to be effective in regions where the drugs chloroquine, halofantrine, mefloquine, and amodiaquine may have unacceptable failure rates, presumably due to drug resistance.

P.

falciparum

PEDIATRIC USE

8.4 Pediatric Use Safety and effectiveness have not been established in pediatric patients who weigh less than 11 kg.

The efficacy and safety of MALARONE have been established for the prophylaxis of malaria in controlled trials involving pediatric patients weighing 11 kg or more .

Prophylaxis of Malaria: [see Clinical Studies (14.1)] Safety and effectiveness have not been established in pediatric patients who weigh less than 5 kg.

The efficacy and safety of MALARONE for the treatment of malaria have been established in controlled trials involving pediatric patients weighing 5 kg or more .

Treatment of Malaria: [see Clinical Studies (14.2)]

PREGNANCY

8.1 Pregnancy Pregnancy Category C Atovaquone was not teratogenic and did not cause reproductive toxicity in rats at doses up to 1,000 mg/kg/day corresponding to maternal plasma concentrations up to 7.3 times the estimated human exposure during treatment of malaria based on AUC.

In rabbits, atovaquone caused adverse fetal effects and maternal toxicity at a dose of 1,200 mg/kg/day corresponding to plasma concentrations that were approximately 1.3 times the estimated human exposure during treatment of malaria based on AUC.

Adverse fetal effects in rabbits, including decreased fetal body lengths and increased early resorptions and post-implantation losses, were observed only in the presence of maternal toxicity.

Atovaquone: In a pre- and post-natal study in rats, atovaquone did not produce adverse effects in offspring at doses up to 1,000 mg/kg/day corresponding to AUC exposures of approximately 7.3 times the estimated human exposure during treatment of malaria.

A pre- and post-natal study in Sprague-Dawley rats revealed no adverse effects at doses up to 16 mg/kg/day of proguanil hydrochloride (up to 0.04-times the average human exposure based on AUC).

Pre- and post-natal studies of proguanil in animals at exposures similar to or greater than those observed in humans have not been conducted.

Proguanil: The combination of atovaquone and proguanil hydrochloride was not teratogenic in pregnant rats at atovaquone:proguanil hydrochloride (50:20 mg/kg/day) corresponding to plasma concentrations up to 1.7 and 0.1 times, respectively, the estimated human exposure during treatment of malaria based on AUC.

In pregnant rabbits, the combination of atovaquone and proguanil hydrochloride was not teratogenic or embryotoxic to rabbit fetuses at atovaquone:proguanil hydrochloride (100:40 mg/kg/day) corresponding to plasma concentrations of approximately 0.3 and 0.5 times, respectively, the estimated human exposure during treatment of malaria based on AUC.

Atovaquone and Proguanil: There are no adequate and well‑controlled studies of atovaquone and/or proguanil hydrochloride in pregnant women.

MALARONE should be used during pregnancy only if the potential benefit justifies the potential risk to the fetus.

Falciparum malaria carries a higher risk of morbidity and mortality in pregnant women than in the general population.

Maternal death and fetal loss are both known complications of falciparum malaria in pregnancy.

In pregnant women who must travel to malaria‑endemic areas, personal protection against mosquito bites should always be employed in addition to antimalarials.

[See Patient Counseling Information (17).] The proguanil component of MALARONE acts by inhibiting the parasitic dihydrofolate reductase .

However, there are no clinical data indicating that folate supplementation diminishes drug efficacy.

For women of childbearing age receiving folate supplements to prevent neural tube birth defects, such supplements may be continued while taking MALARONE.

[see Clinical Pharmacology (12.1)]

NUSRING MOTHERS

8.3 Nursing Mothers It is not known whether atovaquone is excreted into human milk.

In a rat study, atovaquone concentrations in the milk were 30% of the concurrent atovaquone concentrations in the maternal plasma.

Proguanil is excreted into human milk in small quantities.

Caution should be exercised when MALARONE is administered to a nursing woman.

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS Atovaquone absorption may be reduced in patients with diarrhea or vomiting.

If used in patients who are vomiting, parasitemia should be closely monitored and the use of an antiemetic considered.

In patients with severe or persistent diarrhea or vomiting, alternative antimalarial therapy may be required.

( ) • 5.1 In mixed and infection, relapse occurred commonly when patients were treated with MALARONE alone.

( ) • P.

falciparum Plasmodium vivax P.

vivax 5.2 In the event of recrudescent infections after treatment or prophylaxis failure, patients should be treated with a different blood schizonticide.

( ) • P.

falciparum 5.2 Elevated liver laboratory tests and cases of hepatitis and hepatic failure requiring liver transplantation have been reported with prophylactic use.

( ) • 5.3 MALARONE has not been evaluated for the treatment of cerebral malaria or other severe manifestations of complicated malaria.

Patients with severe malaria are not candidates for oral therapy.

( ) • 5.4 5.1 Vomiting and Diarrhea Absorption of atovaquone may be reduced in patients with diarrhea or vomiting.

If MALARONE is used in patients who are vomiting, parasitemia should be closely monitored and the use of an antiemetic considered.

Vomiting occurred in up to 19% of pediatric patients given treatment doses of MALARONE.

In the controlled clinical trials, 15.3% of adults received an antiemetic when they received atovaquone/proguanil and 98.3% of these patients were successfully treated.

In patients with severe or persistent diarrhea or vomiting, alternative antimalarial therapy may be required.

[See Dosage and Administration (2).] 5.2 Relapse of Infection In mixed and infections, parasite relapse occurred commonly when patients were treated with MALARONE alone.

P.

falciparum Plasmodium vivax P.

vivax In the event of recrudescent infections after treatment with MALARONE or failure of chemoprophylaxis with MALARONE, patients should be treated with a different blood schizonticide.

P.

falciparum 5.3 Hepatotoxicity Elevated liver laboratory tests and cases of hepatitis and hepatic failure requiring liver transplantation have been reported with prophylactic use of MALARONE.

5.4 Severe or Complicated Malaria MALARONE has not been evaluated for the treatment of cerebral malaria or other severe manifestations of complicated malaria, including hyperparasitemia, pulmonary edema, or renal failure.

Patients with severe malaria are not candidates for oral therapy.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION Patients should be instructed: to take MALARONE at the same time each day with food or a milky drink.

• to take a repeat dose of MALARONE if vomiting occurs within 1 hour after dosing.

• to take a dose as soon as possible if a dose is missed, then return to their normal dosing schedule.

However, if a dose is skipped, the patient should not double the next dose.

• that rare serious adverse events such as hepatitis, severe skin reactions, neurological, and hematological events have been reported when MALARONE was used for the prophylaxis or treatment of malaria.

• to consult a healthcare professional regarding alternative forms of prophylaxis if prophylaxis with MALARONE is prematurely discontinued for any reason.

• that protective clothing, insect repellents, and bednets are important components of malaria prophylaxis.

• that no chemoprophylactic regimen is 100% effective; therefore, patients should seek medical attention for any febrile illness that occurs during or after return from a malaria‑endemic area and inform their healthcare professional that they may have been exposed to malaria.

• that falciparum malaria carries a higher risk of death and serious complications in pregnant women than in the general population.

Pregnant women anticipating travel to malarious areas should discuss the risks and benefits of such travel with their physicians.

• GlaxoSmithKline Research Triangle Park, NC 27709 ©2013, GlaxoSmithKline.

All rights reserved.

MLR:6PI

DOSAGE AND ADMINISTRATION

2 The daily dose should be taken at the same time each day with food or a milky drink.

In the event of vomiting within 1 hour after dosing, a repeat dose should be taken.

MALARONE may be crushed and mixed with condensed milk just prior to administration to patients who may have difficulty swallowing tablets.

MALARONE should be taken with food or a milky drink.

• Prophylaxis ( ): 2.1 Start prophylaxis 1 or 2 days before entering a malaria‑endemic area and continue daily during the stay and for 7 days after return.

• Adults: One adult strength tablet per day.

• Pediatric Patients: Dosage based on body weight (see Table 1).

• Treatment ( ): 2.2 Adults: Four adult strength tablets as a single daily dose for 3 days.

• Pediatric Patients: Dosage based on body weight (see Table 2).

• Renal Impairment ): (2.3 Do not use for prophylaxis of malaria in patients with severe renal impairment.

• Use with caution for treatment of malaria in patients with severe renal impairment.

• 2.1 Prevention of Malaria Start prophylactic treatment with MALARONE 1 or 2 days before entering a malaria‑endemic area and continue daily during the stay and for 7 days after return.

One MALARONE Tablet (adult strength = 250 mg atovaquone/100 mg proguanil hydrochloride) per day.

Adults: The dosage for prevention of malaria in pediatric patients is based upon body weight (Table 1).

Pediatric Patients: Table 1.

Dosage for Prevention of Malaria in Pediatric Patients Weight (kg) Atovaquone/ Proguanil HCl Total Daily Dose Dosage Regimen 11-20 62.5 mg/25 mg 1 MALARONE Pediatric Tablet daily 21-30 125 mg/50 mg 2 MALARONE Pediatric Tablets as a single daily dose 31-40 187.5 mg/75 mg 3 MALARONE Pediatric Tablets as a single daily dose >40 250 mg/100 mg 1 MALARONE Tablet (adult strength) as a single daily dose 2.2 Treatment of Acute Malaria Four MALARONE Tablets (adult strength; total daily dose 1 g atovaquone/400 mg proguanil hydrochloride) as a single daily dose for 3 consecutive days.

Adults: The dosage for treatment of acute malaria in pediatric patients is based upon body weight (Table 2).

Pediatric Patients: Table 2.

Dosage for Treatment of Acute Malaria in Pediatric Patients Weight (kg) Atovaquone/ Proguanil HCl Total Daily Dose Dosage Regimen 5-8 125 mg/50 mg 2 MALARONE Pediatric Tablets daily for 3 consecutive days 9-10 187.5 mg/75 mg 3 MALARONE Pediatric Tablets daily for 3 consecutive days 11-20 250 mg/100 mg 1 MALARONE Tablet (adult strength) daily for 3 consecutive days 21-30 500 mg/200 mg 2 MALARONE Tablets (adult strength) as a single daily dose for 3 consecutive days 31-40 750 mg/300 mg 3 MALARONE Tablets (adult strength) as a single daily dose for 3 consecutive days >40 1 g/400 mg 4 MALARONE Tablets (adult strength) as a single daily dose for 3 consecutive days 2.3 Renal Impairment Do not use MALARONE for malaria prophylaxis in patients with severe renal impairment (creatinine clearance <30 mL/min) .

Use with caution for the treatment of malaria in patients with severe renal impairment, only if the benefits of the 3-day treatment regimen outweigh the potential risks associated with increased drug exposure.

No dosage adjustments are needed in patients with mild (creatinine clearance 50 to 80 mL/min) or moderate (creatinine clearance 30 to 50 mL/min) renal impairment.

[see Contraindications (4.2)] [See Clinical Pharmacology (12.3).]

topiramate 200 MG Oral Tablet

WARNINGS

Acute Myopia and Secondary Angle Closure Glaucoma A syndrome consisting of acute myopia associated with secondary angle closure glaucoma has been reported in patients receiving topiramate.

Symptoms include acute onset of decreased visual acuity and/or ocular pain.

Ophthalmologic findings can include myopia, anterior chamber shallowing, ocular hyperemia (redness) and increased intraocular pressure.

Mydriasis may or may not be present.

This syndrome may be associated with supraciliary effusion resulting in anterior displacement of the lens and iris, with secondary angle closure glaucoma.

Symptoms typically occur within 1 month of initiating topiramate therapy.

In contrast to primary narrow angle glaucoma, which is rare under 40 years of age, secondary angle closure glaucoma associated with topiramate has been reported in pediatric patients as well as adults.

The primary treatment to reverse symptoms is discontinuation of topiramate as rapidly as possible, according to the judgment of the treating physician.

Other measures, in conjunction with discontinuation of topiramate, may be helpful.

Elevated intraocular pressure of any etiology, if left untreated, can lead to serious sequelae including permanent vision loss.

Oligohidrosis and Hyperthermia Oligohidrosis (decreased sweating), infrequently resulting in hospitalization, has been reported in association with topiramate use.

Decreased sweating and an elevation in body temperature above normal characterized these cases.

Some of the cases were reported after exposure to elevated environmental temperatures.

The majority of the reports have been in children.

Patients, especially pediatric patients, treated with topiramate should be monitored closely for evidence of decreased sweating and increased body temperature, especially in hot weather.

Caution should be used when topiramate is prescribed with other drugs that predispose patients to heat-related disorders; these drugs include, but are not limited to, other carbonic anhydrase inhibitors and drugs with anticholinergic activity.

Suicidal Behavior and Ideation Antiepileptic drugs (AEDs), including topiramate, increase the risk of suicidal thoughts or behavior in patients taking these drugs for any indication.

Patients treated with any AED for any indication should be monitored for the emergence or worsening of depression, suicidal thoughts or behavior, and/or any unusual changes in mood or behavior.

Pooled analyses of 199 placebo-controlled clinical trials (mono- and adjunctive therapy) of 11 different AEDs showed that patients randomized to one of the AEDs had approximately twice the risk (adjusted Relative Risk 1.8, 95% CI:1.2, 2.7) of suicidal thinking or behavior compared to patients randomized to placebo.

In these trials, which had a median treatment duration of 12 weeks, the estimated incidence rate of suicidal behavior or ideation among 27,863 AED-treated patients was 0.43%, compared to 0.24% among 16,029 placebo-treated patients, representing an increase of approximately one case of suicidal thinking or behavior for every 530 patients treated.

There were four suicides in drug-treated patients in the trials and none in placebo-treated patients, but the number is too small to allow any conclusion about drug effect on suicide.

The increased risk of suicidal thoughts or behavior with AEDs was observed as early as one week after starting drug treatment with AEDs and persisted for the duration of treatment assessed.

Because most trials included in the analysis did not extend beyond 24 weeks, the risk of suicidal thoughts or behavior beyond 24 weeks could not be assessed.

The risk of suicidal thoughts or behavior was generally consistent among drugs in the data analyzed.

The finding of increased risk with AEDs of varying mechanisms of action and across a range of indications suggests that the risk applies to all AEDs used for any indication.

The risk did not vary substantially by age (5 to 100 years) in the clinical trials analyzed.

Table 3 shows absolute and relative risk by indication for all evaluated AEDs.

Table 3 Risk by Indication for Antiepileptic Drugs in the Pooled Analysis Indication Placebo Patients with Events Per 1000 Patients Drug Patients with Events Per 1000 Patients Relative Risk: Incidence of Events in Drug Patients/Incidence in Placebo Patients Risk Difference: Additional Drug Patients with Events Per 1000 Patients Epilepsy 1.0 3.4 3.5 2.4 Psychiatric 5.7 8.5 1.5 2.9 Other 1.0 1.8 1.9 0.9 Total 2.4 4.3 1.8 1.9 The relative risk for suicidal thoughts or behavior was higher in clinical trials for epilepsy than in clinical trials for psychiatric or other conditions, but the absolute risk differences were similar for the epilepsy and psychiatric indications.

Anyone considering prescribing topiramate or any other AED must balance the risk of suicidal thoughts or behavior with the risk of untreated illness.

Epilepsy and many other illnesses for which AEDs are prescribed are themselves associated with morbidity and mortality and an increased risk of suicidal thoughts and behavior.

Should suicidal thoughts and behavior emerge during treatment, the prescriber needs to consider whether the emergence of these symptoms in any given patient may be related to the illness being treated.

Patients, their caregivers, and families should be informed that AEDs increase the risk of suicidal thoughts and behavior and should be advised of the need to be alert for the emergence or worsening of the signs and symptoms of depression, any unusual changes in mood or behavior or the emergence of suicidal thoughts, behavior or thoughts about self-harm.

Behaviors of concern should be reported immediately to healthcare providers.

Metabolic Acidosis Hyperchloremic, non-anion gap, metabolic acidosis (i.e., decreased serum bicarbonate below the normal reference range in the absence of chronic respiratory alkalosis) is associated with topiramate treatment.

This metabolic acidosis is caused by renal bicarbonate loss due to the inhibitory effect of topiramate on carbonic anhydrase.

Such electrolyte imbalance has been observed with the use of topiramate in placebo-controlled clinical trials and in the post-marketing period.

Generally, topiramate-induced metabolic acidosis occurs early in treatment although cases can occur at any time during treatment.

Bicarbonate decrements are usually mild-moderate (average decrease of 4 mEq/L at daily doses of 400 mg in adults and at approximately 6 mg/kg/day in pediatric patients); rarely, patients can experience severe decrements to values below 10 mEq/L.

Conditions or therapies that predispose to acidosis (such as renal disease, severe respiratory disorders, status epilepticus, diarrhea, surgery, ketogenic diet, or drugs) may be additive to the bicarbonate lowering effects of topiramate.

In adults, the incidence of persistent treatment-emergent decreases in serum bicarbonate (levels of less than 20 mEq/L at two consecutive visits or at the final visit) in controlled clinical trials for adjunctive treatment of epilepsy was 32% for 400 mg/day, and 1% for placebo.

Metabolic acidosis has been observed at doses as low as 50 mg/day.

The incidence of persistent treatment-emergent decreases in serum bicarbonate in adults in the epilepsy controlled clinical trial for monotherapy was 15% for 50 mg/day and 25% for 400 mg/day.

The incidence of a markedly abnormally low serum bicarbonate (i.e., absolute value less than 17 mEq/L and greater than 5 mEq/L decrease from pretreatment) in the adjunctive therapy trials was 3% for 400 mg/day, and 0% for placebo and in the monotherapy trial was 1% for 50 mg/day and 7% for 400 mg/day.

Serum bicarbonate levels have not been systematically evaluated at daily doses greater than 400 mg/day.

In pediatric patients (less than 16 years of age), the incidence of persistent treatment-emergent decreases in serum bicarbonate in placebo-controlled trials for adjunctive treatment of Lennox-Gastaut syndrome or refractory partial onset seizures was 67% for topiramate (at approximately 6 mg/kg/day), and 10% for placebo.

The incidence of a markedly abnormally low serum bicarbonate (i.e., absolute value less than 17 mEq/L and greater than 5 mEq/L decrease from pretreatment) in these trials was 11% for topiramate and 0% for placebo.

Cases of moderately severe metabolic acidosis have been reported in patients as young as 5 months old, especially at daily doses above 5 mg/kg/day.

In pediatric patients (10 years up to 16 years of age), the incidence of persistent treatment-emergent decreases in serum bicarbonate in the epilepsy controlled clinical trial for monotherapy was 7% for 50 mg/day and 20% for 400 mg/day.

The incidence of a markedly abnormally low serum bicarbonate (i.e., absolute value less than 17 mEq/L and greater than 5 mEq/L decrease from pretreatment) in this trial was 4% for 50 mg/day and 4% for 400 mg/day.

Some manifestations of acute or chronic metabolic acidosis may include hyperventilation, nonspecific symptoms such as fatigue and anorexia, or more severe sequelae including cardiac arrhythmias or stupor.

Chronic, untreated metabolic acidosis may increase the risk for nephrolithiasis or nephrocalcinosis, and may also result in osteomalacia (referred to as rickets in pediatric patients) and/or osteoporosis with an increased risk for fractures.

Chronic metabolic acidosis in pediatric patients may also reduce growth rates.

A reduction in growth rate may eventually decrease the maximal height achieved.

The effect of topiramate on growth and bone-related sequelae has not been systematically investigated.

Measurement of baseline and periodic serum bicarbonate during topiramate treatment is recommended.

If metabolic acidosis develops and persists, consideration should be given to reducing the dose or discontinuing topiramate (using dose tapering).

If the decision is made to continue patients on topiramate in the face of persistent acidosis, alkali treatment should be considered.

Cognitive/Neuropsychiatric Adverse Events Adults Adverse events most often associated with the use of topiramate were related to the central nervous system and were observed in the epilepsy population.

In adults, the most frequent of these can be classified into three general categories: 1) Cognitive-related dysfunction (e.g.

confusion, psychomotor slowing, difficulty with concentration/attention, difficulty with memory, speech or language problems, particularly word-finding difficulties); 2) Psychiatric/behavioral disturbances (e.g.

depression or mood problems); and 3) Somnolence or fatigue.

Cognitive-Related Dysfunction The majority of cognitive-related adverse events were mild to moderate in severity, and they frequently occurred in isolation.

Rapid titration rate and higher initial dose were associated with higher incidences of these events.

Many of these events contributed to withdrawal from treatment [see ADVERSE REACTIONS , Table 5 and Table 7 ].

In the original add-on epilepsy controlled trials (using rapid titration such as 100 to 200 mg/day weekly increments), the proportion of patients who experienced one or more cognitive-related adverse events was 42% for 200 mg/day, 41% for 400 mg/day, 52% for 600 mg/day, 56% for 800 and 1000 mg/day, and 14% for placebo.

These dose-related adverse reactions began with a similar frequency in the titration or in the maintenance phase, although in some patients the events began during titration and persisted into the maintenance phase.

Some patients who experienced one or more cognitive-related adverse events in the titration phase had a dose-related recurrence of these events in the maintenance phase.

In the monotherapy epilepsy controlled trial, the proportion of patients who experienced one or more cognitive-related adverse events was 19% for topiramate 50 mg/day and 26% for 400 mg/day.

Psychiatric/Behavioral Disturbances Psychiatric/behavioral disturbances (depression or mood problems) were dose-related for the epilepsy population.

Somnolence/Fatigue Somnolence and fatigue were the adverse events most frequently reported during clinical trials of topiramate for adjunctive epilepsy.

For the adjunctive epilepsy population, the incidence of somnolence did not differ substantially between 200 mg/day and 1000 mg/day, but the incidence of fatigue was dose-related and increased at dosages above 400 mg/day.

For the monotherapy epilepsy population in the 50 mg/day and 400 mg/day groups, the incidence of somnolence was dose-related (9% for the 50 mg/day group and 15% for the 400 mg/day group) and the incidence of fatigue was comparable in both treatment groups (14% each).

Additional nonspecific CNS events commonly observed with topiramate in the add-on epilepsy population include dizziness or ataxia.

Pediatric Patients In double-blind adjunctive therapy and monotherapy epilepsy clinical studies, the incidences of cognitive/neuropsychiatric adverse events in pediatric patients were generally lower than observed in adults.

These events included psychomotor slowing, difficulty with concentration/attention, speech disorders/related speech problems and language problems.

The most frequently reported neuropsychiatric events in pediatric patients during adjunctive therapy double-blind studies were somnolence and fatigue.

The most frequently reported neuropsychiatric events in pediatric patients in the 50 mg/day and 400 mg/day groups during the monotherapy double-blind study were headache, dizziness, anorexia, and somnolence.

No patients discontinued treatment due to any adverse events in the adjunctive epilepsy double-blind trials.

In the monotherapy epilepsy double-blind trial, 1 pediatric patient (2%) in the 50 mg/day group and 7 pediatric patients (12%) in the 400 mg/day group discontinued treatment due to any adverse events.

The most common adverse event associated with discontinuation of therapy was difficulty with concentration/attention; all occurred in the 400 mg/day group.

Withdrawal of AEDs Antiepileptic drugs, including topiramate, should be withdrawn gradually to minimize the potential of increased seizure frequency.

Sudden Unexplained Death in Epilepsy (SUDEP) During the course of premarketing development of topiramate tablets, 10 sudden and unexplained deaths were recorded among a cohort of treated patients (2,796 subject years of exposure).

This represents an incidence of 0.0035 deaths per patient year.

Although this rate exceeds that expected in a healthy population matched for age and sex, it is within the range of estimates for the incidence of sudden unexplained deaths in patients with epilepsy not receiving topiramate (ranging from 0.0005 for the general population of patients with epilepsy, to 0.003 for a clinical trial population similar to that in the topiramate program, to 0.005 for patients with refractory epilepsy).

OVERDOSAGE

Overdoses of topiramate have been reported.

Signs and symptoms included convulsions, drowsiness, speech disturbance, blurred vision, diplopia, mentation impaired, lethargy, abnormal coordination, stupor, hypotension, abdominal pain, agitation, dizziness and depression.

The clinical consequences were not severe in most cases, but deaths have been reported after poly-drug overdoses involving topiramate.

Topiramate overdose has resulted in severe metabolic acidosis (see WARNINGS ).

A patient who ingested a dose between 96 and 110 g topiramate was admitted to hospital with coma lasting 20 to 24 hours followed by full recovery after 3 to 4 days.

In acute topiramate overdose, if the ingestion is recent, the stomach should be emptied immediately by lavage or by induction of emesis.

Activated charcoal has been shown to adsorb topiramate in vitro .

Treatment should be appropriately supportive.

Hemodialysis is an effective means of removing topiramate from the body.

DESCRIPTION

Topiramate is a sulfamate-substituted monosaccharide.

Topiramate tablets are available as 25 mg, 50 mg, 100 mg, and 200 mg circular tablets for oral administration.

Topiramate is a white crystalline powder with a bitter taste.

Topiramate USP is most soluble in alkaline solutions containing sodium hydroxide or sodium phosphate and having a pH of 9 to 10.

It is freely soluble in acetone, chloroform, dimethylsulfoxide, and ethanol.

The solubility in water is 9.8 mg/mL.

Its saturated solution has a pH of 6.3.

Topiramate has the molecular formula C 12 H 21 NO 8 S and a molecular weight of 339.37.

Topiramate is designated chemically as 2,3:4,5-Di- O -isopropylidene-β-D-fructopyranose sulfamate and has the following structural formula: Topiramate tablets contain the following inactive ingredients: anhydrous lactose, microcrystalline cellulose, pregelatinized starch, sodium starch glycolate, magnesium stearate, purified water, polyvinyl alcohol, titanium dioxide, polyethylene glycol and talc.

In addition, individual tablets contain: 50 mg tablets: iron oxide yellow 100 mg tablets: iron oxide yellow, and D&C Yellow # 10 Aluminum Lake 200 mg tablets: iron oxide red, lecithin (soya), and iron oxide black image of chemical structure

CLINICAL STUDIES

The studies described in the following sections were conducted using topiramate tablets.

Epilepsy Monotherapy Controlled Trial The effectiveness of topiramate as initial monotherapy in adults and children 10 years of age and older with partial onset or primary generalized seizures was established in a multicenter, randomized, double-blind, parallel-group trial.

The trial was conducted in 487 patients diagnosed with epilepsy (6 to 83 years of age) who had 1 or 2 well-documented seizures during the 3-month retrospective baseline phase who then entered the study and received topiramate 25 mg/day for 7 days in an open-label fashion.

Forty-nine percent of subjects had no prior AED treatment and 17% had a diagnosis of epilepsy for greater than 24 months.

Any AED therapy used for temporary or emergency purposes was discontinued prior to randomization.

In the double-blind phase, 470 patients were randomized to titrate up to 50 mg/day or 400 mg/day.

If the target dose could not be achieved, patients were maintained on the maximum tolerated dose.

Fifty eight percent of patients achieved the maximal dose of 400 mg/day for ≥ 2 weeks, and patients who did not tolerate 150 mg/day were discontinued.

The primary efficacy assessment was a between group comparison of time to first seizure during the double-blind phase.

Comparison of the Kaplan-Meier survival curves of time to first seizure favored the topiramate 400 mg/day group over the topiramate 50 mg/day group (p=0.0002, log rank test; Figure 1 ).

The treatment effects with respect to time to first seizure were consistent across various patient subgroups defined by age, sex, geographic region, baseline body weight, baseline seizure type, time since diagnosis, and baseline AED use.

Figure 1: Kaplan-Meier Estimates of Cumulative Rates for Time to First Seizure Adjunctive Therapy Controlled Trials in Patients With Partial Onset Seizures The effectiveness of topiramate as an adjunctive treatment for adults with partial onset seizures was established in six multicenter, randomized, double-blind, placebo-controlled trials, two comparing several dosages of topiramate and placebo and four comparing a single dosage with placebo, in patients with a history of partial onset seizures, with or without secondarily generalized seizures.

Patients in these studies were permitted a maximum of two antiepileptic drugs (AEDs) in addition to topiramate tablets or placebo.

In each study, patients were stabilized on optimum dosages of their concomitant AEDs during baseline phase lasting between 4 and 12 weeks.

Patients who experienced a prespecified minimum number of partial onset seizures, with or without secondary generalization, during the baseline phase (12 seizures for 12-week baseline, 8 for 8-week baseline, or 3 for 4-week baseline) were randomly assigned to placebo or a specified dose of topiramate tablets in addition to their other AEDs.

Following randomization, patients began the double-blind phase of treatment.

In five of the six studies, patients received active drug beginning at 100 mg per day; the dose was then increased by 100 mg or 200 mg/day increments weekly or every other week until the assigned dose was reached, unless intolerance prevented increases.

In the sixth study (119), the 25 or 50 mg/day initial doses of topiramate were followed by respective weekly increments of 25 or 50 mg/day until the target dose of 200 mg/day was reached.

After titration, patients entered a 4, 8, or 12-week stabilization period.

The numbers of patients randomized to each dose, and the actual mean and median doses in the stabilization period are shown in Table 1 .

Adjunctive Therapy Controlled Trial in Pediatric Patients Ages 2 to 16 Years With Partial Onset Seizures The effectiveness of topiramate as an adjunctive treatment for pediatric patients ages 2 to 16 years with partial onset seizures was established in a multicenter, randomized, double-blind, placebo-controlled trial, comparing topiramate and placebo in patients with a history of partial onset seizures, with or without secondarily generalized seizures.

Patients in this study were permitted a maximum of two antiepileptic drugs (AEDs) in addition to topiramate tablets or placebo.

In this study, patients were stabilized on optimum dosages of their concomitant AEDs during an 8-week baseline phase.

Patients who experienced at least six partial onset seizures, with or without secondarily generalized seizures, during the baseline phase were randomly assigned to placebo or topiramate tablets in addition to their other AEDs.

Following randomization, patients began the double-blind phase of treatment.

Patients received active drug beginning at 25 or 50 mg per day; the dose was then increased by 25 mg to 150 mg/day increments every other week until the assigned dosage of 125, 175, 225, or 400 mg/day based on patients’ weight to approximate a dosage of 6 mg/kg per day was reached, unless intolerance prevented increases.

After titration, patients entered an 8-week stabilization period.

Adjunctive Therapy Controlled Trial in Patients With Primary Generalized Tonic-Clonic Seizures The effectiveness of topiramate as an adjunctive treatment for primary generalized tonic-clonic seizures in patients 2 years old and older was established in a multicenter, randomized, double-blind, placebo-controlled trial, comparing a single dosage of topiramate and placebo.

Patients in this study were permitted a maximum of two antiepileptic drugs (AEDs) in addition to topiramate or placebo.

Patients were stabilized on optimum dosages of their concomitant AEDs during an 8-week baseline phase.

Patients who experienced at least three primary generalized tonic-clonic seizures during the baseline phase were randomly assigned to placebo or topiramate in addition to their other AEDs.

Following randomization, patients began the double-blind phase of treatment.

Patients received active drug beginning at 50 mg per day for four weeks; the dose was then increased by 50 mg to 150 mg/day increments every other week until the assigned dose of 175, 225, or 400 mg/day based on patients’ body weight to approximate a dosage of 6 mg/kg per day was reached, unless intolerance prevented increases.

After titration, patients entered a 12-week stabilization period.

Adjunctive Therapy Controlled Trial in Patients With Lennox-Gastaut Syndrome The effectiveness of topiramate as an adjunctive treatment for seizures associated with Lennox-Gastaut syndrome was established in a multicenter, randomized, double-blind, placebo-controlled trial comparing a single dosage of topiramate with placebo in patients 2 years of age and older.

Patients in this study were permitted a maximum of two antiepileptic drugs (AEDs) in addition to topiramate or placebo.

Patients who were experiencing at least 60 seizures per month before study entry were stabilized on optimum dosages of their concomitant AEDs during a 4-week baseline phase.

Following baseline, patients were randomly assigned to placebo or topiramate in addition to their other AEDs.

Active drug was titrated beginning at 1 mg/kg per day for a week; the dose was then increased to 3 mg/kg per day for one week then to 6 mg/kg per day.

After titration, patients entered an 8-week stabilization period.

The primary measures of effectiveness were the percent reduction in drop attacks and a parental global rating of seizure severity.

Table 1: Topiramate Dose Summary During the Stabilization Periods of Each of Six Double-Blind, Placebo-Controlled, Add-On Trials in Adults with Partial Onset Seizuresb Target Topiramate Dosage (mg/day) Protocol Stabilization Dose Placebo a 200 400 600 800 1,000 YD N Mean Dose Median Dose 42 5.9 6.0 42 200 200 40 390 400 41 556 600 – – – – – – YE N Mean Dose Median Dose 44 9.7 10.0 – – – – – – 40 544 600 45 739 800 40 796 1,000 Y1 N Mean Dose Median Dose 23 3.8 4.0 – – – 19 395 400 – – – – – – – – – Y2 N Mean Dose Median Dose 30 5.7 6.0 – – – – – – 28 522 600 – – – – – – Y3 N Mean Dose Median Dose 28 7.9 8.0 – – – – – – – – – 25 568 600 – – – 119 N Mean Dose Median Dose 90 8 8 157 200 200 – – – – – – – – – – – – a Placebo dosages are given as the number of tablets.

Placebo target dosages were as follows: Protocol Y1, 4 tablets/day; Protocols YD and Y2, 6 tablets/day; Protocol Y3 and 119, 8 tablets/day; Protocol YE, 10 tablets/day.

b Dose-response studies were not conducted for other indications or pediatric partial onset seizure.

In all add-on trials, the reduction in seizure rate from baseline during the entire double-blind phase was measured.

The median percent reductions in seizure rates and the responder rates (fraction of patients with at least a 50% reduction) by treatment group for each study are shown below in Table 2 .

As described above, a global improvement in seizure severity was also assessed in the Lennox-Gastaut trial.

Table 2: Efficacy Results in Double-Blind, Placebo-Controlled, Add-On Epilepsy Trials Target Protocol Efficacy Results Placebo 200 400 600 800 1,000 ≈6 mg/kg/day* Partial Onset Seizures Studies in Adults YD Median % Reduction % Responders N 45 11.6 18 45 27.2 a 24 45 47.5 b 44 d 46 44.7 c 46 d – – – – – – – – – YE Median % Reduction % Responders N 1.7 9 24 – – – – – – 48 40.8 c 40 c 48 41.0 c 41 c 47 36.0 c 36 d – – – Y1 Median % Reduction % Responders N 24 1.1 8 – – – 23 40.7 e 35 d – – – – – – – – – – – – Y2 Median % Reduction % Responders N 30 -12.2 10 – – – – – – 30 46.4 f 47 c – – – – – – – – – Y3 Median % Reduction % Responders N 28 -20.6 0 – – – – – – – – – 28 24.3 c 43 c – – – – – – 119N Median % Reduction % Responders 91 168 20.0 24 – 44.2 c 45 c – – – – – – – – – – – – – – Studies in Pediatric Patients YP Median T Reduction % Responders N 45 10.5 20 – – – – – – – – – – – – – – – 41 33.1 d 39 Primary Generalized Tonic-Clonic h YTC Median % Reduction % Responders N 40 9.0 20 – – – – – – – – – – – – – – – 39 56.7 d 56 c Lennox-Gastuat Syndrome i YL Median % Reduction % Responders N 49 -5.1 14 – – – – – – – – – – – – – – – 46 14.8 d 28 g Improvement in Seizure Severity j 28 – – – – – – 52 d Comparisons with placebo: a p=0.080; b p less than 0.010; c p less than 0.001; d p less than 0.050; e p=0.065; f p less than 0.005; g p=0.071; h Median % reduction and % responders are reported for PGTC Seizures; i Median % reduction and % responders for drop attacks, i.e., tonic or atonic seizures; j Percent of subjects who were minimally, much, or very much improved from baseline * For Protocols YP and YTC, protocol-specified target dosages (less than 9.3 mg/kg/day) were assigned based on subject’s weight to approximate a dosage of 6 mg/kg per day; these dosages corresponded to mg/day dosages of 125, 175, 225, and 400 mg/day.

Subset analyses of the antiepileptic efficacy of topiramate tablets in these studies showed no differences as a function of gender, race, age, baseline seizure rate, or concomitant AED.

image of figure 1

HOW SUPPLIED

Topiramate tablets are available as debossed, film-coated, circular tablets in the following strengths and colors: 25 mg white (coded “S” on one side; “707” on the other) 50 mg yellow (coded “S” on one side; “710” on the other) 100 mg yellow (coded “S” on one side; “711” on the other) 200 mg brown (coded “S” on one side; “712” on the other) They are supplied as follows: 25 mg tablets Bottles of 30 NDC 54868-6016-1 Bottles of 60 NDC 54868-6016-0 Bottles of 90 NDC 54868-6016-2 Bottles of 120 NDC 54868-6016-3 50 mg tablets Bottles of 30 NDC 54868-6017-1 Bottles of 60 NDC 54868-6017-0 Bottles of 90 NDC 54868-6017-2 100 mg tablets Bottles of 30 NDC 54868-6014-1 Bottles of 60 NDC 54868-6014-0 Bottles of 90 NDC 54868-6014-2 200 mg tablets Bottles of 30 NDC 54868-6015-1 Bottles of 60 NDC 54868-6015-0 Store at 20° to 25°C (68° to 77°F); excursions permitted to 15° to 30°C (59° to 86°F) [See USP Controlled Room Temperature].

Protect from moisture.

Dispense in a tight container.

INDICATIONS AND USAGE

Monotherapy Epilepsy Topiramate tablets are indicated as initial monotherapy in patients 10 years of age and older with partial onset or primary generalized tonic-clonic seizures.

Effectiveness was demonstrated in a controlled trial in patients with epilepsy who had no more than 2 seizures in the 3 months prior to enrollment.

Safety and effectiveness in patients who were converted to monotherapy from a previous regimen of other anticonvulsant drugs have not been established in controlled trials.

Adjunctive Therapy Epilepsy Topiramate tablets are indicated as adjunctive therapy for adults and pediatric patients ages 2 to 16 years with partial onset seizures, or primary generalized tonic-clonic seizures, and in patients 2 years of age and older with seizures associated with Lennox-Gastaut syndrome.

DOSAGE AND ADMINISTRATION

Epilepsy In the controlled add-on trials, no correlation has been demonstrated between trough plasma concentrations of topiramate and clinical efficacy.

No evidence of tolerance has been demonstrated in humans.

Doses above 400 mg/day (600, 800, or 1000 mg/day) have not been shown to improve responses in dose-response studies in adults with partial onset seizures.

It is not necessary to monitor topiramate plasma concentrations to optimize topiramate therapy.

On occasion, the addition of topiramate to phenytoin may require an adjustment of the dose of phenytoin to achieve optimal clinical outcome.

Addition or withdrawal of phenytoin and/or carbamazepine during adjunctive therapy with topiramate may require adjustment of the dose of topiramate.

Because of the bitter taste, tablets should not be broken.

Topiramate tablets can be taken without regard to meals.

Monotherapy Use The recommended dose for topiramate monotherapy in adults and children 10 years of age and older is 400 mg/day in two divided doses.

Approximately 58% of patients randomized to 400 mg/day achieved this maximal dose in the monotherapy controlled trial; the mean dose achieved in the trial was 275 mg/day.

The dose should be achieved by titrating according to the following schedule: Morning Dose Evening Dose Week 1 25 mg 25 mg Week 2 50 mg 50 mg Week 3 75 mg 75 mg Week 4 100 mg 100 mg Week 5 150 mg 150 mg Week 6 200 mg 200 mg Adjunctive Therapy Use Adults (17 Years of Age and Over) – Partial Seizures, Primary Generalized Tonic-Clonic Seizures, or Lennox-Gastaut Syndrome The recommended total daily dose of topiramate as adjunctive therapy in adults with partial seizures is 200 to 400 mg/day in two divided doses, and 400 mg/day in two divided doses as adjunctive treatment in adults with primary generalized tonic-clonic seizures.

It is recommended that therapy be initiated at 25 to 50 mg/day followed by titration to an effective dose in increments of 25 to 50 mg/week.

Titrating in increments of 25 mg/week may delay the time to reach an effective dose.

Daily doses above 1,600 mg have not been studied.

In the study of primary generalized tonic-clonic seizures the initial titration rate was slower than in previous studies; the assigned dose was reached at the end of 8 weeks (see CLINICAL STUDIES, Adjunctive Therapy Controlled Trials in Patients With Primary Generalized Tonic-Clonic Seizures ).

Pediatric Patients (Ages 2 to 16 Years)– Partial Seizures, Primary Generalized Tonic-Clonic Seizures, or Lennox-Gastaut Syndrome The recommended total daily dose of topiramate as adjunctive therapy for patients with partial seizures, primary generalized tonic-clonic seizures, or seizures associated with Lennox-Gastaut syndrome is approximately 5 to 9 mg/kg/day in two divided doses.

Titration should begin at 25 mg (or less, based on a range of 1 to 3 mg/kg/day) nightly for the first week.

The dosage should then be increased at 1- or 2-week intervals by increments of 1 to 3 mg/kg/day (administered in two divided doses), to achieve optimal clinical response.

Dose titration should be guided by clinical outcome.

In the study of primary generalized tonic-clonic seizures the initial titration rate was slower than in previous studies; the assigned dose of 6 mg/kg/day was reached at the end of 8 weeks (see CLINICAL STUDIES, Adjunctive Therapy Controlled Trials in Patients With Primary Generalized Tonic-Clonic Seizures ).

Patients with Renal Impairment: In renally impaired subjects (creatinine clearance less than 70 mL/min/1.73 m 2 ), one half of the usual adult dose is recommended.

Such patients will require a longer time to reach steady-state at each dose.

Geriatric Patients (Ages 65 Years and Over): Dosage adjustment may be indicated in the elderly patient when impaired renal function (creatinine clearance rate ≤70 mL/min/1.73 m 2 ) is evident (see : Patients with Renal Impairment and CLINICAL PHARMACOLOGY: Special Populations: Age, Gender, and Race ).

Patients Undergoing Hemodialysis: Topiramate is cleared by hemodialysis at a rate that is 4 to 6 times greater than a normal individual.

Accordingly, a prolonged period of dialysis may cause topiramate concentration to fall below that required to maintain an anti-seizure effect.

To avoid rapid drops in topiramate plasma concentration during hemodialysis, a supplemental dose of topiramate may be required.

The actual adjustment should take into account 1) the duration of dialysis period, 2) the clearance rate of the dialysis system being used, and 3) the effective renal clearance of topiramate in the patient being dialyzed.

Patients with Hepatic Disease: In hepatically impaired patients topiramate plasma concentrations may be increased.

The mechanism is not well understood.

Pyridostigmine Bromide 180 MG Extended Release Oral Tablet

Generic Name: PYRIDOSTIGMINE BROMIDE
Brand Name: Pyridostigmine Bromide
  • Substance Name(s):
  • PYRIDOSTIGMINE BROMIDE

WARNINGS

Although failure of patients to show clinical improvement may reflect underdosage, it can also be indicative of overdosage.

As is true of all cholinergic drugs, overdosage of pyridostigmine bromide may result in cholinergic crisis, a state characterized by increasing muscle weakness which, through involvement of the muscles of respiration, may lead to death.

Myasthenic crisis due to an increase in the severity of the disease is also accompanied by extreme muscle weakness, and thus may be difficult to distinguish from cholinergic crisis on a symptomatic basis.

Such differentiation is extremely important, since increases in doses of pyridostigmine bromide or other drugs of this class in the presence of cholinergic crisis or of a refractory or “insensitive” state could have grave consequences.

Osserman and Genkins 1 indicate that the differential diagnosis of the two types of crisis may require the use of edrophonium chloride as well as clinical judgment.

The treatment of the two conditions obviously differs radically.

Whereas the presence of myasthenic crisis suggests the need for more intensive anticholinesterase therapy, the diagnosis of cholinergic crisis, according to Osserman and Genkins 1 , calls for the prompt withdrawal of all drugs of this type.

The immediate use of atropine in cholinergic crisis is also recommended.

Atropine may also be used to abolish or obtund gastrointestinal side effects or other muscarinic reactions; but such use, by masking signs of overdosage, can lead to inadvertent induction of cholinergic crisis.

For detailed information on the management of patients with myasthenia gravis, the physician is referred to one of the excellent reviews such as those by Osserman and Genkins 2 , Grob 3 or Schwab 4,5 .

Usage in Pregnancy: The safety of pyridostigmine bromide during pregnancy or lactation in humans has not been established.

Therefore, use of pyridostigmine bromide in women who may become pregnant requires weighing the drug’s potential benefits against its possible hazards to mother and child.

DESCRIPTION

Pyridostigmine bromide is an orally active cholinesterase inhibitor.

Chemically, pyridostigmine bromide is 3-hydroxy-1-methylpyridinium bromide dimethylcarbamate.

Its structural formula is: Pyridostigmine bromide extended-release tablets are available as extended-release tablets containing 180 mg pyridostigmine bromide; each tablet also contains carnauba wax, copovidone, lactose, magnesium stearate, and silicon dioxide.

1

HOW SUPPLIED

Pyridostigmine Bromide Extended-Release Tablets, 180 mg are available as light brown to pale yellow, capsule-shaped tablets, debossed with “W1” on one side and single-scored on the other side.

They are supplied as follows: Bottles of 30: NDC 0115-1404-08 Note: Because of the hygroscopic nature of the extended-release tablets, mottling may occur.

This does not affect their efficacy.

Store at 20° to 25°C (68° to 77°F) [see USP Controlled Room Temperature].

Dispense in a tight, light-resistant container.

Keep pyridostigmine bromide extended-release tablets in a dry place with the silica gel enclosed.

INDICATIONS AND USAGE

Pyridostigmine bromide is useful in the treatment of myasthenia gravis.

DOSAGE AND ADMINISTRATION

Pyridostigmine bromide is available in extended-release dosage form: Extended-Release Tablets — each containing 180 mg pyridostigmine bromide.

This form provides uniformly slow release, hence prolonged duration of drug action; it facilitates control of myasthenic symptoms with fewer individual doses daily.

The immediate effect of a 180 mg extended-release tablet is about equal to that of a 60 mg immediate-release tablet; however, its duration of effectiveness, although varying in individual patients, averages 2 1/2 times that of a 60 mg dose.

Dosage: The size and frequency of the dosage must be adjusted to the needs of the individual patient.

Extended-Release Tablets — One to three 180 mg tablets, once or twice daily, will usually be sufficient to control symptoms; however, the needs of certain individuals may vary markedly from this average.

The interval between doses should be at least 6 hours.

For optimum control, it may be necessary to use the more rapidly acting regular tablets or syrup in conjunction with extended-release therapy.

Note: For information on a diagnostic test for myasthenia gravis, and for the evaluation and stabilization of therapy, please see product literature on edrophonium chloride.

lamoTRIgine 50 MG Disintegrating Oral Tablet

Generic Name: LAMOTRIGINE
Brand Name: Lamotrigine
  • Substance Name(s):
  • LAMOTRIGINE

DRUG INTERACTIONS

7 Significant drug interactions with lamotrigine are summarized in this section.

Uridine 5´-diphospho-glucuronyl transferases (UGT) have been identified as the enzymes responsible for metabolism of lamotrigine.

Drugs that induce or inhibit glucuronidation may, therefore, affect the apparent clearance of lamotrigine.

Strong or moderate inducers of the cytochrome P450 3A4 (CYP3A4) enzyme, which are also known to induce UGT, may also enhance the metabolism of lamotrigine.

Those drugs that have been demonstrated to have a clinically significant impact on lamotrigine metabolism are outlined in Table 13.

Specific dosing guidance for these drugs is provided in the Dosage and Administration section [see Dosage and Administration (2.1)] .

Additional details of these drug interaction studies are provided in the Clinical Pharmacology section [see Clinical Pharmacology (12.3)] .

Table 13.

Established and Other Potentially Significant Drug Interactions Concomitant Drug Effect on Concentration of Lamotrigine or Concomitant Drug Clinical Comment Estrogen-containing oral contraceptive preparations containing 30 mcg ethinylestradiol and 150 mcg levonorgestrel ↓ lamotrigine ↓ levonorgestrel Decreased lamotrigine concentrations approximately 50%.

Decrease in levonorgestrel component by 19%.

Carbamazepine and carbamazepine epoxide ↓ lamotrigine ? carbamazepine epoxide Addition of carbamazepine decreases lamotrigine concentration approximately 40%.

May increase carbamazepine epoxide levels.

Lopinavir/ritonavir ↓ lamotrigine Decreased lamotrigine concentration approximately 50%.

Atazanavir/ritonavir ↓ lamotrigine Decreased lamotrigine AUC approximately 32%.

Phenobarbital/Primidone ↓ lamotrigine Decreased lamotrigine concentration approximately 40%.

Phenytoin ↓ lamotrigine Decreased lamotrigine concentration approximately 40%.

Rifampin ↓ lamotrigine Decreased lamotrigine AUC approximately 40%.

Valproate ↑ lamotrigine ? valproate Increased lamotrigine concentrations slightly more than 2-fold.

There are conflicting study results regarding effect of lamotrigine on valproate concentrations: 1) a mean 25% decrease in valproate concentrations in healthy volunteers, 2) no change in valproate concentrations in controlled clinical trials in patients with epilepsy.

↓= Decreased (induces lamotrigine glucuronidation).

↑= Increased (inhibits lamotrigine glucuronidation).

? = Conflicting data.

Effect of lamotrigine on Organic Cationic Transporter 2 Substrates Lamotrigine is an inhibitor of renal tubular secretion via organic cationic transporter 2 (OCT2) proteins [see Clinical Pharmacology (12.3)].

This may result in increased plasma levels of certain drugs that are substantially excreted via this route.

Coadministration of lamotrigine with OCT2 substrates with a narrow therapeutic index (e.g., dofetilide) is not recommended.

Valproate increases lamotrigine concentrations more than 2-fold.

(7, 12.3) Carbamazepine, phenytoin, phenobarbital, primidone, and rifampin decrease lamotrigine concentrations by approximately 40%.

(7, 12.3) Estrogen-containing oral contraceptives decrease lamotrigine concentrations by approximately 50%.

(7, 12.3) Protease inhibitors lopinavir/ritonavir and atazanavir/lopinavir decrease lamotrigine exposure by approximately 50% and 32%, respectively.

(7, 12.3) Coadministration with organic cationic transporter 2 substrates with narrow therapeutic index is not recommended (7, 12.3)

OVERDOSAGE

10 10.1 Human Overdose Experience Overdoses involving quantities up to 15 g have been reported for lamotrigine, some of which have been fatal.

Overdose has resulted in ataxia, nystagmus, seizures (including tonic-clonic seizures), decreased level of consciousness, coma, and intraventricular conduction delay.

10.2 Management of Overdose There are no specific antidotes for lamotrigine.

Following a suspected overdose, hospitalization of the patient is advised.

General supportive care is indicated, including frequent monitoring of vital signs and close observation of the patient.

If indicated, emesis should be induced; usual precautions should be taken to protect the airway.

It should be kept in mind that immediate-release lamotrigine is rapidly absorbed [see Clinical Pharmacology (12.3)].

It is uncertain whether hemodialysis is an effective means of removing lamotrigine from the blood.

In 6 renal failure patients, about 20% of the amount of lamotrigine in the body was removed by hemodialysis during a 4-hour session.

A Poison Control Center should be contacted for information on the management of overdosage of lamotrigine.

DESCRIPTION

11 Lamotrigine, USP an AED of the phenyltriazine class, is chemically unrelated to existing AEDs.

Lamotrigine’s chemical name is 3,5-diamino-6-(2,3-dichlorophenyl)- as -triazine, its molecular formula is C 9 H 7 N 5 Cl 2 , and its molecular weight is 256.09.

Lamotrigine, USP is a white to pale cream-colored powder and has a pK a of 5.7.

Lamotrigine, USP is slightly soluble in 0.1 N hydrochloric acid, in acetone, in methanol and in water.

The structural formula is: Lamotrigine orally disintegrating tablets are supplied for oral administration.

The tablets contain 25 mg (white), 50 mg (white), 100 mg (Peach), 200 mg (White) of lamotrigine, USP and the following inactive ingredients: For lamotrigine orally disintegrating tablets 25 mg, 50 mg and 200 mg: Colloidal silicon dioxide, croscarmellose sodium, magnesium stearate, mannitol, starch (maize), microcrystalline cellulose, pregelatinized starch, peppermint flavor, sodium stearyl fumarate and sucralose.

For lamotrigine orally disintegrating tablets 100 mg: Colloidal silicon dioxide, croscarmellose sodium, magnesium stearate, mannitol, starch (maize), microcrystalline cellulose, pregelatinized starch, peppermint flavor, sodium stearyl fumarate, sucralose and idacol red oxide of iron.

Lamotrigine orally disintegrating tablets are formulated using in-house technologies designed to mask the bitter taste of lamotrigine and achieve a rapid dissolution profile.

lamotrigine

CLINICAL STUDIES

14 14.1 Epilepsy Monotherapy with Lamotrigine in Adults with Partial-Onset Seizures Already Receiving Treatment with Carbamazepine, Phenytoin, Phenobarbital, or Primidone as the Single Antiepileptic Drug The effectiveness of monotherapy with lamotrigine was established in a multicenter, double-blind clinical trial enrolling 156 adult outpatients with partial-onset seizures.

The patients experienced at least 4 simple partial-onset, complex partial-onset, and/or secondarily generalized seizures during each of 2 consecutive 4-week periods while receiving carbamazepine or phenytoin monotherapy during baseline.

Lamotrigine (target dose of 500 mg/day) or valproate (1,000 mg/day) was added to either carbamazepine or phenytoin monotherapy over a 4-week period.

Patients were then converted to monotherapy with lamotrigine or valproate during the next 4 weeks, then continued on monotherapy for an additional 12-week period.

Trial endpoints were completion of all weeks of trial treatment or meeting an escape criterion.

Criteria for escape relative to baseline were: (1) doubling of average monthly seizure count, (2) doubling of highest consecutive 2-day seizure frequency, (3) emergence of a new seizure type (defined as a seizure that did not occur during the 8-week baseline) that is more severe than seizure types that occur during study treatment, or (4) clinically significant prolongation of generalized tonic-clonic seizures.

The primary efficacy variable was the proportion of patients in each treatment group who met escape criteria.

The percentages of patients who met escape criteria were 42% (32/76) in the group receiving lamotrigine and 69% (55/80) in the valproate group.

The difference in the percentage of patients meeting escape criteria was statistically significant ( P = 0.0012) in favor of lamotrigine.

No differences in efficacy based on age, sex, or race were detected.

Patients in the control group were intentionally treated with a relatively low dose of valproate; as such, the sole objective of this trial was to demonstrate the effectiveness and safety of monotherapy with lamotrigine, and cannot be interpreted to imply the superiority of lamotrigine to an adequate dose of valproate.

Adjunctive Therapy with Lamotrigine in Adults with Partial-Onset Seizures The effectiveness of lamotrigine as adjunctive therapy (added to other AEDs) was initially established in 3 pivotal, multicenter, placebo-controlled, double-blind clinical trials in 355 adults with refractory partial-onset seizures.

The patients had a history of at least 4 partial-onset seizures per month in spite of receiving 1 or more AEDs at therapeutic concentrations and in 2 of the trials were observed on their established AED regimen during baselines that varied between 8 to 12 weeks.

In the third trial, patients were not observed in a prospective baseline.

In patients continuing to have at least 4 seizures per month during the baseline, lamotrigine or placebo was then added to the existing therapy.

In all 3 trials, change from baseline in seizure frequency was the primary measure of effectiveness.

The results given below are for all partial-onset seizures in the intent-to-treat population (all patients who received at least 1 dose of treatment) in each trial, unless otherwise indicated.

The median seizure frequency at baseline was 3 per week while the mean at baseline was 6.6 per week for all patients enrolled in efficacy trials.

One trial (n = 216) was a double-blind, placebo-controlled, parallel trial consisting of a 24-week treatment period.

Patients could not be on more than 2 other anticonvulsants and valproate was not allowed.

Patients were randomized to receive placebo, a target dose of 300 mg/day of lamotrigine, or a target dose of 500 mg/day of lamotrigine.

The median reductions in the frequency of all partial-onset seizures relative to baseline were 8% in patients receiving placebo, 20% in patients receiving 300 mg/day of lamotrigine, and 36% in patients receiving 500 mg/day of lamotrigine.

The seizure frequency reduction was statistically significant in the 500-mg/day group compared with the placebo group, but not in the 300-mg/day group.

A second trial (n = 98) was a double-blind, placebo-controlled, randomized, crossover trial consisting of two 14-week treatment periods (the last 2 weeks of which consisted of dose tapering) separated by a 4-week washout period.

Patients could not be on more than 2 other anticonvulsants and valproate was not allowed.

The target dose of lamotrigine was 400 mg/day.

When the first 12 weeks of the treatment periods were analyzed, the median change in seizure frequency was a 25% reduction on lamotrigine compared with placebo ( P <0.001).

The third trial (n = 41) was a double-blind, placebo-controlled, crossover trial consisting of two 12-week treatment periods separated by a 4-week washout period.

Patients could not be on more than 2 other anticonvulsants.

Thirteen patients were on concomitant valproate; these patients received 150 mg/day of lamotrigine.

The 28 other patients had a target dose of 300 mg/day of lamotrigine.

The median change in seizure frequency was a 26% reduction on lamotrigine compared with placebo ( P <0.01).

No differences in efficacy based on age, sex, or race, as measured by change in seizure frequency, were detected.

Adjunctive Therapy with Lamotrigine in Pediatric Patients with Partial-Onset Seizures The effectiveness of lamotrigine as adjunctive therapy in pediatric patients with partial-onset seizures was established in a multicenter, double-blind, placebo-controlled trial in 199 patients aged 2 to 16 years (n = 98 on lamotrigine, n = 101 on placebo).

Following an 8-week baseline phase, patients were randomized to 18 weeks of treatment with lamotrigine or placebo added to their current AED regimen of up to 2 drugs.

Patients were dosed based on body weight and valproate use.

Target doses were designed to approximate 5 mg/kg/day for patients taking valproate (maximum dose: 250 mg/day) and 15 mg/kg/day for the patients not taking valproate (maximum dose: 750 mg/day).

The primary efficacy endpoint was percentage change from baseline in all partial-onset seizures.

For the intent-to-treat population, the median reduction of all partial-onset seizures was 36% in patients treated with lamotrigine and 7% on placebo, a difference that was statistically significant ( P <0.01).

Adjunctive Therapy with Lamotrigine in Pediatric and Adult Patients with Lennox-Gastaut Syndrome The effectiveness of lamotrigine as adjunctive therapy in patients with Lennox-Gastaut syndrome was established in a multicenter, double-blind, placebo-controlled trial in 169 patients aged 3 to 25 years (n = 79 on lamotrigine, n = 90 on placebo).

Following a 4-week, single-blind, placebo phase, patients were randomized to 16 weeks of treatment with lamotrigine or placebo added to their current AED regimen of up to 3 drugs.

Patients were dosed on a fixed-dose regimen based on body weight and valproate use.

Target doses were designed to approximate 5 mg/kg/day for patients taking valproate (maximum dose: 200 mg/day) and 15 mg/kg/day for patients not taking valproate (maximum dose: 400 mg/day).

The primary efficacy endpoint was percentage change from baseline in major motor seizures (atonic, tonic, major myoclonic, and tonic-clonic seizures).

For the intent-to-treat population, the median reduction of major motor seizures was 32% in patients treated with lamotrigine and 9% on placebo, a difference that was statistically significant ( P <0.05).

Drop attacks were significantly reduced by lamotrigine (34%) compared with placebo (9%), as were tonic-clonic seizures (36% reduction versus 10% increase for lamotrigine and placebo, respectively).

Adjunctive Therapy with Lamotrigine in Pediatric and Adult Patients with Primary Generalized Tonic-Clonic Seizures The effectiveness of lamotrigine as adjunctive therapy in patients with PGTC seizures was established in a multicenter, double-blind, placebo-controlled trial in 117 pediatric and adult patients aged 2 years and older (n = 58 on lamotrigine, n = 59 on placebo).

Patients with at least 3 PGTC seizures during an 8-week baseline phase were randomized to 19 to 24 weeks of treatment with lamotrigine or placebo added to their current AED regimen of up to 2 drugs.

Patients were dosed on a fixed-dose regimen, with target doses ranging from 3 to 12 mg/kg/day for pediatric patients and from 200 to 400 mg/day for adult patients based on concomitant AEDs.

The primary efficacy endpoint was percentage change from baseline in PGTC seizures.

For the intent-to-treat population, the median percent reduction in PGTC seizures was 66% in patients treated with lamotrigine and 34% on placebo, a difference that was statistically significant ( P = 0.006).

14.2 Bipolar Disorder Adults The effectiveness of lamotrigine in the maintenance treatment of bipolar I disorder was established in 2 multicenter, double-blind, placebo-controlled trials in adult patients (aged 18 to 82 years) who met DSM-IV criteria for bipolar I disorder.

Trial 1 enrolled patients with a current or recent (within 60 days) depressive episode as defined by DSM-IV and Trial 2 included patients with a current or recent (within 60 days) episode of mania or hypomania as defined by DSM-IV.

Both trials included a cohort of patients (30% of 404 subjects in Trial 1 and 28% of 171 patients in Trial 2) with rapid cycling bipolar disorder (4 to 6 episodes per year).

In both trials, patients were titrated to a target dose of 200 mg of lamotrigine as add-on therapy or as monotherapy with gradual withdrawal of any psychotropic medications during an 8- to 16-week open-label period.

Overall 81% of 1,305 patients participating in the open-label period were receiving 1 or more other psychotropic medications, including benzodiazepines, selective serotonin reuptake inhibitors (SSRIs), atypical antipsychotics (including olanzapine), valproate, or lithium, during titration of lamotrigine.

Patients with a CGI-severity score of 3 or less maintained for at least 4 continuous weeks, including at least the final week on monotherapy with lamotrigine, were randomized to a placebo-controlled, double-blind treatment period for up to 18 months.

The primary endpoint was TIME (time to intervention for a mood episode or one that was emerging, time to discontinuation for either an adverse event that was judged to be related to bipolar disorder, or for lack of efficacy).

The mood episode could be depression, mania, hypomania, or a mixed episode.

In Trial 1, patients received double-blind monotherapy with lamotrigine 50 mg/day (n = 50), lamotrigine 200 mg/day (n = 124), lamotrigine 400 mg/day (n = 47), or placebo (n = 121).

Lamotrigine (200- and 400-mg/day treatment groups combined) was superior to placebo in delaying the time to occurrence of a mood episode (Figure 1).

Separate analyses of the 200- and 400-mg/day dose groups revealed no added benefit from the higher dose.

In Trial 2, patients received double-blind monotherapy with lamotrigine (100 to 400 mg/day, n = 59), or placebo (n = 70).

Lamotrigine was superior to placebo in delaying time to occurrence of a mood episode (Figure 2).

The mean dose of lamotrigine was about 211 mg/day.

Although these trials were not designed to separately evaluate time to the occurrence of depression or mania, a combined analysis for the 2 trials revealed a statistically significant benefit for lamotrigine over placebo in delaying the time to occurrence of both depression and mania, although the finding was more robust for depression.

Figure 1: Kaplan-Meier Estimation of Cumulative Proportion of Patients with Mood Episode (Trial 1) Figure 2: Kaplan-Meier Estimation of Cumulative Proportion of Patients with Mood Episode (Trial 2) Lamotrigine Lamotrigine

HOW SUPPLIED

16 /STORAGE AND HANDLING Lamotrigine Orally Disintegrating Tablets 25-mg, white colored, round shaped, flat-faced, bevel-edged tablets debossed with “NT” on one side and “123” on the other side.

Maintenance Packs of 30 (NDC 49884-484-11).

50-mg, white colored, round shaped, flat-faced, bevel-edged tablets debossed with “EP” on one side and “191” on the other side.

Maintenance Packs of 30 (NDC 49884-485-11).

100-mg, Peach colored, round shaped, flat-faced, bevel-edged tablets debossed with “E” on one side and “432” on the other side.

Maintenance Packs of 30 (NDC 49884-486-11).

200-mg, White colored, round shaped, flat-faced, bevel-edged tablets debossed with “EP” on one side and “433” on the other side.

Maintenance Packs of 30 (NDC 49884-487-11).

Store at 20° to 25°C (68° to 77°F); with excursions permitted to 15° to 30°C (59° to 86°F).

[See USP Controlled Room Temperature].

Lamotrigine Orally Disintegrating Tablets Patient Titration Kit for Patients Taking Valproate (Blue ODT Kit) 25-mg, white colored, round shaped, flat-faced, bevel-edged tablets debossed with “NT” on one side and “123” on the other side and 50 mg, white colored, round shaped, flat-faced, bevel-edged tablets debossed with “EP” on one side and “191” on the other side, blister pack of 28 tablets (21/25-mg tablets and 7/50-mg tablets) (NDC 49884-880-99).

Lamotrigine Orally Disintegrating Tablets Patient Titration Kit for Patients Taking Carbamazepine, Phenytoin, Phenobarbital, or Primidone and Not Taking Valproate (Green ODT Kit) 50-mg, white colored, round shaped, flat-faced, bevel-edged tablets debossed with “EP” on one side and “191” on the other side.

and 100 mg, Peach colored, round shaped, flat-faced, bevel-edged tablets debossed with “E” on one side and “432” on the other side, blister pack of 56 tablets (42/50-mg tablets and 14/100-mg tablets) (NDC 49884-881-99).

Lamotrigine Oally Disintegrating Tablets Patient Titration Kit for Patients Not Taking Carbamazepine, Phenytoin, Phenobarbital, Primidone, or Valproate (Orange ODT Kit) 25-mg, white colored, round shaped, flat-faced, bevel-edged tablets debossed with “NT” on one side and “123” on the other side., 50 mg, white colored, round shaped, flat-faced, bevel-edged tablets debossed with “EP” on one side and “191” on the other side, and 100 mg, Peach colored, round shaped, flat-faced, bevel-edged tablets debossed with “E” on one side and “432” on the other side, blister pack of 35 tablets (14/25-mg tablets, 14/50-mg tablets, and 7/100-mg tablets) (NDC 49884-882-99).

Store at 20°C to 25°C (68°F to 77°F); with excursions permitted to 15°C to 30°C (59°F to 86°F).

[See USP Controlled Room Temperature].

Blister packs If the product is dispensed in a blister pack, the patient should be advised to examine the blister pack before use and not use if blisters are torn, broken, or missing.

RECENT MAJOR CHANGES

Warnings and Precautions, Hemophagocytic Lymphohistiocytosis (5.2) 8/2019

GERIATRIC USE

8.5 Geriatric Use Clinical trials of lamotrigine for epilepsy and bipolar disorder did not include sufficient numbers of patients aged 65 years and older to determine whether they respond differently from younger patients or exhibit a different safety profile than that of younger patients.

In general, dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range, reflecting the greater frequency of decreased hepatic, renal, or cardiac function and of concomitant disease or other drug therapy.

DOSAGE FORMS AND STRENGTHS

3 Orally disintegrating tablets: 25 mg, 50 mg, 100 mg, and 200 mg.

(3.3, 16) 3.3 Orally Disintegrating Tablets 25-mg, White colored, round shaped, flat-faced, bevel-edged tablets debossed with “NT” on one side and “123” on the other side.

50-mg, White colored, round shaped, flat-faced, bevel-edged tablets debossed with “EP” on one side and “191”on the other side.

100-mg, Peach colored, round shaped, flat-faced, bevel-edged tablets debossed with “E” on one side and “432” on the other side.

200-mg, White colored, round shaped, flat-faced, bevel-edged tablets debossed with “EP” on one side and “433” on the other side.

MECHANISM OF ACTION

12.1 Mechanism of Action The precise mechanism(s) by which lamotrigine exerts its anticonvulsant action are unknown.

In animal models designed to detect anticonvulsant activity, lamotrigine was effective in preventing seizure spread in the maximum electroshock (MES) and pentylenetetrazol (scMet) tests, and prevented seizures in the visually and electrically evoked after-discharge (EEAD) tests for antiepileptic activity.

Lamotrigine also displayed inhibitory properties in the kindling model in rats both during kindling development and in the fully kindled state.

The relevance of these models to human epilepsy, however, is not known.

One proposed mechanism of action of lamotrigine, the relevance of which remains to be established in humans, involves an effect on sodium channels.

In vitro pharmacological studies suggest that lamotrigine inhibits voltage-sensitive sodium channels, thereby stabilizing neuronal membranes and consequently modulating presynaptic transmitter release of excitatory amino acids (e.g., glutamate and aspartate).

Effect of Lamotrigine on N-Methyl d-Aspartate-Receptor Mediated Activity Lamotrigine did not inhibit N-methyl d-aspartate (NMDA)-induced depolarizations in rat cortical slices or NMDA-induced cyclic GMP formation in immature rat cerebellum, nor did lamotrigine displace compounds that are either competitive or noncompetitive ligands at this glutamate receptor complex (CNQX, CGS, TCHP).

The IC 50 for lamotrigine effects on NMDA-induced currents (in the presence of 3 µM of glycine) in cultured hippocampal neurons exceeded 100 µM.

The mechanisms by which lamotrigine exerts its therapeutic action in bipolar disorder have not been established.

INDICATIONS AND USAGE

1 Lamotrigine orally disintegrating tablets are indicated for: Epilepsy—adjunctive therapy in patients aged 2 years and older: partial-onset seizures.

primary generalized tonic-clonic seizures.

generalized seizures of Lennox-Gastaut syndrome.

(1.1) Epilepsy—monotherapy in patients aged 16 years and older: Conversion to monotherapy in patients with partial-onset seizures who are receiving treatment with carbamazepine, phenytoin, phenobarbital, primidone, or valproate as the single antiepileptic drug.

(1.1) Bipolar disorder : Maintenance treatment of bipolar I disorder to delay the time to occurrence of mood episodes in patients treated for acute mood episodes with standard therapy.

(1.2) Limitations of Use: Treatment of acute manic or mixed episodes is not recommended.

Effectiveness of lamotrigine in the acute treatment of mood episodes has not been established.

1.1 Epilepsy Adjunctive Therapy Lamotrigine orally disintegrating tablets are indicated as adjunctive therapy for the following seizure types in patients aged 2 years and older: partial-onset seizures.

primary generalized tonic-clonic (PGTC) seizures.

generalized seizures of Lennox-Gastaut syndrome.

Monotherapy Lamotrigine orally disintegrating tablets are indicated for conversion to monotherapy in adults (aged 16 years and older) with partial-onset seizures who are receiving treatment with carbamazepine, phenytoin, phenobarbital, primidone, or valproate as the single antiepileptic drug (AED).

Safety and effectiveness of lamotrigine orally disintegrating tablets have not been established (1) as initial monotherapy; (2) for conversion to monotherapy from AEDs other than carbamazepine, phenytoin, phenobarbital, primidone, or valproate; or (3) for simultaneous conversion to monotherapy from 2 or more concomitant AEDs.

1.2 Bipolar Disorder Lamotrigine orally disintegrating tablets are indicated for the maintenance treatment of bipolar I disorder to delay the time to occurrence of mood episodes (depression, mania, hypomania, mixed episodes) in patients treated for acute mood episodes with standard therapy [see Clinical Studies (14.1)].

Limitations of Use Treatment of acute manic or mixed episodes is not recommended.

Effectiveness of lamotrigine orally disintegrating tablets in the acute treatment of mood episodes has not been established.

PEDIATRIC USE

8.4 Pediatric Use Epilepsy Lamotrigine is indicated as adjunctive therapy in patients aged 2 years and older for partial-onset seizures, the generalized seizures of Lennox-Gastaut syndrome, and PGTC seizures.

Safety and efficacy of lamotrigine used as adjunctive treatment for partial-onset seizures were not demonstrated in a small, randomized, double-blind, placebo-controlled withdrawal trial in very young pediatric patients (aged 1 to 24 months).

Lamotrigine was associated with an increased risk for infectious adverse reactions (lamotrigine 37%, placebo 5%), and respiratory adverse reactions (lamotrigine 26%, placebo 5%).

Infectious adverse reactions included bronchiolitis, bronchitis, ear infection, eye infection, otitis externa, pharyngitis, urinary tract infection, and viral infection.

Respiratory adverse reactions included nasal congestion, cough, and apnea.

Bipolar Disorder Safety and efficacy of lamotrigine for the maintenance treatment of bipolar disorder were not established in a double-blind, randomized withdrawal, placebo-controlled trial that evaluated 301 pediatric patients aged 10 to 17 years with a current manic/hypomanic, depressed, or mixed mood episode as defined by DSM-IV-TR.

In the randomized phase of the trial, adverse reactions that occurred in at least 5% of patients taking lamotrigine (n = 87) and were twice as common compared with patients taking placebo (n = 86) were influenza (lamotrigine 8%, placebo 2%), oropharyngeal pain (lamotrigine 8%, placebo 2%), vomiting (lamotrigine 6%, placebo 2%), contact dermatitis (lamotrigine 5%, placebo 2%), upper abdominal pain (lamotrigine 5%, placebo 1%), and suicidal ideation (lamotrigine 5%, placebo 0%).

Juvenile Animal Data In a juvenile animal study in which lamotrigine (oral doses of 0, 5, 15, or 30 mg/kg) was administered to young rats from postnatal day 7 to 62, decreased viability and growth were seen at the highest dose tested and long-term Neurobehavioral abnormalities (decreased locomotor activity, increased reactivity, and learning deficits in animals tested as adults) were observed at the 2 highest doses.

The no-effect dose for adverse developmental effects in juvenile animals is less than the human dose of 400 mg/day on a mg/m 2 basis.

PREGNANCY

8.1 Pregnancy Pregnancy Exposure Registry There is a pregnancy exposure registry that monitors pregnancy outcomes in women exposed to AEDs, including lamotrigine, during pregnancy.

Encourage women who are taking lamotrigine during pregnancy to enroll in the North American Antiepileptic Drug (NAAED) Pregnancy Registry by calling 1-888-233-2334 or visiting http://www.aedpregnancyregistry.org/.

Risk Summary Data from several prospective pregnancy exposure registries and epidemiological studies of pregnant women have not detected an increased frequency of major congenital malformations or a consistent pattern of malformations among women exposed to lamotrigine compared with the general population (see Data) .

The majority of lamotrigine pregnancy exposure data are from women with epilepsy.

In animal studies, administration of lamotrigine during pregnancy resulted in developmental toxicity (increased mortality, decreased body weight, increased structural variation, neurobehavioral abnormalities) at doses lower than those administered clinically.

Lamotrigine decreased fetal folate concentrations in rats, an effect known to be associated with adverse pregnancy outcomes in animals and humans (see Data) .

The estimated background risk of major birth defects and miscarriage for the indicated population is unknown.

In the U.S.

general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2% to 4% and 15% to 20%, respectively.

Clinical Considerations As with other AEDs, physiological changes during pregnancy may affect lamotrigine concentrations and/or therapeutic effect.

There have been reports of decreased lamotrigine concentrations during pregnancy and restoration of pre-pregnancy concentrations after delivery.

Dose adjustments may be necessary to maintain clinical response.

Data Human Data: Data from several international pregnancy registries have not shown an increased risk for malformations overall.

The International Lamotrigine Pregnancy Registry reported major congenital malformations in 2.2% (95% CI: 1.6%, 3.1%) of 1,558 infants exposed to lamotrigine monotherapy in the first trimester of pregnancy.

The NAAED Pregnancy Registry reported major congenital malformations among 2.0% of 1,562 infants exposed to lamotrigine monotherapy in the first trimester.

EURAP, a large international pregnancy registry focused outside of North America, reported major birth defects in 2.9% (95% CI: 2.3%, 3.7%) of 2,514 exposures to lamotrigine monotherapy in the first trimester.

The frequency of major congenital malformations was similar to estimates from the general population.

The NAAED Pregnancy Registry observed an increased risk of isolated oral clefts: among 2,200 infants exposed to lamotrigine early in pregnancy, the risk of oral clefts was 3.2 per 1,000 (95% CI: 1.4, 6.3), a 3-fold increased risk versus unexposed healthy controls.

This finding has not been observed in other large international pregnancy registries.

Furthermore, a case-control study based on 21 congenital anomaly registries covering over 10 million births in Europe reported an adjusted odds ratio for isolated oral clefts with lamotrigine exposure of 1.45 (95% CI: 0.8, 2.63).

Several meta-analyses have not reported an increased risk of major congenital malformations following lamotrigine exposure in pregnancy compared with healthy and disease-matched controls.

No patterns of specific malformation types were observed.

The same meta-analyses evaluated the risk of additional maternal and infant outcomes including fetal death, stillbirth, preterm birth, small for gestational age, and neurodevelopmental delay.

Although there are no data suggesting an increased risk of these outcomes with lamotrigine monotherapy exposure, differences in outcome definition, ascertainment methods, and comparator groups limit the conclusions that can be drawn.

Animal Data: When lamotrigine was administered to pregnant mice, rats, or rabbits during the period of organogenesis (oral doses of up to 125, 25, and 30 mg/kg, respectively), reduced fetal body weight and increased incidences of fetal skeletal variations were seen in mice and rats at doses that were also maternally toxic.

The no-effect doses for embryofetal developmental toxicity in mice, rats, and rabbits (75, 6.25, and 30 mg/kg, respectively) are similar to (mice and rabbits) or less than (rats) the human dose of 400 mg/day on a body surface area (mg/m 2 ) basis.

In a study in which pregnant rats were administered lamotrigine (oral doses of 0, 5 or 25 mg/kg) during the period of organogenesis and offspring were evaluated postnatally, neurobehavioral abnormalities were observed in exposed offspring at both doses.

The lowest effect dose for developmental neurotoxicity in rats is less than the human dose of 400 mg/day on a mg/m 2 basis.

Maternal toxicity was observed at the higher dose tested.

When pregnant rats were administered lamotrigine (oral doses of 0, 5, 10, or 20 mg/kg) during the latter part of gestation and throughout lactation, increased offspring mortality (including stillbirths) was seen at all doses.

The lowest effect dose for pre- and post-natal developmental toxicity in rats is less than the human dose of 400 mg/day on a mg/m 2 basis.

Maternal toxicity was observed at the 2 highest doses tested.

When administered to pregnant rats, lamotrigine decreased fetal folate concentrations at doses greater than or equal to 5 mg/kg/day, which is less than the human dose of 400 mg/day on a mg/m 2 basis.

BOXED WARNING

WARNING: SERIOUS SKIN RASHES Lamotrigine ODT can cause serious rashes requiring hospitalization and discontinuation of treatment.

The incidence of these rashes, which have included Stevens-Johnson syndrome, is approximately 0.3% to 0.8% in pediatric patients (aged 2 to 17 years) and 0.08% to 0.3% in adults receiving lamotrigine.

One rash-related death was reported in a prospectively followed cohort of 1,983 pediatric patients (aged 2 to 16 years) with epilepsy taking lamotrigine as adjunctive therapy.

In worldwide postmarketing experience, rare cases of toxic epidermal necrolysis and/or rash-related death have been reported in adult and pediatric patients, but their numbers are too few to permit a precise estimate of the rate.

Other than age, there are as yet no factors identified that are known to predict the risk of occurrence or the severity of rash caused by lamotrigine.

There are suggestions, yet to be proven, that the risk of rash may also be increased by (1) coadministration of lamotrigine with valproate (includes valproic acid and divalproex sodium), (2) exceeding the recommended initial dose of lamotrigine, or (3) exceeding the recommended dose escalation for lamotrigine.

However, cases have occurred in the absence of these factors.

Nearly all cases of life-threatening rashes caused by lamotrigine have occurred within 2 to 8 weeks of treatment initiation.

However, isolated cases have occurred after prolonged treatment (e.g., 6 months).

Accordingly, duration of therapy cannot be relied upon as means to predict the potential risk heralded by the first appearance of a rash.

Although benign rashes are also caused by lamotrigine, it is not possible to predict reliably which rashes will prove to be serious or life threatening.

Accordingly, lamotrigine should ordinarily be discontinued at the first sign of rash, unless the rash is clearly not drug related.

Discontinuation of treatment may not prevent a rash from becoming life threatening or permanently disabling or disfiguring [see Warnings and Precautions (5.1)] .

WARNING: SERIOUS SKIN RASHES See full prescribing information for complete boxed warning.

Cases of life-threatening serious rashes, including Stevens-Johnson syndrome and toxic epidermal necrolysis, and/or rash-related death have been caused by lamotrigine.

The rate of serious rash is greater in pediatric patients than in adults.

Additional factors that may increase the risk of rash include: coadministration with valproate.

exceeding recommended initial dose of lamotrigine.

exceeding recommended dose escalation for lamotrigine.

(5.1) Benign rashes are also caused by lamotrigine; however, it is not possible to predict which rashes will prove to be serious or life threatening.

Lamotrigine should be discontinued at the first sign of rash, unless the rash is clearly not drug related.

(5.1)

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS Life-threatening serious rash and/or rash-related death: Discontinue at the first sign of rash, unless the rash is clearly not drug related.

(Boxed Warning, 5.1) Hemophagocytic lymphohistiocytosis: Consider this diagnosis and evaluate patients immediately if they develop signs or symptoms of systemic inflammation.

Discontinue lamotrigine if an alternative etiology is not established.

(5.2) Fatal or life-threatening hypersensitivity reaction: Multiorgan hypersensitivity reactions, also known as drug reaction with eosinophilia and systemic symptoms, may be fatal or life threatening.

Early signs may include rash, fever, and lymphadenopathy.

These reactions may be associated with other organ involvement, such as hepatitis, hepatic failure, blood dyscrasias, or acute multiorgan failure.

Lamotrigine should be discontinued if alternate etiology for this reaction is not found.

(5.3) Blood dyscrasias (e.g., neutropenia, thrombocytopenia, pancytopenia): May occur, either with or without an associated hypersensitivity syndrome.

Monitor for signs of anemia, unexpected infection, or bleeding.

(5.4) Suicidal behavior and ideation: Monitor for suicidal thoughts or behaviors.

(5.5) Aseptic meningitis: Monitor for signs of meningitis.

(5.6) Medication errors due to product name confusion: Strongly advise patients to visually inspect tablets to verify the received drug is correct.

(5.7, 16, 17) 5.1 Serious Skin Rashes [see Boxed Warning] Pediatric Population The incidence of serious rash associated with hospitalization and discontinuation of lamotrigine in a prospectively followed cohort of pediatric patients (aged 2 to 17 years) is approximately 0.3% to 0.8%.

One rash-related death was reported in a prospectively followed cohort of 1,983 pediatric patients (aged 2 to 16 years) with epilepsy taking lamotrigine as adjunctive therapy.

Additionally, there have been rare cases of toxic epidermal necrolysis with and without permanent sequelae and/or death in U.S.

and foreign postmarketing experience.

There is evidence that the inclusion of valproate in a multidrug regimen increases the risk of serious, potentially life-threatening rash in pediatric patients.

In pediatric patients who used valproate concomitantly for epilepsy, 1.2% (6 of 482) experienced a serious rash compared with 0.6% (6 of 952) patients not taking valproate.

Adult Population Serious rash associated with hospitalization and discontinuation of lamotrigine occurred in 0.3% (11 of 3,348) of adult patients who received lamotrigine in premarketing clinical trials of epilepsy.

In the bipolar and other mood disorders clinical trials, the rate of serious rash was 0.08% (1 of 1,233) of adult patients who received lamotrigine as initial monotherapy and 0.13% (2 of 1,538) of adult patients who received lamotrigine as adjunctive therapy.

No fatalities occurred among these individuals.

However, in worldwide postmarketing experience, rare cases of rash-related death have been reported, but their numbers are too few to permit a precise estimate of the rate.

Among the rashes leading to hospitalization were Stevens-Johnson syndrome, toxic epidermal necrolysis, angioedema, and those associated with multi-organ hypersensitivity [see Warnings and Precautions (5.3)].

There is evidence that the inclusion of valproate in a multidrug regimen increases the risk of serious, potentially life-threatening rash in adults.

Specifically, of 584 patients administered lamotrigine with valproate in epilepsy clinical trials, 6 (1%) were hospitalized in association with rash; in contrast, 4 (0.16%) of 2,398 clinical trial patients and volunteers administered lamotrigine in the absence of valproate were hospitalized.

Patients with History of Allergy or Rash to Other Antiepileptic Drugs The risk of nonserious rash may be increased when the recommended initial dose and/or the rate of dose escalation for lamotrigine is exceeded and in patients with a history of allergy or rash to other AEDs.

5.2 Hemophagocytic Lymphohistiocytosis Hemophagocytic lymphohistiocytosis (HLH) has occurred in pediatric and adult patients taking lamotrigine for various indications.

HLH is a life-threatening syndrome of pathologic immune activation characterized by clinical signs and symptoms of extreme systemic inflammation.

It is associated with high mortality rates if not recognized early and treated.

Common findings include fever, hepatosplenomegaly, rash, lymphadenopathy, neurologic symptoms, cytopenias, high serum ferritin, hypertriglyceridemia, and liver function and coagulation abnormalities.

In cases of HLH reported with lamotrigine, patients have presented with signs of systemic inflammation (fever, rash, hepatosplenomegaly, and organ system dysfunction) and blood dyscrasias.

Symptoms have been reported to occur within 8 to 24 days following the initiation of treatment.

Patients who develop early manifestations of pathologic immune activation should be evaluated immediately, and a diagnosis of HLH should be considered.

Lamotrigine should be discontinued if an alternative etiology for the signs or symptoms cannot be established.

5.3 Multiorgan Hypersensitivity Reactions and Organ Failure Multiorgan hypersensitivity reactions, also known as drug reaction with eosinophilia and systemic symptoms (DRESS), have occurred with lamotrigine.

Some have been fatal or life threatening.

DRESS typically, although not exclusively, presents with fever, rash, and/or lymphadenopathy in association with other organ system involvement, such as hepatitis, nephritis, hematologic abnormalities, myocarditis, or myositis, sometimes resembling an acute viral infection.

Eosinophilia is often present.

This disorder is variable in its expression, and other organ systems not noted here may be involved.

Fatalities associated with acute multiorgan failure and various degrees of hepatic failure have been reported in 2 of 3,796 adult patients and 4 of 2,435 pediatric patients who received lamotrigine in epilepsy clinical trials.

Rare fatalities from multiorgan failure have also been reported in postmarketing use.

Isolated liver failure without rash or involvement of other organs has also been reported with lamotrigine.

It is important to note that early manifestations of hypersensitivity (e.g., fever, lymphadenopathy) may be present even though a rash is not evident.

If such signs or symptoms are present, the patient should be evaluated immediately.

Lamotrigine should be discontinued if an alternative etiology for the signs or symptoms cannot be established.

Prior to initiation of treatment with lamotrigine, the patient should be instructed that a rash or other signs or symptoms of hypersensitivity (e.g., fever, lymphadenopathy) may herald a serious medical event and that the patient should report any such occurrence to a healthcare provider immediately.

5.4 Blood Dyscrasias There have been reports of blood dyscrasias that may or may not be associated with multiorgan hypersensitivity (also known as DRESS) [see Warnings and Precautions (5.3)] .

These have included neutropenia, leukopenia, anemia, thrombocytopenia, pancytopenia, and, rarely, aplastic anemia and pure red cell aplasia.

5.5 Suicidal Behavior and Ideation AEDs, including lamotrigine, increase the risk of suicidal thoughts or behavior in patients taking these drugs for any indication.

Patients treated with any AED for any indication should be monitored for the emergence or worsening of depression, suicidal thoughts or behavior, and/or any unusual changes in mood or behavior.

Pooled analyses of 199 placebo-controlled clinical trials (monotherapy and adjunctive therapy) of 11 different AEDs showed that patients randomized to 1 of the AEDs had approximately twice the risk (adjusted Relative Risk 1.8, 95% CI: 1.2, 2.7) of suicidal thinking or behavior compared with patients randomized to placebo.

In these trials, which had a median treatment duration of 12 weeks, the estimated incidence of suicidal behavior or ideation among 27,863 AED-treated patients was 0.43%, compared with 0.24% among 16,029 placebo-treated patients, representing an increase of approximately 1 case of suicidal thinking or behavior for every 530 patients treated.

There were 4 suicides in drug-treated patients in the trials and none in placebo-treated patients, but the number of events is too small to allow any conclusion about drug effect on suicide.

The increased risk of suicidal thoughts or behavior with AEDs was observed as early as 1 week after starting treatment with AEDs and persisted for the duration of treatment assessed.

Because most trials included in the analysis did not extend beyond 24 weeks, the risk of suicidal thoughts or behavior beyond 24 weeks could not be assessed.

The risk of suicidal thoughts or behavior was generally consistent among drugs in the data analyzed.

The finding of increased risk with AEDs of varying mechanism of action and across a range of indications suggests that the risk applies to all AEDs used for any indication.

The risk did not vary substantially by age (5 to 100 years) in the clinical trials analyzed.

Table 7 shows absolute and relative risk by indication for all evaluated AEDs.

Table 7.

Risk by Indication for Antiepileptic Drugs in the Pooled Analysis Indication Placebo Patients With Events per 1,000 Patients Drug Patients With Events per 1,000 Patients Relative Risk: Incidence of Events in Drug Patients/Incidence in Placebo Patients Risk Difference: Additional Drug Patients With Events per 1,000 Patients Epilepsy 1.0 3.4 3.5 2.4 Psychiatric 5.7 8.5 1.5 2.9 Other 1.0 1.8 1.9 0.9 Total 2.4 4.3 1.8 1.9 The relative risk for suicidal thoughts or behavior was higher in clinical trials for epilepsy than in clinical trials for psychiatric or other conditions, but the absolute risk differences were similar for the epilepsy and psychiatric indications.

Anyone considering prescribing lamotrigine or any other AED must balance the risk of suicidal thoughts or behavior with the risk of untreated illness.

Epilepsy and many other illnesses for which AEDs are prescribed are themselves associated with morbidity and mortality and an increased risk of suicidal thoughts and behavior.

Should suicidal thoughts and behavior emerge during treatment, the prescriber needs to consider whether the emergence of these symptoms in any given patient may be related to the illness being treated.

Patients, their caregivers, and families should be informed that AEDs increase the risk of suicidal thoughts and behavior and should be advised of the need to be alert for the emergence or worsening of the signs and symptoms of depression, any unusual changes in mood or behavior, the emergence of suicidal thoughts or suicidal behavior, or thoughts about self-harm.

Behaviors of concern should be reported immediately to healthcare providers.

5.6 Aseptic Meningitis Therapy with lamotrigine increases the risk of developing aseptic meningitis.

Because of the potential for serious outcomes of untreated meningitis due to other causes, patients should also be evaluated for other causes of meningitis and treated as appropriate.

Postmarketing cases of aseptic meningitis have been reported in pediatric and adult patients taking lamotrigine for various indications.

Symptoms upon presentation have included headache, fever, nausea, vomiting, and nuchal rigidity.

Rash, photophobia, myalgia, chills, altered consciousness, and somnolence were also noted in some cases.

Symptoms have been reported to occur within 1 day to one and a half months following the initiation of treatment.

In most cases, symptoms were reported to resolve after discontinuation of lamotrigine.

Re-exposure resulted in a rapid return of symptoms (from within 30 minutes to 1 day following re-initiation of treatment) that were frequently more severe.

Some of the patients treated with lamotrigine who developed aseptic meningitis had underlying diagnoses of systemic lupus erythematosus or other autoimmune diseases.

Cerebrospinal fluid (CSF) analyzed at the time of clinical presentation in reported cases was characterized by a mild to moderate pleocytosis, normal glucose levels, and mild to moderate increase in protein.

CSF white blood cell count differentials showed a predominance of neutrophils in a majority of the cases, although a predominance of lymphocytes was reported in approximately one third of the cases.

Some patients also had new onset of signs and symptoms of involvement of other organs (predominantly hepatic and renal involvement), which may suggest that in these cases the aseptic meningitis observed was part of a hypersensitivity reaction [see Warnings and Precautions (5.3)] .

5.7 Potential Medication Errors Medication errors involving lamotrigine have occurred.

In particular, the name lamotrigine can be confused with the names of other commonly used medications.

Medication errors may also occur between the different formulations of lamotrigine.

To reduce the potential of medication errors, write and say lamotrigine clearly.

Depictions of Lamotrigine orally disintegrating tablets can be found in the Medication Guide that accompanies the product to highlight the distinctive markings, colors, and shapes that serve to identify the different presentations of the drug and thus may help reduce the risk of medication errors.

To avoid the medication error of using the wrong drug or formulation, patients should be strongly advised to visually inspect their tablets to verify that they are lamotrigine, as well as the correct formulation of lamotrigine, each time they fill their prescription.

5.8 Concomitant Use with Oral Contraceptives Some estrogen-containing oral contraceptives have been shown to decrease serum concentrations of lamotrigine [see Clinical Pharmacology (12.3)] .

Dosage adjustments will be necessary in most patients who start or stop estrogen-containing oral contraceptives while taking lamotrigine [see Dosage and Administration (2.1)] .

During the week of inactive hormone preparation (pill-free week) of oral contraceptive therapy, plasma lamotrigine levels are expected to rise, as much as doubling at the end of the week.

Adverse reactions consistent with elevated levels of lamotrigine, such as dizziness, ataxia, and diplopia, could occur.

5.9 Withdrawal Seizures As with other AEDs, lamotrigine should not be abruptly discontinued.

In patients with epilepsy there is a possibility of increasing seizure frequency.

In clinical trials in adults with bipolar disorder, 2 patients experienced seizures shortly after abrupt withdrawal of lamotrigine.

Unless safety concerns require a more rapid withdrawal, the dose of lamotrigine should be tapered over a period of at least 2 weeks (approximately 50% reduction per week) [see Dosage and Administration (2.1)] .

5.10 Status Epilepticus Valid estimates of the incidence of treatment-emergent status epilepticus among patients treated with lamotrigine are difficult to obtain because reporters participating in clinical trials did not all employ identical rules for identifying cases.

At a minimum, 7 of 2,343 adult patients had episodes that could unequivocally be described as status epilepticus.

In addition, a number of reports of variably defined episodes of seizure exacerbation (e.g., seizure clusters, seizure flurries) were made.

5.11 Sudden Unexplained Death in Epilepsy (SUDEP) During the premarketing development of lamotrigine, 20 sudden and unexplained deaths were recorded among a cohort of 4,700 patients with epilepsy (5,747 patient-years of exposure).

Some of these could represent seizure-related deaths in which the seizure was not observed, e.g., at night.

This represents an incidence of 0.0035 deaths per patient-year.

Although this rate exceeds that expected in a healthy population matched for age and sex, it is within the range of estimates for the incidence of sudden unexplained death in epilepsy (SUDEP) in patients not receiving lamotrigine (ranging from 0.0005 for the general population of patients with epilepsy, to 0.004 for a recently studied clinical trial population similar to that in the clinical development program for lamotrigine, to 0.005 for patients with refractory epilepsy).

Consequently, whether these figures are reassuring or suggest concern depends on the comparability of the populations reported upon with the cohort receiving lamotrigine and the accuracy of the estimates provided.

Probably most reassuring is the similarity of estimated SUDEP rates in patients receiving lamotrigine and those receiving other AEDs, chemically unrelated to each other, that underwent clinical testing in similar populations.

This evidence suggests, although it certainly does not prove, that the high SUDEP rates reflect population rates, not a drug effect.

5.12 Addition of Lamotrigine to a Multidrug Regimen that Includes Valproate Because valproate reduces the clearance of lamotrigine, the dosage of lamotrigine in the presence of valproate is less than half of that required in its absence [see Dosage and Administration (2.2, 2.3, 2.4), Drug Interactions (7)].

5.13 Binding in the Eye and Other Melanin-Containing Tissues Because lamotrigine binds to melanin, it could accumulate in melanin-rich tissues over time.

This raises the possibility that lamotrigine may cause toxicity in these tissues after extended use.

Although ophthalmological testing was performed in 1 controlled clinical trial, the testing was inadequate to exclude subtle effects or injury occurring after long-term exposure.

Moreover, the capacity of available tests to detect potentially adverse consequences, if any, of lamotrigine’s binding to melanin is unknown [see Clinical Pharmacology (12.2)] .

Accordingly, although there are no specific recommendations for periodic ophthalmological monitoring, prescribers should be aware of the possibility of long-term ophthalmologic effects.

5.14 Laboratory Tests False-Positive Drug Test Results Lamotrigine has been reported to interfere with the assay used in some rapid urine drug screens, which can result in false-positive readings, particularly for phencyclidine (PCP).

A more specific analytical method should be used to confirm a positive result.

Plasma Concentrations of Lamotrigine The value of monitoring plasma concentrations of lamotrigine in patients treated with lamotrigine has not been established.

Because of the possible pharmacokinetic interactions between lamotrigine and other drugs, including AEDs (see Table 13), monitoring of the plasma levels of lamotrigine and concomitant drugs may be indicated, particularly during dosage adjustments.

In general, clinical judgment should be exercised regarding monitoring of plasma levels of lamotrigine and other drugs and whether or not dosage adjustments are necessary.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION Advise the patient to read the FDA-approved patient labeling (Medication Guide).

Rash Prior to initiation of treatment with lamotrigine, inform patients that a rash or other signs or symptoms of hypersensitivity (e.g., fever, lymphadenopathy) may herald a serious medical event and instruct them to report any such occurrence to their healthcare providers immediately.

Hemophagocytic Lymphohistiocytosis Prior to initiation of treatment with lamotrigine, inform patients that excessive immune activation may occur with lamotrigine and that they should report signs or symptoms such as fever, rash, or lymphadenopathy to a healthcare provider immediately.

Multiorgan Hypersensitivity Reactions, Blood Dyscrasias, and Organ Failure Inform patients that multiorgan hypersensitivity reactions and acute multiorgan failure may occur with lamotrigine.

Isolated organ failure or isolated blood dyscrasias without evidence of multiorgan hypersensitivity may also occur.

Instruct patients to contact their healthcare providers immediately if they experience any signs or symptoms of these conditions [see Warnings and Precautions (5.3, 5.4)].

Suicidal Thinking and Behavior Inform patients, their caregivers, and families that AEDs, including lamotrigine, may increase the risk of suicidal thoughts and behavior.

Instruct them to be alert for the emergence or worsening of symptoms of depression, any unusual changes in mood or behavior, or the emergence of suicidal thoughts or behavior or thoughts about self-harm.

Instruct them to immediately report behaviors of concern to their healthcare providers.

Worsening of Seizures Instruct patients to notify their healthcare providers if worsening of seizure control occurs.

Central Nervous System Adverse Effects Inform patients that lamotrigine may cause dizziness, somnolence, and other symptoms and signs of central nervous system depression.

Accordingly, instruct them neither to drive a car nor to operate other complex machinery until they have gained sufficient experience on lamotrigine to gauge whether or not it adversely affects their mental and/or motor performance.

Pregnancy and Nursing Instruct patients to notify their healthcare providers if they become pregnant or intend to become pregnant during therapy and if they intend to breastfeed or are breastfeeding an infant.

Encourage patients to enroll in the NAAED Pregnancy Registry if they become pregnant.

This registry is collecting information about the safety of antiepileptic drugs during pregnancy.

To enroll, patients can call the toll-free number 1-888-233-2334 [see Use in Specific Populations (8.1)].

Inform patients who intend to breastfeed that lamotrigine is present in breast milk and advise them to monitor their child for potential adverse effects of this drug.

Discuss the benefits and risks of continuing breastfeeding.

Oral Contraceptive Use Instruct women to notify their healthcare providers if they plan to start or stop use of oral contraceptives or other female hormonal preparations.

Starting estrogen-containing oral contraceptives may significantly decrease lamotrigine plasma levels and stopping estrogen-containing oral contraceptives (including the pill-free week) may significantly increase lamotrigine plasma levels [ see Warnings and Precautions (5.8), Clinical Pharmacology (12.3)] .

Also instruct women to promptly notify their healthcare providers if they experience adverse reactions or changes in menstrual pattern (e.g., break-through bleeding) while receiving lamotrigine in combination with these medications.

Discontinuing Lamotrigine Instructpatients to notify their healthcare providers if they stop taking lamotrigine for any reason and not to resume lamotrigine without consulting their healthcare providers.

Aseptic Meningitis Inform patients that lamotrigine may cause aseptic meningitis.

Instruct them to notify their healthcare providers immediately if they develop signs and symptoms of meningitis such as headache, fever, nausea, vomiting, stiff neck, rash, abnormal sensitivity to light, myalgia, chills, confusion, or drowsiness while taking lamotrigine.

Potential Medication Errors To avoid a medication error of using the wrong drug or formulation, strongly advise patients to visually inspect their tablets to verify that they are lamotrigine, as well as the correct formulation of lamotrigine, each time they fill their prescription [see Dosage Forms and Strengths ( 3.3), How Supplied/Storage And Handling (16)] .

Refer the patient to the Medication Guide that provides depictions of the lamotrigine orally disintegrating tablets.

Other Brands listed are the trademarks of their respective owners.

DOSAGE AND ADMINISTRATION

2 Dosing is based on concomitant medications, indication, and patient age.

(2.1, 2.2, 2.3, 2.4) To avoid an increased risk of rash, the recommended initial dose and subsequent dose escalations should not be exceeded.

Lamotrigine Starter Kits and Lamotrigine Orally Disintegrating Tablets Patient Titration Kits are available for the first 5 weeks of treatment.

(2.1, 16) Do not restart lamotrigine orally disintegrating tablets in patients who discontinued due to rash unless the potential benefits clearly outweigh the risks.

(2.1, 5.1) Adjustments to maintenance doses will be necessary in most patients starting or stopping estrogen-containing oral contraceptives.

(2.1, 5.8) Discontinuation: Taper over a period of at least 2 weeks (approximately 50% dose reduction per week).

(2.1, 5.9) Epilepsy: Adjunctive therapy—See Table 1 for patients older than 12 years and Tables 2 and 3 for patients aged 2 to 12 years.

(2.2) Conversion to monotherapy—See Table 4.

(2.3) Bipolar disorder: See Tables 5 and 6.

(2.4) 2.1 General Dosing Considerations Rash There are suggestions, yet to be proven, that the risk of severe, potentially life-threatening rash may be increased by (1) coadministration of lamotrigine with valproate, (2) exceeding the recommended initial dose of lamotrigine, or (3) exceeding the recommended dose escalation for lamotrigine.

However, cases have occurred in the absence of these factors [see Boxed Warning].

Therefore, it is important that the dosing recommendations be followed closely.

The risk of nonserious rash may be increased when the recommended initial dose and/or the rate of dose escalation for lamotrigine orally disintegrating tablets are exceeded and in patients with a history of allergy or rash to other AEDs.

Lamotrigine ODT Patient Titration Kits provide lamotrigine at doses consistent with the recommended titration schedule for the first 5 weeks of treatment, based upon concomitant medications, for patients with epilepsy (older than 12 years) and bipolar I disorder (adults) and are intended to help reduce the potential for rash.

The use of lamotrigine ODT Patient Titration Kits is recommended for appropriate patients who are starting or restarting lamotrigine orally disintegrating tablets [see How Supplied/Storage and Handling (16)].

It is recommended that lamotrigine orally disintegrating tablets not be restarted in patients who discontinued due to rash associated with prior treatment with lamotrigine unless the potential benefits clearly outweigh the risks.

If the decision is made to restart a patient who has discontinued lamotrigine orally disintegrating tablets, the need to restart with the initial dosing recommendations should be assessed.

The greater the interval of time since the previous dose, the greater consideration should be given to restarting with the initial dosing recommendations.

If a patient has discontinued lamotrigine for a period of more than 5 half-lives, it is recommended that initial dosing recommendations and guidelines be followed.

The half-life of lamotrigine is affected by other concomitant medications [see Clinical pharmacology (12.3)].

Lamotrigine Added to Drugs Known to Induce or Inhibit Glucuronidation Because lamotrigine orally disintegrating tablets are metabolized predominantly by glucuronic acid conjugation, drugs that are known to induce or inhibit glucuronidation may affect the apparent clearance of lamotrigine.

Drugs that induce glucuronidation include carbamazepine, phenytoin, phenobarbital, primidone, rifampin, estrogen-containing oral contraceptives, and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir.

Valproate inhibits glucuronidation.

For dosing considerations for lamotrigine orally disintegrating tablets in patients on estrogen-containing contraceptives and atazanavir/ritonavir, see below and Table 13.

For dosing considerations for lamotrigine orally disintegrating tablets in patients on other drugs known to induce or inhibit glucuronidation, see Tables 1, 2, 5 to 6, and 13.

Target Plasma Levels for Patients with Epilepsy or Bipolar Disorder A therapeutic plasma concentration range has not been established for lamotrigine.

Dosing of lamotrigine orally disintegrating tablets should be based on therapeutic response [ see Clinical Pharmacology (12.3)].

Women Taking Estrogen-Containing Oral Contraceptives Starting Lamotrigine orally disintegrating tablets in Women Taking Estrogen-Containing Oral Contraceptives: Although estrogen-containing oral contraceptives have been shown to increase the clearance of lamotrigine [see Clinical Pharmacology (12.3)], no adjustments to the recommended dose-escalation guidelines for lamotrigine orally disintegrating tablets should be necessary solely based on the use of estrogen-containing oral contraceptives.

Therefore, dose escalation should follow the recommended guidelines for initiating adjunctive therapy with lamotrigine orally disintegrating tablets based on the concomitant AED or other concomitant medications (see Tables 1, 5, and 7).

See below for adjustments to maintenance doses of lamotrigine orally disintegrating tablets in women taking estrogen-containing oral contraceptives.

Adjustments to the Maintenance Dose of Lamotrigine in Women Taking Estrogen-Containing Oral Contraceptives: (1) Taking Estrogen-Containing Oral Contraceptives : In women not taking carbamazepine, phenytoin, phenobarbital, primidone, or other drugs such as rifampin and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine glucuronidation [see Drug Interactions (7), Clinical Pharmacology (12.3)], the maintenance dose of lamotrigine orally disintegrating tablets will in most cases need to be increased by as much as 2-fold over the recommended target maintenance dose to maintain a consistent lamotrigine plasma level.

(2) Starting Estrogen-Containing Oral Contraceptives: In women taking a stable dose of lamotrigine orally disintegrating tablets and not taking carbamazepine, phenytoin, phenobarbital, primidone, or other drugs such as rifampin and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine glucuronidation [see Drug Interactions (7), Clinical Pharmacology (12.3)], the maintenance dose will in most cases need to be increased by as much as 2-fold to maintain a consistent lamotrigine plasma level.

The dose increases should begin at the same time that the oral contraceptive is introduced and continue, based on clinical response, no more rapidly than 50 to 100 mg/day every week.

Dose increases should not exceed the recommended rate (see Tables 1 and 5) unless lamotrigine plasma levels or clinical response support larger increases.

Gradual transient increases in lamotrigine plasma levels may occur during the week of inactive hormonal preparation (pill-free week), and these increases will be greater if dose increases are made in the days before or during the week of inactive hormonal preparation.

Increased lamotrigine plasma levels could result in additional adverse reactions, such as dizziness, ataxia, and diplopia.

If adverse reactions attributable to lamotrigine orally disintegrating tablets consistently occur during the pill-free week, dose adjustments to the overall maintenance dose may be necessary.

Dose adjustments limited to the pill-free week are not recommended.

For women taking lamotrigine orally disintegrating tablets in addition to carbamazepine, phenytoin, phenobarbital, primidone, or other drugs such as rifampin and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine glucuronidation [see Drug Interactions (7), Clinical Pharmacology (12.3)], no adjustment to the dose of lamotrigine orally disintegrating tablets should be necessary.

(3) Stopping Estrogen-Containing Oral Contraceptives : In women not taking carbamazepine, phenytoin, phenobarbital, primidone, or other drugs such as rifampin and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine glucuronidation [see Drug Interactions (7), Clinical Pharmacology (12.3)], the maintenance dose of lamotrigine orally disintegrating tablets will in most cases need to be decreased by as much as 50% in order to maintain a consistent lamotrigine plasma level.

The decrease in dose of lamotrigine orally disintegrating tablets should not exceed 25% of the total daily dose per week over a 2-week period, unless clinical response or lamotrigine plasma levels indicate otherwise [see Clinical Pharmacology (12.3)].

In women taking lamotrigine in addition to carbamazepine, phenytoin, phenobarbital, primidone, or other drugs such as rifampin and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine glucuronidation [see Drug Interactions (7), Clinical Pharmacology (12.3)], no adjustment to the dose of lamotrigine orally disintegrating tablets should be necessary.

Women and Other Hormonal Contraceptive Preparations or Hormone Replacement Therapy The effect of other hormonal contraceptive preparations or hormone replacement therapy on the pharmacokinetics of lamotrigine has not been systematically evaluated.

It has been reported that ethinylestradiol, not progestogens, increased the clearance of lamotrigine up to 2-fold, and the progestin-only pills had no effect on lamotrigine plasma levels.

Therefore, adjustments to the dosage of lamotrigine orally disintegrating tablets in the presence of progestogens alone will likely not be needed.

Patients Taking Atazanavir/Ritonavir While atazanavir/ritonavir does reduce the lamotrigine plasma concentration, no adjustments to the recommended dose-escalation guidelines for lamotrigine orally disintegrating tablets should be necessary solely based on the use of atazanavir/ritonavir.

Dose escalation should follow the recommended guidelines for initiating adjunctive therapy with lamotrigine orally disintegrating tablets based on concomitant AED or other concomitant medications (see Tables 1, 2, and 5).

In patients already taking maintenance doses of lamotrigine orally disintegrating tablets and not taking glucuronidation inducers, the dose of lamotrigine orally disintegrating tablets may need to be increased if atazanavir/ritonavir is added, or decreased if atazanavir/ritonavir is discontinued [see Clinical Pharmacology (12.3)].

Patients with Hepatic Impairment Experience in patients with hepatic impairment is limited.

Based on a clinical pharmacology study in 24 subjects with mild, moderate, and severe liver impairment [see Use in Specific Populations (8.6), Clinical Pharmacology (12.3)], the following general recommendations can be made.

No dosage adjustment is needed in patients with mild liver impairment.

Initial, escalation, and maintenance doses should generally be reduced by approximately 25% in patients with moderate and severe liver impairment without ascites and 50% in patients with severe liver impairment with ascites.

Escalation and maintenance doses may be adjusted according to clinical response.

Patients with Renal Impairment Initial doses of lamotrigine orally disintegrating tablets should be based on patients’ concomitant medications (see Tables 1 to 3 and 5); reduced maintenance doses may be effective for patients with significant renal impairment [see Use in Specific Populations (8.7), Clinical Pharmacology (12.3)].

Few patients with severe renal impairment have been evaluated during chronic treatment with lamotrigine orally disintegrating tablets.

Because there is inadequate experience in this population, lamotrigine orally disintegrating tablets should be used with caution in these patients.

Discontinuation Strategy Epilepsy : For patients receiving lamotrigine orally disintegrating tablets in combination with other AEDs, a re-evaluation of all AEDs in the regimen should be considered if a change in seizure control or an appearance or worsening of adverse reactions is observed.

If a decision is made to discontinue therapy with lamotrigine orally disintegrating tablets, a step-wise reduction of dose over at least 2 weeks (approximately 50% per week) is recommended unless safety concerns require a more rapid withdrawal [see Warnings and Precautions (5.9)].

Discontinuing carbamazepine, phenytoin, phenobarbital, primidone, or other drugs such as rifampin and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine glucuronidation should prolong the half-life of lamotrigine; discontinuing valproate should shorten the half-life of lamotrigine.

Bipolar Disorder: In the controlled clinical trials, there was no increase in the incidence, type, or severity of adverse reactions following abrupt termination of lamotrigine orally disintegrating tablets.

In the clinical development program in adults with bipolar disorder, 2 patients experienced seizures shortly after abrupt withdrawal of lamotrigine orally disintegrating tablets.

Discontinuation of lamotrigine orally disintegrating tablets should involve a step-wise reduction of dose over at least 2 weeks (approximately 50% per week) unless safety concerns require a more rapid withdrawal [see Warnings and Precautions (5.9)].

2.2 Epilepsy – Adjunctive Therapy This section provides specific dosing recommendations for patients older than 12 years and patients aged 2 to 12 years.

Within each of these age-groups, specific dosing recommendations are provided depending upon the concomitant AEDs or other concomitant medications (see Table 1 for patients older than 12 years and Table 2 for patients aged 2 to 12 years).

A weight-based dosing guide for patients aged 2 to 12 years on concomitant valproate is provided in Table 3.

Patients Older than 12 Years Recommended dosing guidelines are summarized in Table 1.

Table 1.

Escalation Regimen for Lamotrigine Orally Disintegrating Tablets in Patients Older than 12 Years with Epilepsy In Patients TAKING Valproate a In Patients NOT TAKING Carbamazepine, Phenytoin, Phenobarbital, Primidone, b or Valproate a In Patients TAKING Carbamazepine, Phenytoin, Phenobarbital, or Primidone b and NOT TAKING Valproate a Weeks 1 and 2 25 mg every other day 25 mg every day 50 mg/day Weeks 3 and 4 25 mg every day 50 mg/day 100 mg/day (in 2 divided doses) Week 5 onward to maintenance Increase by 25 to 50 mg/day every 1 to 2 weeks.

Increase by 50 mg/day every 1 to 2 weeks.

Increase by 100 mg/day every 1 to 2 weeks.

Usual maintenance dose 100 to 200 mg/day with valproate alone 100 to 400 mg/day with valproate and other drugs that induce glucuronidation (in 1 or 2 divided doses) 225 to 375 mg/day (in 2 divided doses) 300 to 500 mg/day (in 2 divided doses) a Valproate has been shown to inhibit glucuronidation and decrease the apparent clearance of lamotrigine [see Drug Interactions (7), Clinical Pharmacology (12.3)].

b Drugs that induce lamotrigine glucuronidation and increase clearance, other than the specified antiepileptic drugs, include estrogen-containing oral contraceptives, rifampin, and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir.

Dosing recommendations for oral contraceptives and the protease inhibitor atazanavir/ritonavir can be found in General Dosing Considerations [see Dosage and Administration (2.1)].

Patients on rifampin and the protease inhibitor lopinavir/ritonavir should follow the same dosing titration/maintenance regimen used with antiepileptic drugs that induce glucuronidation and increase clearance [see Dosage and Administration (2.1), Drug Interactions (7), Clinical Pharmacology (12.3)].

Patients Aged 2 to 12 Years Recommended dosing guidelines are summarized in Table 2.

Lower starting doses and slower dose escalations than those used in clinical trials are recommended because of the suggestion that the risk of rash may be decreased by lower starting doses and slower dose escalations.

Therefore, maintenance doses will take longer to reach in clinical practice than in clinical trials.

It may take several weeks to months to achieve an individualized maintenance dose.

Maintenance doses in patients weighing <30 kg, regardless of age or concomitant AED, may need to be increased as much as 50%, based on clinical response.

Table 2.

Escalation Regimen for Lamotrigine Orally Disintegrating Tablets in Patients Aged 2 to 12 Years with Epilepsy In Patients TAKING Valproate a In Patients NOT TAKING Carbamazepine, Phenytoin, Phenobarbital, Primidone, b or Valproate a In Patients TAKING Carbamazepine, Phenytoin, Phenobarbital, or Primidone b and NOT TAKING Valproate a Weeks 1 and 2 0.15 mg/kg/day in 1 or 2 divided doses, rounded down to the nearest whole tablet (see Table 3 for weight-based dosing guide) 0.3 mg/kg/day in 1 or 2 divided doses, rounded down to the nearest whole tablet 0.6 mg/kg/day in 2 divided doses, rounded down to the nearest whole tablet Weeks 3 and 4 0.3 mg/kg/day in 1 or 2 divided doses, rounded down to the nearest whole tablet (see Table 3 for weight-based dosing guide) 0.6 mg/kg/day in 2 divided doses, rounded down to the nearest whole tablet 1.2 mg/kg/day in 2 divided doses, rounded down to the nearest whole tablet Week 5 onward to maintenance The dose should be increased every 1 to 2 weeks as follows: calculate 0.3 mg/kg/day, round this amount down to the nearest whole tablet, and add this amount to the previously administered daily dose.

The dose should be increased every 1 to 2 weeks as follows: calculate 0.6 mg/kg/day, round this amount down to the nearest whole tablet, and add this amount to the previously administered daily dose.

The dose should be increased every 1 to 2 weeks as follows: calculate 1.2 mg/kg/day, round this amount down to the nearest whole tablet, and add this amount to the previously administered daily dose.

Usual maintenance dose 1 to 5 mg/kg/day (maximum 200 mg/day in 1 or 2 divided doses) 1 to 3 mg/kg/day with valproate alone 4.5 to 7.5 mg/kg/day (maximum 300 mg/day in 2 divided doses) 5 to 15 mg/kg/day (maximum 400 mg/day in 2 divided doses) Maintenance dose in patients <30 kg May need to be increased by as much as 50%, based on clinical response.

May need to be increased by as much as 50%, based on clinical response.

May need to be increased by as much as 50%, based on clinical response.

Note: Only whole tablets should be used for dosing.

a Valproate has been shown to inhibit glucuronidation and decrease the apparent clearance of lamotrigine [see Drug Interactions (7), Clinical Pharmacology (12.3)].

b Drugs that induce lamotrigine glucuronidation and increase clearance, other than the specified antiepileptic drugs, include estrogen-containing oral contraceptives, rifampin, and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir.

Dosing recommendations for oral contraceptives and the protease inhibitor atazanavir/ritonavir can be found in General Dosing Considerations [see Dosage and Administration (2.1)].

Patients on rifampin and the protease inhibitor lopinavir/ritonavir should follow the same dosing titration/maintenance regimen used with antiepileptic drugs that induce glucuronidation and increase clearance [see Dosage and Administration (2.1), Drug Interactions (7), Clinical Pharmacology (12.3)].

Table 3.

The Initial Weight-Based Dosing Guide for Patients Aged 2 to 12 Years Taking Valproate (Weeks 1 to 4) with Epilepsy If the patient’s weight is Give this daily dose, using the most appropriate combination of lamotrigine 2- and 5-mg tablets Greater than And less than Weeks 1 and 2 Weeks 3 and 4 6.7 kg 14 kg 2 mg every other day 2 mg every day 14.1 kg 27 kg 2 mg every day 4 mg every day 27.1 kg 34 kg 4 mg every day 8 mg every day 34.1 kg 40 kg 5 mg every day 10 mg every day Usual Adjunctive Maintenance Dose for Epilepsy The usual maintenance doses identified in Tables 1 and 2 are derived from dosing regimens employed in the placebo-controlled adjunctive trials in which the efficacy of lamotrigine orally disintegrating tablets was established.

In patients receiving multidrug regimens employing carbamazepine, phenytoin, phenobarbital, or primidone without valproate , maintenance doses of adjunctive lamotrigine orally disintegrating tablets as high as 700 mg/day have been used.

In patients receiving valproate alone , maintenance doses of adjunctive lamotrigine orally disintegrating tablets as high as 200 mg/day have been used.

The advantage of using doses above those recommended in Tables 1 to 4 has not been established in controlled trials.

2.3 Epilepsy – Conversion from Adjunctive Therapy to Monotherapy The goal of the transition regimen is to attempt to maintain seizure control while mitigating the risk of serious rash associated with the rapid titration of lamotrigine orally disintegrating tablets.

The recommended maintenance dose of lamotrigine orally disintegrating tablets as monotherapy is 500 mg/day given in 2 divided doses.

To avoid an increased risk of rash, the recommended initial dose and subsequent dose escalations for lamotrigine should not be exceeded [see Boxed Warning].

Conversion from Adjunctive Therapy with Carbamazepine, Phenytoin, Phenobarbital, or Primidone to Monotherapy with Lamotrigine Orally Disintegrating Tablets Orally Disintegrating Tablets After achieving a dose of 500 mg/day of lamotrigine orally disintegrating tablets using the guidelines in Table 1, the concomitant enzyme-inducing AED should be withdrawn by 20% decrements each week over a 4-week period.

The regimen for the withdrawal of the concomitant AED is based on experience gained in the controlled monotherapy clinical trial.

Conversion from Adjunctive Therapy with Valproate to Monotherapy with Lamotrigine Orally Disintegrating Tablets The conversion regimen involves the 4 steps outlined in Table 4.

Table 4.

Conversion from Adjunctive Therapy with Valproate to Monotherapy with Lamotrigine Orally Disintegrating Tablets in Patients Aged 16 Years and Older with Epilepsy Lamotrigine Orally Disintegrating Tablets Valproate Step 1 Achieve a dose of 200 mg/day according to guidelines in Table 1.

Maintain established stable dose.

Step 2 Maintain at 200 mg/day.

Decrease dose by decrements no greater than 500 mg/day/week to 500 mg/day and then maintain for 1 week.

Step 3 Increase to 300 mg/day and maintain for 1 week.

Simultaneously decrease to 250 mg/day and maintain for 1 week.

Step 4 Increase by 100 mg/day every week to achieve maintenance dose of 500 mg/day.

Discontinue.

Conversion from Adjunctive Therapy with Antiepileptic Drugs other than Carbamazepine, Phenytoin, Phenobarbital, Primidone, or Valproate to Monotherapy with Lamotrigine Orally Disintegrating Tablets No specific dosing guidelines can be provided for conversion to monotherapy with lamotrigine orally disintegrating tablets with AEDs other than carbamazepine, phenytoin, phenobarbital, primidone, or valproate.

2.4 Bipolar Disorder The goal of maintenance treatment with lamotrigine orally disintegrating tablet is to delay the time to occurrence of mood episodes (depression, mania, hypomania, mixed episodes) in patients treated for acute mood episodes with standard therapy [see Indications and Usage (1.2)] .

Patients taking lamotrigine orally disintegrating tablets for more than 16 weeks should be periodically reassessed to determine the need for maintenance treatment.

Adults The target dose of lamotrigine orally disintegrating tablet is 200 mg/day (100 mg/day in patients taking valproate, which decreases the apparent clearance of lamotrigine, and 400 mg/day in patients not taking valproate and taking either carbamazepine, phenytoin, phenobarbital, primidone, or other drugs such as rifampin and the protease inhibitor lopinavir/ritonavir that increase the apparent clearance of lamotrigine).

In the clinical trials, doses up to 400 mg/day as monotherapy were evaluated; however, no additional benefit was seen at 400 mg/day compared with 200 mg/day [see Clinical Studies (14.2)] .

Accordingly, doses above 200 mg/day are not recommended.

Treatment with lamotrigine orally disintegrating tablets are introduced, based on concurrent medications, according to the regimen outlined in Table 5.

If other psychotropic medications are withdrawn following stabilization, the dose of lamotrigine orally disintegrating tablets should be adjusted.

In patients discontinuing valproate, the dose of lamotrigine orally disintegrating tablets should be doubled over a 2-week period in equal weekly increments (see Table 6).

In patients discontinuing carbamazepine, phenytoin, phenobarbital, primidone, or other drugs such as rifampin and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir that induce lamotrigine glucuronidation, the dose of lamotrigine orally disintegrating tablets should remain constant for the first week and then should be decreased by half over a 2-week period in equal weekly decrements (see Table 6).

The dose of lamotrigine orally disintegrating tablets may then be further adjusted to the target dose (200 mg) as clinically indicated.

If other drugs are subsequently introduced, the dose of lamotrigine orally disintegrating tablets may need to be adjusted.

In particular, the introduction of valproate requires reduction in the dose of lamotrigine orally disintegrating tablets [see Drug Interactions (7), Clinical Pharmacology (12.3)].

To avoid an increased risk of rash, the recommended initial dose and subsequent dose escalations of lamotrigine orally disintegrating tablets should not be exceeded [see Boxed Warning].

Table 5.

Escalation Regimen for Lamotrigine Orally Disintegrating Tablets in Adults with Bipolar Disorder In Patients TAKING Valproate a In Patients NOT TAKING Carbamazepine, Phenytoin, Phenobarbital, Primidone, b or Valproate a In Patients TAKING Carbamazepine, Phenytoin, Phenobarbital, or Primidone b and NOT TAKING Valproate a Weeks 1 and 2 25 mg every other day 25 mg daily 50 mg daily Weeks 3 and 4 25 mg daily 50 mg daily 100 mg daily, in divided doses Week 5 50 mg daily 100 mg daily 200 mg daily, in divided doses Week 6 100 mg daily 200 mg daily 300 mg daily, in divided doses Week 7 100 mg daily 200 mg daily up to 400 mg daily, in divided doses a Valproate has been shown to inhibit glucuronidation and decrease the apparent clearance of lamotrigine [see Drug Interactions (7), Clinical Pharmacology (12.3)] .

b Drugs that induce lamotrigine glucuronidation and increase clearance, other than the specified antiepileptic drugs, include estrogen-containing oral contraceptives, rifampin, and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir.

Dosing recommendations for oral contraceptives and the protease inhibitor atazanavir/ritonavir can be found in General Dosing Considerations [see Dosage and Administration (2.1)] .

Patients on rifampin and the protease inhibitor lopinavir/ritonavir should follow the same dosing titration/maintenance regimen used with antiepileptic drugs that induce glucuronidation and increase clearance [see Dosage and Administration (2.1), Drug Interactions (7), Clinical Pharmacology (12.3)].

Table 6.

Dosage Adjustments to Lamotrigine Orally Disintegrating Tablets in Adults with Bipolar Disorder Following Discontinuation of Psychotropic Medications Discontinuation of Psychotropic Drugs (excluding Valproate, a Carbamazepine, Phenytoin, Phenobarbital, or Primidone b ) After Discontinuation of Valproate a After Discontinuation of Carbamazepine, Phenytoin, Phenobarbital, or Primidone b Current Dose of Lamotrigine Orally Disintegrating Tablets (mg/day) 100 Current Dose of Lamotrigine Orally Disintegrating Tablets(mg/day) 400 Week 1 Maintain current dose of lamotrigine orally disintegrating tablets 150 400 Week 2 Maintain current dose of lamotrigine orally disintegrating tablets 200 300 Week 3 onward Maintain current dose of lamotrigine orally disintegrating tablets 200 200 a Valproate has been shown to inhibit glucuronidation and decrease the apparent clearance of lamotrigine [see Drug Interactions (7), Clinical Pharmacology (12.3) ].

b Drugs that induce lamotrigine glucuronidation and increase clearance, other than the specified antiepileptic drugs, include estrogen-containing oral contraceptives, rifampin, and the protease inhibitors lopinavir/ritonavir and atazanavir/ritonavir.

Dosing recommendations for oral contraceptives and the protease inhibitor atazanavir/ritonavir can be found in General Dosing Considerations [see Dosage and Administration (2.1)] .

Patients on rifampin and the protease inhibitor lopinavir/ritonavir should follow the same dosing titration/maintenance regimen used with antiepileptic drugs that induce glucuronidation and increase clearance [see Dosage and Administration (2.1), Drug Interactions (7), Clinical Pharmacology (12.3)].

2.6 Administration of Lamotrigine Orally Disintegrating Tablets Lamotrigine orally disintegrating tablets should be placed onto the tongue and moved around in the mouth.

The tablet will disintegrate rapidly, can be swallowed with or without water, and can be taken with or without food.

Dicyclomine Hydrochloride 10 MG Oral Capsule

DRUG INTERACTIONS

7 • Antiglaucoma agents : anticholinergics antagonize antiglaucoma agents and may increase intraoccular pressure ( 7 ) • Anticholinergic agents : may affect the gastrointestinal absorption of various drugs; may also increase certain actions or side effects of other anticholinergic drugs ( 7 ) • Antacids : interfere with the absorption of anticholinergic agents ( 7 ) 7.1 Antiglaucoma Agents Anticholinergics antagonize the effects of antiglaucoma agents.

Anticholinergic drugs in the presence of increased intraocular pressure may be hazardous when taken concurrently with agents such as corticosteroids.

Use of dicyclomine in patients with glaucoma is not recommended [see Contraindications (4) ].

7.2 Other Drugs with Anticholinergic Activity The following agents may increase certain actions or side effects of anticholinergic drugs including dicyclomine: amantadine, antiarrhythmic agents of Class I (e.g., quinidine), antihistamines, antipsychotic agents (e.g., phenothiazines), benzodiazepines, MAO inhibitors, narcotic analgesics (e.g., meperidine), nitrates and nitrites, sympathomimetic agents, tricyclic antidepressants and other drugs having anticholinergic activity.

7.3 Other Gastrointestinal Motility Drugs Interaction with other gastrointestinal motility drugs may antagonize the effects of drugs that alter gastrointestinal motility, such as metoclopramide.

7.4 Effect of Antacids Because antacids may interfere with the absorption of anticholinergic agents including dicyclomine, simultaneous use of these drugs should be avoided.

7.5 Effect on Absorption of Other Drugs Anticholinergic agents may affect gastrointestinal absorption of various drugs by affecting on gastrointestinal motility, such as slowly dissolving dosage forms of digoxin; increased serum digoxin concentration may result.

7.6 Effect on Gastric Acid Secretion The inhibiting effects of anticholinergic drugs on gastric hydrochloric acid secretion are antagonized by agents used to treat achlorhydria and those used to test gastric secretion.

OVERDOSAGE

10 In case of an overdose, patients should contact a physician, poison control center (1-800-222-1222), or emergency room.

The signs and symptoms of overdosage include: headache; nausea; vomiting; blurred vision; dilated pupils; hot, dry skin; dizziness; dryness of the mouth; difficulty in swallowing; and CNS stimulation including convulsion.

A curare-like action may occur (i.e., neuromuscular blockade leading to muscular weakness and possible paralysis).

One reported event included a 37 year old who reported numbness on the left side, cold fingertips, blurred vision, abdominal and flank pain, decreased appetite, dry mouth and nervousness following ingestion of 320 mg daily (four 20 mg tablets 4 times daily).

These events resolved after discontinuing the dicyclomine.

The acute oral LD 50 of the drug is 625 mg/kg in mice.

The amount of drug in a single dose that is ordinarily associated with symptoms of overdosage or that is likely to be life threatening, has not been defined.

The maximum human oral dose recorded was 600 mg by mouth in a 10 month old child and approximately 1500 mg in an adult, each of whom survived.

In three of the infants who died following administration of dicyclomine hydrochloride [see Warnings and Precautions (5.1) ], the blood concentrations of drug were 200 ng/mL, 220 ng/mL, and 505 ng/mL.

It is not known if dicyclomine is dialyzable.

Treatment should consist of gastric lavage, emetics and activated charcoal.

Sedatives (e.g., short-acting barbiturates, benzodiazepines) may be used for management of overt signs of excitement.

If indicated, an appropriate parenteral cholinergic agent may be used as an antidote.

DESCRIPTION

11 Dicyclomine hydrochloride is an antispasmodic and anticholinergic (antimuscarinic) agent.

Chemically, dicyclomine hydrochloride is [bicyclohexyl]-1-carboxylic acid, 2-(diethylamino) ethyl ester, hydrochloride with the following structural formula, molecular weight, and molecular formula: C19H35NO2 • HCl — M.W.

345.96 C19H35NO2 • HCl — M.W.

345.96 Dicyclomine hydrochloride, USP occurs as a fine, white, crystalline, practically odorless powder with a bitter taste.

It is soluble in water, freely soluble in alcohol and chloroform, and very slightly soluble in ether.

Dicyclomine hydrochloride capsules, USP, for oral administration, contain 10 mg of dicyclomine hydrochloride, USP.

Each capsule contains the following inactive ingredients: anhydrous lactose, colloidal silicon dioxide, FD&C Blue No.

1, gelatin, magnesium stearate, microcrystalline cellulose, pregelatinized starch (corn), sodium lauryl sulfate and titanium dioxide.

In addition the imprinting ink contains the following: black iron oxide, D&C Yellow No.

10 Aluminum Lake, FD&C Blue No.

1 Aluminum Lake, FD&C Blue No.

2 Aluminum Lake, FD&C Red No.

40 Aluminum Lake, pharmaceutical glaze and propylene glycol.

Dicyclomine hydrochloride tablets, USP, for oral administration, contain 20 mg of dicyclomine hydrochloride, USP.

In addition, each tablet contains the following inactive ingredients: anhydrous lactose, colloidal silicon dioxide, FD&C Blue No.

1 Aluminum Lake, magnesium stearate, microcrystalline cellulose, pregelatinized starch (corn) and sodium lauryl sulfate.

Dicyclomine Hydrochloride Structural Formula

CLINICAL STUDIES

14 In controlled clinical trials involving over 100 patients who received drug, 82% of patients treated for functional bowel/irritable bowel syndrome with dicyclomine hydrochloride at initial doses of 160 mg daily (40 mg 4 times daily) demonstrated a favorable clinical response compared with 55% treated with placebo (p < 0.05).

HOW SUPPLIED

16 /STORAGE AND HANDLING Product: 63629-6320 NDC: 63629-6320-1 90 CAPSULE in a BOTTLE NDC: 63629-6320-2 30 CAPSULE in a BOTTLE

RECENT MAJOR CHANGES

Warnings and Precautions: Peripheral and Central Nervous System ( 5.3 ) 07/2012

GERIATRIC USE

8.5 Geriatric Use Clinical studies of dicyclomine did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects.

Other reported clinical experience has not identified differences in responses between the elderly and younger patients.

In general, dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range in adults, reflecting the greater frequency of decreased hepatic, renal or cardiac function and of concomitant disease or other drug therapy .

Because elderly patients are more likely to have decreased renal function, care should be taken in dose selection, and it may be useful to monitor renal function.

DOSAGE FORMS AND STRENGTHS

3 Dicyclomine Hydrochloride Capsules, USP are available containing 10 mg of dicyclomine hydrochloride, USP.

• The 10 mg capsules are a hard-shell gelatin capsule with a light turquoise blue opaque cap and light turquoise blue opaque body filled with a white to off-white powder.

The capsule is axially printed with MYLAN over 1610 in black ink on both the cap and the body.

Dicyclomine Hydrochloride Tablets, USP are available containing 20 mg of dicyclomine hydrochloride, USP.

• The 20 mg tablets are blue, round, unscored tablets debossed with M over D6 on one side of the tablet and blank on the other side.

• Dicyclomine hydrochloride capsules 10 mg ( 3 ) • Dicyclomine hydrochloride tablets 20 mg ( 3 )

MECHANISM OF ACTION

12.1 Mechanism of Action Dicyclomine relieves smooth muscle spasm of the gastrointestinal tract.

Animal studies indicate that this action is achieved via a dual mechanism: • a specific anticholinergic effect (antimuscarinic) at the acetylcholine-receptor sites with approximately 1/8 the milligram potency of atropine ( in vitro , guinea pig ileum); and • a direct effect upon smooth muscle (musculotropic) as evidenced by dicyclomine’s antagonism of bradykinin- and histamine-induced spasms of the isolated guinea pig ileum.

Atropine did not affect responses to these two agonists.

In vivo studies in cats and dogs showed dicyclomine to be equally potent against acetylcholine (ACh)- or barium chloride (BaCl 2 )-induced intestinal spasm while atropine was at least 200 times more potent against effects of ACh than BaCl 2 .

Tests for mydriatic effects in mice showed that dicyclomine was approximately 1/500 as potent as atropine; antisialagogue tests in rabbits showed dicyclomine to be 1/300 as potent as atropine.

INDICATIONS AND USAGE

1 Dicyclomine hydrochloride is indicated for the treatment of patients with functional bowel/irritable bowel syndrome.

Dicyclomine is an antispasmodic and anticholinergic (antimuscarinic) agent indicated for the treatment of functional bowel/irritable bowel syndrome ( 1 )

PEDIATRIC USE

8.4 Pediatric Use Safety and effectiveness in pediatric patients have not been established.

Dicyclomine is contraindicated in infants less than 6 months of age [see Contraindications (4) ].

There are published cases reporting that the administration of dicyclomine hydrochloride to infants has been followed by serious respiratory symptoms (dyspnea, shortness of breath, breathlessness, respiratory collapse, apnea and asphyxia), seizures, syncope, pulse rate fluctuations, muscular hypotonia, and coma and death, however; no causal relationship has been established.

PREGNANCY

8.1 Pregnancy Teratogenic Effects.

Pregnancy Category B Adequate and well-controlled studies have not been conducted with dicyclomine in pregnant women at the recommended doses of 80 mg/day to 160 mg/day.

However, epidemiologic studies did not show an increased risk of structural malformations among babies born to women who took products containing dicyclomine hydrochloride at doses up to 40 mg/day during the first trimester of pregnancy.

Reproduction studies have been performed in rats and rabbits at doses up to 33 times the maximum recommended human dose based on 160 mg/day (3 mg/kg) and have revealed no evidence of harm to the fetus due to dicyclomine.

Because animal reproduction studies are not always predictive of human response, this drug should be used during pregnancy only if clearly needed.

NUSRING MOTHERS

8.3 Nursing Mothers Dicyclomine is contraindicated in women who are breastfeeding.

Dicyclomine hydrochloride is excreted in human milk.

Because of the potential for serious adverse reactions in breast-fed infants from dicyclomine, a decision should be made whether to discontinue nursing or to discontinue the drug, taking into account the importance of the drug to the mother [see Use in Specific Populations (8.4) ] .

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS • Cardiovascular conditions : worsening of conditions ( 5.2 ) • Peripheral and central nervous system : heat prostration can occur with drug use (fever and heat stroke due to decreased sweating); drug should be discontinued and supportive measures instituted ( 5.3 ) • Psychosis and delirium have been reported in patients sensitive to anticholinergic drugs (such as elderly patients and/or in patients with mental illness) : signs and symptoms resolve within 12 to 24 hours after discontinuation of dicyclomine ( 5.3 ) • Myasthenia Gravis : overdose may lead to muscular weakness and paralysis.

Dicyclomine should be given to patients with myasthenia gravis only to reduce adverse muscarinic effects of an anticholinesterase ( 5.4 ) • Incomplete intestinal obstruction : diarrhea may be an early symptom especially in patients with ileostomy or colostomy.

Treatment with dicyclomine would be inappropriate and possibly fatal ( 5.5 ) • Salmonella dysenteric patients : due to risk of toxic megacolon ( 5.6 ) • Ulcerative colitis : Dicyclomine should be used with caution in these patients; large doses may suppress intestinal motility or aggravate the serious complications of toxic megacolon ( 5.7 ) • Prostatic hypertrophy : Dicyclomine should be used with caution in these patients; may lead to urinary retention ( 5.8 ) • Hepatic and renal disease : should be used with caution ( 5.9 ) • Geriatric : use with caution in elderly who may be more susceptible to dicyclomine’s adverse events ( 5.10 ) 5.2 Cardiovascular Conditions Dicyclomine hydrochloride needs to be used with caution in conditions characterized by tachyarrhythmia such as thyrotoxicosis, congestive heart failure and in cardiac surgery, where they may further accelerate the heart rate.

Investigate any tachycardia before administration of dicyclomine hydrochloride.

Care is required in patients with coronary heart disease, as ischemia and infarction may be worsened, and in patients with hypertension [see Adverse Reactions (6.3) ] .

5.3 Peripheral and Central Nervous System The peripheral effects of dicyclomine hydrochloride are a consequence of their inhibitory effect on muscarinic receptors of the autonomic nervous system.

They include dryness of the mouth with difficulty in swallowing and talking, thirst, reduced bronchial secretions, dilatation of the pupils (mydriasis) with loss of accommodation (cycloplegia) and photophobia, flushing and dryness of the skin, transient bradycardia followed by tachycardia, with palpitations and arrhythmias, and difficulty in micturition, as well as reduction in the tone and motility of the gastrointestinal tract leading to constipation [see Adverse Reactions (6) ] .

In the presence of high environmental temperature heat prostration can occur with drug use (fever and heat stroke due to decreased sweating).

It should also be used cautiously in patients with fever.

If symptoms occur, the drug should be discontinued and supportive measures instituted.

Because of the inhibitory effect on muscarinic receptors within the autonomic nervous system, caution should be taken in patients with autonomic neuropathy.

Central nervous system (CNS) signs and symptoms include confusional state, disorientation, amnesia, hallucinations, dysarthria, ataxia, coma, euphoria, fatigue, insomnia, agitation and mannerisms and inappropriate affect.

Psychosis and delirium have been reported in sensitive individuals (such as elderly patients and/or in patients with mental illness) given anticholinergic drugs.

These CNS signs and symptoms usually resolve within 12 to 24 hours after discontinuation of the drug.

Dicyclomine may produce drowsiness, dizziness or blurred vision.

The patient should be warned not to engage in activities requiring mental alertness, such as operating a motor vehicle or other machinery or performing hazardous work while taking dicyclomine.

5.4 Myasthenia Gravis With overdosage, a curare-like action may occur (i.e., neuromuscular blockade leading to muscular weakness and possible paralysis).

It should not be given to patients with myasthenia gravis except to reduce adverse muscarinic effects of an anticholinesterase [see Contraindications (4) ].

5.5 Intestinal Obstruction Diarrhea may be an early symptom of incomplete intestinal obstruction, especially in patients with ileostomy or colostomy.

In this instance, treatment with this drug would be inappropriate and possibly harmful [see Contraindications (4) ].

Rarely development of Ogilvie’s syndrome (colonic pseudo-obstruction) has been reported.

Ogilvie’s syndrome is a clinical disorder with signs, symptoms and radiographic appearance of an acute large bowel obstruction but with no evidence of distal colonic obstruction 5.6 Toxic Dilatation of Intestinemegacolon Toxic dilatation of intestine and intestinal perforation is possible when anticholinergic agents are administered in patients with Salmonella dysentery.

5.7 Ulcerative Colitis Caution should be taken in patients with ulcerative colitis.

Large doses may suppress intestinal motility to the point of producing a paralytic ileus and the use of this drug may precipitate or aggravate the serious complication of toxic megacolon [see Adverse Reactions (6.3) ] .

Dicyclomine is contraindicated in patients with severe ulcerative colitis [see Contraindications (4) ].

5.8 Prostatic Hypertrophy Dicyclomine should be used with caution in patients with known or suspected prostatic enlargement, in whom prostatic enlargement may lead to urinary retention [see Adverse Reactions (6.3) ] .

5.9 Hepatic and Renal Disease Dicyclomine should be used with caution in patients with known hepatic and renal impairment.

5.10 Geriatric Population Dicyclomine hydrochloride should be used with caution in elderly who may be more susceptible to its adverse effects.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION 17.2 Use in Infants Inform parents and caregivers not to administer dicyclomine in infants less than 6 months of age [see Use in Specific Populations (8.4)] .

17.3 Use in Nursing Mothers Advise lactating women that dicyclomine should not be used while breastfeeding their infants [see Use in Specific Populations (8.3 , 8.4) ] .

17.4 Peripheral and Central Nervous System In the presence of a high environmental temperature, heat prostration can occur with dicyclomine use (fever and heat stroke due to decreased sweating).

If symptoms occur, the drug should be discontinued and a physician contacted.

Dicyclomine may produce drowsiness or blurred vision.

The patient should be warned not to engage in activities requiring mental alertness, such as operating a motor vehicle or other machinery or to perform hazardous work while taking dicyclomine [see Warnings and Precautions (5.3) ].

Mylan Pharmaceuticals Inc.

Morgantown, WV 26505 U.S.A.

REVISED FEBRUARY 2016 DICY:R6

DOSAGE AND ADMINISTRATION

2 Dosage must be adjusted to individual patient needs.

Dosage for dicyclomine must be adjusted to individual patient needs ( 2 ).

If a dose is missed, patients should continue the normal dosing schedule ( 2 ).

Oral in adults ( 2.1 ): • Starting dose: 20 mg 4 times a day.

After a week treatment with the starting dose, the dose may be escalated to 40 mg 4 times a day, unless side effects limit dosage escalation • Discontinue dicyclomine if efficacy not achieved or side effects require doses less than 80 mg per day after 2 weeks of treatment 2.1 Oral Dosage and Administration in Adults The recommended initial dose is 20 mg 4 times a day.

After one week treatment with the initial dose, the dose may be increased to 40 mg 4 times a day unless side effects limit dosage escalation.

If efficacy is not achieved within 2 weeks or side effects require doses below 80 mg per day, the drug should be discontinued.

Documented safety data are not available for doses above 80 mg daily for periods longer than 2 weeks.

amoxicillin (as amoxicillin trihydrate) 250 MG per 5 ML Oral Suspension

DRUG INTERACTIONS

7 Probenicid decreases renal tubular secretion of amoxicillin which may result in increased blood levels of amoxicillin.

( 7.1 ) Concomitant use of Amoxicillin and oral anticoagulants may increase the prolongation of prothrombin time.

( 7.2 ) Coadministration with allopurinol increases the risk of rash.

( 7.3 ) Amoxicillin may reduce the efficacy of oral contraceptives.

( 7.4 ) 7.1 Probenecid Probenecid decreases the renal tubular secretion of amoxicillin.

Concurrent use of amoxicillin and probenecid may result in increased and prolonged blood levels of amoxicillin.

7.2 Oral Anticoagulants Abnormal prolongation of prothrombin time (increased international normalized ratio [INR]) has been reported in patients receiving amoxicillin and oral anticoagulants.

Appropriate monitoring should be undertaken when anticoagulants are prescribed concurrently.

Adjustments in the dose of oral anticoagulants may be necessary to maintain the desired level of anticoagulation.

7.3 Allopurinol The concurrent administration of allopurinol and amoxicillin increases the incidence of rashes in patients receiving both drugs as compared to patients receiving amoxicillin alone.

It is not known whether this potentiation of amoxicillin rashes is due to allopurinol or the hyperuricemia present in these patients.

7.4 Oral Contraceptives AMOXICILLIN may affect the gut flora, leading to lower estrogen reabsorption and reduced efficacy of combined oral estrogen/progesterone contraceptives.

7.5 Other Antibacterials Chloramphenicol, macrolides, sulfonamides, and tetracyclines may interfere with the bactericidal effects of penicillin.

This has been demonstrated in vitro; however, the clinical significance of this interaction is not well documented.

7.6 Effects on Laboratory Tests High urine concentrations of ampicillin may result in false-positive reactions when testing for the presence of glucose in urine using CLINITEST ® , Benedict’s Solution, or Fehling’s Solution.

Since this effect may also occur with amoxicillin, it is recommended that glucose tests based on enzymatic glucose oxidase reactions (such as CLINISTIX ® ) be used.

Following administration of ampicillin or amoxicillin to pregnant women, a transient decrease in plasma concentration of total conjugated estriol, estriol-glucuronide, conjugated estrone, and estradiol has been noted.

OVERDOSAGE

10 In case of overdosage, discontinue medication, treat symptomatically, and institute supportive measures as required.

A prospective study of 51 pediatric patients at a poison-control center suggested that overdosages of less than 250 mg/kg of amoxicillin are not associated with significant clinical symptoms.

Interstitial nephritis resulting in oliguric renal failure has been reported in a small number of patients after overdosage with amoxicillin1.

Crystalluria, in some cases leading to renal failure, has also been reported after amoxicillin overdosage in adult and pediatric patients.

In case of overdosage, adequate fluid intake and diuresis should be maintained to reduce the risk of amoxicillin crystalluria.

Renal impairment appears to be reversible with cessation of drug administration.

High blood levels may occur more readily in patients with impaired renal function because of decreased renal clearance of amoxicillin.

Amoxicillin may be removed from circulation by hemodialysis.

DESCRIPTION

11 Formulations of AMOXICILLIN contain amoxicillin, a semisynthetic antibiotic, an analog of ampicillin, with a broad spectrum of bactericidal activity against many gram-positive and gram-negative microorganisms.

Chemically, it is (2 S ,5 R ,6 R )-6-[( R )-(-)-2-amino-2-( p -hydroxyphenyl)acetamido]-3,3-dimethyl-7-oxo-4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylic acid trihydrate.

It may be represented structurally as: The amoxicillin molecular formula is C 16 H 19 N 3 O 5 S•3H 2 O, and the molecular weight is 419.45.

Capsules: Each capsule of AMOXICILLIN, with royal blue opaque cap and pink opaque body, contains 250 mg or 500 mg amoxicillin as the trihydrate.

The cap and body of the 250-mg capsule are imprinted with the product name AMOXIL and 250; the cap and body of the 500 mg capsule are imprinted with AMOXIL and 500.

Inactive ingredients: D&C Red No.

28, FD&C Blue No.

1, FD&C Red No.

40, gelatin, magnesium stearate, and titanium dioxide.

Tablets: Each tablet contains 500 mg or 875 mg amoxicillin as the trihydrate.

Each film-coated, capsule-shaped, pink tablet is debossed with AMOXIL centered over 500 or 875, respectively.

The 875-mg tablet is scored on the reverse side.

Inactive ingredients: Colloidal silicon dioxide, crospovidone, FD&C Red No.

30 aluminum lake, hypromellose, magnesium stearate, microcrystalline cellulose, polyethylene glycol, sodium starch glycolate, and titanium dioxide.

Powder for Oral Suspension: Each 5 mL of reconstituted suspension contains 125 mg, 200 mg, 250 mg or 400 mg amoxicillin as the trihydrate.

Each 5 mL of the 125-mg reconstituted suspension contains 0.11 mEq (2.51 mg) of sodium.

Each 5 mL of the 200-mg reconstituted suspension contains 0.15 mEq (3.39 mg) of sodium.

Each 5 mL of the 250 mg reconstituted suspension contains 0.15 mEq (3.36 mg) of sodium; each 5 mL of the 400 mg reconstituted suspension contains 0.19 mEq (4.33 mg) of sodium.

Inactive ingredients: FD&C Red No.

3, flavorings, silica gel, sodium benzoate, sodium citrate, sucrose, and xanthan gum.

amoxicillin-chemstruc

CLINICAL STUDIES

14 14.1 H.

pylori Eradication to Reduce the Risk of Duodenal Ulcer Recurrence Randomized, double-blind clinical studies performed in the United States in patients with H.

pylori and duodenal ulcer disease (defined as an active ulcer or history of an ulcer within 1 year) evaluated the efficacy of lansoprazole in combination with amoxicillin capsules and clarithromycin tablets as triple 14 day therapy, or in combination with amoxicillin capsules as dual 14 day therapy, for the eradication of H.

pylori.

Based on the results of these studies, the safety and efficacy of 2 different eradication regimens were established: Triple therapy : Amoxicillin 1 gram twice daily/clarithromycin 500 mg twice daily/lansoprazole 30 mg twice daily (see Table 6) .

Dual therapy : Amoxicillin 1 gram three times daily/lansoprazole 30 mg three times daily (see Table 7) .

All treatments were for 14 days.

H.

pylori eradication was defined as 2 negative tests (culture and histology) at 4 to 6 weeks following the end of treatment.

Triple therapy was shown to be more effective than all possible dual therapy combinations.

Dual therapy was shown to be more effective than both monotherapies.

Eradication of H.

pylori has been shown to reduce the risk of duodenal ulcer recurrence.

Table 6.

H.

pylori Eradication Rates When Amoxicillin is Administered as Part of a Triple Therapy Regimen Study Triple Therapy Triple Therapy ​Evaluable Analysis a [95% Confidence Interval] (number of patients) Intent-to-Treat Analysis b [95% Confidence Interval] (number of patients) Study 1 92 [80.0 – 97.7] (n = 48) 86 [73.3 – 93.5] (n = 55) Study 2 86 [75.7 – 93.6] (n = 66) 83 [72.0 – 90.8] (n = 70) a This analysis was based on evaluable patients with confirmed duodenal ulcer (active or within 1 year) and H.

pylori infection at baseline defined as at least 2 of 3 positive endoscopic tests from CLOtest®, histology, and/or culture.

Patients were included in the analysis if they completed the study.

Additionally, if patients dropped out of the study due to an adverse event related to the study drug, they were included in the analysis as failures of therapy.

b Patients were included in the analysis if they had documented H.

pylori infection at baseline as defined above and had a confirmed duodenal ulcer (active or within 1 year).

All dropouts were included as failures of therapy.

Table 7.

H.

pylori Eradication Rates When Amoxicillin is Administered as Part of a Dual Therapy Regimen Study Dual Therapy Dual Therapy Evaluable Analysis a [95% Confidence Interval] (number of patients) Intent-to-Treat Analysis b [95% Confidence Interval] (number of patients) Study 1 77 [62.5 – 87.2] (n = 51) 70 [56.8 – 81.2] (n = 60) Study 2 66 [51.9 – 77.5] (n = 58) 61 [48.5 – 72.9] (n = 67) a This analysis was based on evaluable patients with confirmed duodenal ulcer (active or within 1 year) and H.

pylori infection at baseline defined as at least 2 of 3 positive endoscopic tests from CLOtest®, histology, and/or culture.

Patients were included in the analysis if they completed the study.

Additionally, if patients dropped out of the study due to an adverse event related to the study drug, they were included in the analysis as failures of therapy.

b Patients were included in the analysis if they had documented H.

pylori infection at baseline as defined above and had a confirmed duodenal ulcer (active or within 1 year).

All dropouts were included as failures of therapy.

HOW SUPPLIED

16 /STORAGE AND HANDLING Capsules: Each capsule of AMOXICILLIN, with royal blue opaque cap and pink opaque body, contains 250 mg or 500 mg amoxicillin as the trihydrate.

The cap and body of the 250-mg capsule are imprinted with the product name AMOXIL and 250; the cap and body of the 500 mg capsule are imprinted with AMOXIL and 500 250-mg Capsule NDC 43598-225-01 Bottles of 100 NDC 43598-225-05 Bottles of 500 500-mg Capsule NDC 43598-205-01 Bottles of 100 NDC 43598-205-05 Bottles of 500 Tablets: Each tablet contains 500 mg or 875 mg amoxicillin as the trihydrate.

Each film-coated, capsule-shaped, pink tablet is debossed with AMOXIL centered over 500 or 875, respectively.

The 875-mg tablet is scored on the reverse side.

500-mg Tablet NDC 43598-224-14 Bottles of 20 NDC 43598-224-01 Bottles of 100 NDC 43598-224-05 Bottles of 500 875-mg Tablet NDC 43598-219-14 Bottles of 20 NDC 43598-219-01 Bottles of 100 Powder for Oral Suspension: Each 5 mL of reconstituted strawberry-flavored suspension contains 125 mg amoxicillin as the trihydrate.

Each 5 mL of reconstituted bubble-gum-flavored suspension contains 200 mg, 250 mg or 400 mg amoxicillin as the trihydrate.

125 mg/5 mL NDC 43598-222-80 80-mL bottle NDC 43598-222-52 100-mL bottle NDC 43598-222-53 150-mL bottle 200 mg/5 mL NDC 43598-223-50 50-mL bottle NDC 43598-223-51 75-mL bottle NDC 43598-223-52 100-mL bottle 250 mg/5 mL NDC 43598-209-80 80-mL bottle NDC 43598-209-52 100-mL bottle NDC 43598-209-53 150-mL bottle 400 mg/5 mL NDC 43598-207-50 50-mL bottle NDC 43598-207-51 75-mL bottle NDC 43598-207-52 100-mL bottle Store at or below 25ºC (77ºF) 250 mg and 500 mg Capsules 500 mg and 875 mg Tablets 200 mg and 400 mg unreconstituted powder Store Dry Powder at 20ºC-25ºC (68ºF-77ºF) 125 mg and 250 mg unreconstituted powder

RECENT MAJOR CHANGES

Indications and Usage, Gonorrhea ( 1.5 )………………………………………………………………………………

Removed 9/2015 Dosage and Administration, Gonorrhea ( 2.1 ) …………………………………………………………………………Removed 9/2015

DOSAGE FORMS AND STRENGTHS

3 Capsules: 250 mg, 500 mg.

Each capsule of AMOXICILLIN, with royal blue opaque cap and pink opaque body, contains 250 mg or 500 mg amoxicillin as the trihydrate.

The cap and body of the 250-mg capsule are imprinted with the product name AMOXIL and 250; the cap and body of the 500 mg capsule are imprinted with AMOXIL and 500.

Tablets: 500 mg, 875 mg.

Each tablet contains 500 mg or 875 mg amoxicillin as the trihydrate.

Each film-coated, capsule-shaped, pink tablet is debossed with AMOXIL centered over 500 or 875, respectively.

The 875-mg tablet is scored on the reverse side.

Powder for Oral Suspension: 125 mg/5 mL, 200 mg/5 mL, 250 mg/5 mL, 400 mg/5 mL.

Each 5 mL of reconstituted strawberry-flavored suspension contains 125 mg amoxicillin as the trihydrate.

Each 5 mL of reconstituted bubble-gum-flavored suspension contains 200 mg, 250 mg or 400 mg amoxicillin as the trihydrate.

Capsules: 250 mg, 500 mg ( 3 ) Tablets: 500 mg, 875 mg ( 3 ) Powder for Oral Suspension: 125 mg/5 mL, 200 mg/5 mL, 250 mg/5 mL, 400 mg/5 mL ( 3 )

INDICATIONS AND USAGE

1 AMOXICILLIN is a penicillin-class antibacterial indicated for treatment of infections due to susceptible strains of designated microorganisms.

Infections of the ear, nose, throat, genitourinary tract, skin and skin structure, and lower respiratory tract.

( 1.1 – 1.4 ) In combination for treatment of H.

pylori infection and duodenal ulcer disease.

( 1.5 ) To reduce the development of drug-resistant bacteria and maintain the effectiveness of AMOXICILLIN and other antibacterial drugs, AMOXICILLIN should be used only to treat infections that are proven or strongly suspected to be caused by bacteria.

( 1.6 ) 1.1 Infections of the Ear, Nose, and Throat AMOXICILLIN is indicated in the treatment of infections due to susceptible (ONLY β-lactamase–negative) isolates of Streptococcus species.

(α and β hemolytic isolates only), Streptococcus pneumoniae , Staphylococcus spp., or Haemophilus influenzae .

1.2 Infections of the Genitourinary Tract AMOXICILLIN is indicated in the treatment of infections due to susceptible (ONLY β-lactamase–negative) isolates of Escherichia coli , Proteus mirabilis , or Enterococcus faecalis .

1.3 Infections of the Skin and Skin Structure AMOXICILLIN is indicated in the treatment of infections due to susceptible (ONLY β-lactamase–negative) isolates of Streptococcus spp.

(α and β hemolytic isolates only), Staphylococcus spp., or E.

coli.

1.4 Infections of the Lower Respiratory Tract AMOXICILLIN is indicated in the treatment of infections due to susceptible (ONLY β-lactamase–negative) isolates of Streptococcus spp.

(α and β hemolytic isolates only), S.

pneumoniae , S taphylococcus spp ., or H.

influenzae .

1.5 Helicobacter pylori Infection Triple therapy for Helicobacter pylori with clarithromycin and lansoprazole: AMOXICILLIN, in combination with clarithromycin plus lansoprazole as triple therapy, is indicated for the treatment of patients with H.

pylori infection and duodenal ulcer disease (active or 1 year history of a duodenal ulcer) to eradicate H.

pylori .

Eradication of H.

pylori has been shown to reduce the risk of duodenal ulcer recurrence.

Dual therapy for H.

pylori with lansoprazole : AMOXICILLIN, in combination with lansoprazole delayed release capsules as dual therapy, is indicated for the treatment of patients with H.

pylori infection and duodenal ulcer disease (active or 1 year history of a duodenal ulcer) who are either allergic or intolerant to clarithromycin or in whom resistance to clarithromycin is known or suspected .

(See the clarithromycin package insert, MICROBIOLOGY.) Eradication of H.

pylori has been shown to reduce the risk of duodenal ulcer recurrence.

1.6 Usage To reduce the development of drug resistant bacteria and maintain the effectiveness of AMOXICILLIN and other antibacterial drugs, AMOXICILLIN should be used only to treat infections that are proven or strongly suspected to be caused by bacteria.

When culture and susceptibility information are available, they should be considered in selecting or modifying antibacterial therapy.

In the absence of such data, local epidemiology and susceptibility patterns may contribute to the empiric selection of therapy.

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS Anaphylactic reactions: Serious and occasionally fatal anaphylactic reactions have been reported in patients on penicillin therapy.

Serious anaphylactic reactions require immediate emergency treatment with supportive measures.

( 5.1 ) Clostridium difficile -associated diarrhea (ranging from mild diarrhea to fatal colitis): Evaluate if diarrhea occurs.

( 5.2 ) 5.1 Anaphylactic Reactions Serious and occasionally fatal hypersensitivity (anaphylactic) reactions have been reported in patients on penicillin therapy including amoxicillin.

Although anaphylaxis is more frequent following parenteral therapy, it has occurred in patients on oral penicillins.

These reactions are more likely to occur in individuals with a history of penicillin hypersensitivity and/or a history of sensitivity to multiple allergens.

There have been reports of individuals with a history of penicillin hypersensitivity who have experienced severe reactions when treated with cephalosporins.

Before initiating therapy with AMOXICILLIN, careful inquiry should be made regarding previous hypersensitivity reactions to penicillins, cephalosporins, or other allergens.

If an allergic reaction occurs, AMOXICILLIN should be discontinued and appropriate therapy instituted.

5.2 Clostridium difficile Associated Diarrhea Clostridium difficile associated diarrhea (CDAD) has been reported with use of nearly all antibacterial agents, including AMOXICILLIN, and may range in severity from mild diarrhea to fatal colitis.

Treatment with antibacterial agents alters the normal flora of the colon leading to overgrowth of C.

difficile .

C.

difficile produces toxins A and B which contribute to the development of CDAD.

Hypertoxin-producing strains of C.

difficile cause increased morbidity and mortality, as these infections can be refractory to antimicrobial therapy and may require colectomy.

CDAD must be considered in all patients who present with diarrhea following antibacterial use.

Careful medical history is necessary since CDAD has been reported to occur over 2 months after the administration of antibacterial agents.

If CDAD is suspected or confirmed, ongoing antibiotic use not directed against C.

difficile may need to be discontinued.

Appropriate fluid and electrolyte management, protein supplementation, antibiotic treatment of C.

difficile , and surgical evaluation should be instituted as clinically indicated.

5.3 Development of Drug-Resistant Bacteria Prescribing AMOXICILLIN in the absence of a proven or strongly suspected bacterial infection is unlikely to provide benefit to the patient and increases the risk of the development of drug-resistant bacteria.

5.4 Use in Patients With Mononucleosis A high percentage of patients with mononucleosis who receive amoxicillin develop an erythematous skin rash.

Thus amoxicillin should not be administered to patients with mononucleosis.

5.5 Phenylketonurics Amoxicillin chewable tablets contain aspartame which contains phenylalanine.

Each 200 mg chewable tablet contains 1.82 mg phenylalanine; each 400 mg chewable tablet contains 3.64 mg phenylalanine.

The oral suspensions of Amoxicillin do not contain phenylalanine and can be used by phenylketonurics.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION Information for Patients Patients should be advised that AMOXICILLIN may be taken every 8 hours or every 12 hours, depending on the dose prescribed.

Patients should be counseled that antibacterial drugs, including AMOXICILLIN, should only be used to treat bacterial infections.

They do not treat viral infections (e.g., the common cold).

When AMOXICILLIN is prescribed to treat a bacterial infection, patients should be told that although it is common to feel better early in the course of therapy, the medication should be taken exactly as directed.

Skipping doses or not completing the full course of therapy may: (1) decrease the effectiveness of the immediate treatment, and (2) increase the likelihood that bacteria will develop resistance and will not be treatable by AMOXICILLIN or other antibacterial drugs in the future.

Patients should be counseled that diarrhea is a common problem caused by antibiotics, and it usually ends when the antibiotic is discontinued.

Sometimes after starting treatment with antibiotics, patients can develop watery and bloody stools (with or without stomach cramps and fever) even as late as 2 or more months after having taken their last dose of the antibiotic.

If this occurs, patients should contact their physician as soon as possible.

Patients should be aware that AMOXICILLIN contains a penicillin class drug product that can cause allergic reactions in some individuals.

AMOXIL is registered trademark of GlaxoSmithKline and is licensed to Dr.

Reddy’s Laboratories Inc.

Manufactured.

By: Dr.

Reddy’s Laboratories Tennessee LLC.

Bristol, TN 37620 Issued: 052016

DOSAGE AND ADMINISTRATION

2 In adults, 750-1750 mg/day in divided doses every 8-12 hours.

In Pediatric Patients > 3 Months of Age, 20-45 mg/kg/day in divided doses every 8-12 hours.

Refer to full prescribing information for specific dosing regimens.

( 2.1 , 2.2 , 2.3 ) The upper dose for neonates and infants ≤ 3 months is 30 mg/kg/day divided every 12 hours.

( 2.2 ) Dosing for H.

pylori Infection: Triple therapy: 1 gram AMOXICILLIN, 500 mg clarithromycin, and 30 mg lansoprazole, all given twice daily (every 12 hours) for 14 days.

Dual therapy: 1 gram AMOXICILLIN and 30 mg lansoprazole, each given three times daily (every 8 hours) for 14 days.

( 2.3 ) Reduce the dose in patients with severe renal impairment (GFR <30 mL/min).

( 2.4 ) 2.1 Dosing for Adult and Pediatric Patients > 3 Months of Age Treatment should be continued for a minimum of 48 to 72 hours beyond the time that the patient becomes asymptomatic or evidence of bacterial eradication has been obtained.

It is recommended that there be at least 10 days treatment for any infection caused by Streptococcus pyogenes to prevent the occurrence of acute rheumatic fever.

In some infections, therapy may be required for several weeks.

It may be necessary to continue clinical and/or bacteriological follow-up for several months after cessation of therapy.

Table 1.

Dosing Recommendations for Adult and Pediatric Patients > 3 Months of Age Infection Severity a Usual Adult Dose Usual Dose for Children > 3 Months b Ear/Nose/Throat Skin/Skin Structure Genitourinary Tract Mild/Moderate 500 mg every 12 hours or 250 mg every 8 hours 25 mg/kg/day in divided doses every 12 hours or 20 mg/kg/day in divided doses every 8 hours Severe 875 mg every 12 hours or 500 mg every 8 hours 45 mg/kg/day in divided doses every 12 hours or 40 mg/kg/day in divided doses every 8 hours Lower Respiratory Tract Mild/Moderate or Severe 875 mg every 12 hours or 500 mg every 8 hours 45 mg/kg/day in divided doses every 12 hours or 40 mg/kg/day in divided doses every 8 hours a Dosing for infections caused by bacteria that are intermediate in their susceptibility to amoxicillin should follow the recommendations for severe infections.

b The children’s dosage is intended for individuals whose weight is less than 40 kg.

Children weighing 40 kg or more should be dosed according to the adult recommendations.

2.2 Dosing in Neonates and Infants Aged ≤ 12 Weeks (≤ 3 Months) Treatment should be continued for a minimum of 48 to 72 hours beyond the time that the patient becomes asymptomatic or evidence of bacterial eradication has been obtained.

It is recommended that there be at least 10 days’ treatment for any infection caused by Streptococcus pyogenes to prevent the occurrence of acute rheumatic fever.

Due to incompletely developed renal function affecting elimination of amoxicillin in this age group, the recommended upper dose of AMOXICILLIN is 30 mg/kg/day divided every 12 hours.

There are currently no dosing recommendations for pediatric patients with impaired renal function.

2.3 Dosing for H.

pylori Infection Triple therapy: The recommended adult oral dose is 1 gram AMOXICILLIN, 500 mg clarithromycin, and 30 mg lansoprazole, all given twice daily (every 12 hours) for 14 days.

Dual therapy: The recommended adult oral dose is 1 gram AMOXICILLIN and 30 mg lansoprazole, each given three times daily (every 8 hours) for 14 days.

Please refer to clarithromycin and lansoprazole full prescribing information.

2.4 Dosing in Renal Impairment Patients with impaired renal function do not generally require a reduction in dose unless the impairment is severe.

Severely impaired patients with a glomerular filtration rate of < 30 mL/min.

should not receive a 875 mg dose.

Patients with a glomerular filtration rate of 10 to 30 mL/min should receive 500 mg or 250 mg every 12 hours, depending on the severity of the infection.

Patients with a glomerular filtration rate less than 10 mL/min should receive 500 mg or 250 mg every 24 hours, depending on severity of the infection.

Hemodialysis patients should receive 500 mg or 250 mg every 24 hours, depending on severity of the infection.

They should receive an additional dose both during and at the end of dialysis.

2.5 Directions for Mixing Oral Suspension Tap bottle until all powder flows freely.

Add approximately 1/3 of the total amount of water for reconstitution (see Table 2) and shake vigorously to wet powder.

Add remainder of the water and again shake vigorously.

Table 2.

Amount of Water for Mixing Oral Suspension Strength Bottle Size Amount of Water Required for Reconstitution Oral Suspension 125 mg/5 mL 80 mL 62 mL 100 mL 78 mL 150 mL 116 mL Oral Suspension 200 mg/5 mL 50 mL 39 mL 75 mL 57 mL 100 mL 76 mL Oral Suspension 250 mg/5 mL 80 mL 59 mL 100 mL 74 mL 150 mL 111 mL Oral Suspension 400 mg/5 mL 50 mL 36 mL 75 mL 54 mL 100 mL 71 mL After reconstitution, the required amount of suspension should be placed directly on the child’s tongue for swallowing.

Alternate means of administration are to add the required amount of suspension to formula, milk, fruit juice, water, ginger ale, or cold drinks.

These preparations should then be taken immediately.

NOTE: SHAKE ORAL SUSPENSION WELL BEFORE USING.

Keep bottle tightly closed.

Any unused portion of the reconstituted suspension must be discarded after 14 days.

Refrigeration is preferable, but not required.

Diltiazem Hydrochloride 120 MG Oral Tablet

WARNINGS

1.

Cardiac Conduction Diltiazem prolongs AV node refractory periods without significantly prolonging sinus node recovery time, except in patients with sick sinus syndrome.

This effect may rarely result in abnormally slow heart rates (particularly in patients with sick sinus syndrome) or second- or-third-degree AV block (six of 1,243 patients for 0.48%).

Concomitant use of diltiazem with beta-blockers or digitalis may result in additive effects on cardiac conduction.

A patient with Prinzmetal’s angina developed periods of asystole (2 to 5 seconds) after a single dose of 60 mg of diltiazem.

(See ADVERSE REACTIONS .) 2.

Congestive Heart Failure Although diltiazem has a negative inotropic effect in isolated animal tissue preparations, hemodynamic studies in humans with normal ventricular function have not shown a reduction in cardiac index nor consistent negative effects on contractility (dp/dt).

Experience with the use of diltiazem alone or in combination with beta-blockers in patients with impaired ventricular function is very limited.

Caution should be exercised when using the drug in such patients.

3.

Hypotension Decreases in blood pressure associated with diltiazem therapy may occasionally result in symptomatic hypotension.

4.

Acute Hepatic Injury In rare instances, significant elevations in enzymes such as alkaline phosphatase, LDH, SGOT, SGPT and other phenomena consistent with acute hepatic injury have been noted.

These reactions have been reversible upon discontinuation of drug therapy.

The relationship to diltiazem is uncertain in most cases, but probable in some.

(See PRECAUTIONS .)

DRUG INTERACTIONS

Drug Interactions Due to the potential for additive effects, caution and careful titration are warranted in patients receiving diltiazem concomitantly with any agents known to affect cardiac contractility and/or conduction.

(See WARNINGS .) Pharmacologic studies indicate that there may be additive effects in prolonging AV conduction when using beta-blockers or digitalis concomitantly with diltiazem.

(See WARNINGS .) As with all drugs, care should be exercised when treating patients with multiple medications.

Diltiazem is both a substrate and an inhibitor of the cytochrome P-450 3A4 enzyme treatment.

Other drugs that are specific substrates, inhibitors, or inducers of this enzyme system may have a significant impact on the efficacy and side effect profile of diltiazem.

Patients taking other drugs that are substrates of CYP450 3A4, especially patients with renal and/or hepatic impairment, may require dosage adjustment when starting or stopping concomitantly administered diltiazem in order to maintain optimum therapeutic blood levels.

Anesthetics The depression of cardiac contractility, conductivity, and automaticity, as well as the vascular dilation associated with anesthetics, may be potentiated by calcium channel blockers.

When used concomitantly, anesthetics and calcium blockers should be titrated carefully.

Benzodiazepines Studies showed that diltiazem increased the AUC of midazolam and triazolam by 3- to 4-fold and the C max by 2-fold, compared to placebo.

The elimination half-life of midazolam and triazolam also increased (1.5- to 2.5-fold) during coadministration with diltiazem.

These pharmacokinetic effects seen during diltiazem coadministration can result in increased clinical effects ( e.g ., prolonged sedation) of both midazolam and triazolam.

Beta-Blockers Controlled and uncontrolled domestic studies suggest that concomitant use of diltiazem and beta-blockers is usually well tolerated.

Available data are not sufficient, however, to predict the effects of concomitant treatment, particularly in patients with left ventricular dysfunction or cardiac conduction abnormalities.

Administration of diltiazem concomitantly with propranolol in five normal volunteers resulted in increased propranolol levels in all subjects, and bioavailability of propranolol was increased approximately 50%.

In vitro , propranolol appears to be displaced from its binding sites by diltiazem.

If combination therapy is initiated or withdrawn in conjunction with propranolol, an adjustment in the propranolol dose may be warranted.

(See WARNINGS .) Buspirone In nine healthy subjects, diltiazem significantly increased the mean buspirone AUC 5.5-fold and C max 4.1-fold compared to placebo.

The T 1/2 and T max of buspirone were not significantly affected by diltiazem.

Enhanced effects and increased toxicity of buspirone may be possible during concomitant administration with diltiazem.

Subsequent dose adjustments may be necessary during coadministration, and should be based on clinical assessment.

Carbamazepine Concomitant administration of diltiazem with carbamazepine has been reported to result in elevated serum levels of carbamazepine (40% to 72% increase) resulting in toxicity in some cases.

Patients receiving these drugs concurrently should be monitored for a potential drug interaction.

Cimetidine A study in six healthy volunteers has shown a significant increase in peak diltiazem plasma levels (58%) and area-under-the-curve (53%) after a one-week course of cimetidine at 1,200 mg per day and a single dose of diltiazem 60 mg.

Ranitidine produced smaller, non-significant increases.

The effect may be mediated by cimetidine’s known inhibition of hepatic cytochrome P-450, the enzyme system responsible for the first-pass metabolism of diltiazem.

Patients currently receiving diltiazem therapy should be carefully monitored for a change in pharmacological effect when initiating and discontinuing therapy with cimetidine.

An adjustment in the diltiazem dose may be warranted.

Clonidine Sinus bradycardia resulting in hospitalization and pacemaker insertion has been reported in association with the use of clonidine concurrently with diltiazem.

Monitor heart rate in patients receiving concomitant diltiazem and clonidine.

Cyclosporine A pharmacokinetic interaction between diltiazem and cyclosporine has been observed during studies involving renal and cardiac transplant patients.

In renal and cardiac transplant recipients, a reduction of cyclosporine trough dose ranging from 15% to 48% was necessary to maintain cyclosporine trough concentrations similar to those seen prior to the addition of diltiazem.

If these agents are to be administered concurrently, cyclosporine concentrations should be monitored, especially when diltiazem therapy is initiated, adjusted or discontinued.

The effect of cyclosporine on diltiazem plasma concentrations has not been evaluated.

Digitalis Administration of diltiazem with digoxin in 24 healthy male subjects increased plasma digoxin concentrations approximately 20%.

Another investigator found no increase in digoxin levels in 12 patients with coronary artery disease.

Since there have been conflicting results regarding the effect of digoxin levels, it is recommended that digoxin levels be monitored when initiating, adjusting, and discontinuing diltiazem therapy to avoid possible over- or under-digitalization.

(See WARNINGS.) Quinidine Diltiazem significantly increases the AUC (0→∞) of quinidine by 51%, T 1/2 by 36%, and decreases its CL oral by 33%.

Monitoring for quinidine adverse effects may be warranted and the dose adjusted accordingly.

Rifampin Coadministration of rifampin with diltiazem lowered the diltiazem plasma concentrations to undetectable levels.

Coadministration of diltiazem with rifampin or any known CYP3A4 inducer should be avoided when possible, and alternative therapy considered.

Statins Diltiazem is an inhibitor of CYP3A4 and has been shown to increase significantly the AUC of some statins.

The risk of myopathy and rhabdomyolysis with statins metabolized by CYP3A4 may be increased with concomitant use of diltiazem.

When possible, use a non-CYP3A4-metabolized statin together with diltiazem; otherwise, dose adjustments for both diltiazem and the statin should be considered along with close monitoring for signs and symptoms of any statin related adverse events.

In a healthy volunteer cross-over study (N = 10), coadministration of a single 20 mg dose of simvastatin at the end of a 14-day regimen with 120 mg BID diltiazem SR resulted in a 5-fold increase in mean simvastatin AUC vs.

simvastatin alone.

Subjects with increased average steady-state exposures of diltiazem showed a greater fold increase in simvastatin exposure.

Computer-based simulations showed that at a daily dose of 480 mg of diltiazem, an 8- to 9-fold mean increase in simvastatin AUC can be expected.

If coadministration of simvastatin with diltiazem is required, limit the daily doses of simvastatin to 10 mg and diltiazem to 240 mg.

In a ten-subject randomized, open-label, 4-way cross-over study, coadministration of diltiazem (120 mg BID diltiazem SR for 2 weeks) with a single 20 mg dose of lovastatin resulted in 3- to 4-fold increase in mean lovastatin AUC and C max vs.

lovastatin alone.

In the same study, there was no significant change in 20 mg single dose pravastatin AUC and C max during diltiazem coadministration.

Diltiazem plasma levels were not significantly affected by lovastatin or pravastatin.

OVERDOSAGE

The oral LD 50 s in mice and rats range from 415 to 740 mg/kg and from 560 to 810 mg/kg, respectively.

The intravenous LD 50 s in these species were 60 and 38 mg/kg, respectively.

The oral LD 50 in dogs is considered to be in excess of 50 mg/kg, while lethality was seen in monkeys at 360 mg/kg.

The toxic dose in man is not known.

Due to extensive metabolism, blood levels after a standard dose of diltiazem can vary over 10-fold, limiting the usefulness of blood levels in overdose cases.

There have been reports of diltiazem overdose in amounts ranging from < 1 g to 18 g.

Of cases with known outcome, most patients recovered and in cases with a fatal outcome, the majority involved multiple drug ingestion.

Events observed following diltiazem overdose included bradycardia, hypotension, heart block, and cardiac failure.

Most reports of overdose described some supportive medical measure and/or drug treatment.

Bradycardia frequently responded favorably to atropine, as did heart block, although cardiac pacing was also frequently utilized to treat heart block.

Fluids and vasopressors were used to maintain blood pressure, and in cases of cardiac failure, inotropic agents were administered.

In addition, some patients received treatment with ventilatory support, gastric lavage, activated charcoal, and/or intravenous calcium.

The effectiveness of intravenous calcium administration to reverse the pharmacological effects of diltiazem overdose has been inconsistent.

In a few reported cases, overdose with calcium channel blockers associated with hypotension and bradycardia that was initially refractory to atropine became more responsive to atropine after the patients received intravenous calcium.

In some cases intravenous calcium has been administered (1 g calcium chloride or 3 g calcium gluconate) over 5 minutes and repeated every 10 to 20 minutes as necessary.

Calcium gluconate has also been administered as a continuous infusion at a rate of 2 g per hour for 10 hours.

Infusions of calcium for 24 hours or more may be required.

Patients should be monitored for signs of hypercalcemia.

In the event of overdose or exaggerated response, appropriate supportive measures should be employed in addition to gastrointestinal decontamination.

Diltiazem does not appear to be removed by peritoneal or hemodialysis.

Limited data suggest that plasmapheresis or charcoal hemoperfusion may hasten diltiazem elimination following overdose.

Based on the known pharmacological effects of diltiazem and/or reported clinical experiences, the following measures may be considered: Bradycardia: Administer atropine (0.60 mg to 1 mg).

If there is no response to vagal blockade, administer isoproterenol cautiously.

High Degree AV Block : Treat as for bradycardia above.

Fixed high-degree AV block should be treated with cardiac pacing.

Cardiac Failure : Administer inotropic agents (isoproterenol, dopamine, or dobutamine) and diuretics.

Hypotension: Vasopressors (e.g., dopamine or norepinephrine).

Actual treatment and dosage should depend on the severity of the clinical situation and the judgment and experience of the treating physician.

DESCRIPTION

Diltiazem hydrochloride is a calcium ion cellular influx inhibitor (slow channel blocker or calcium antagonist).

Chemically, diltiazem hydrochloride is 1,5-Benzothiazepin-4(5H)one, 3-(acetyloxy)-5-[2-(dimethylamino)ethyl]-2,3-dihydro-2-(4-methoxyphenyl), monohydrochloride, (+)-cis-.

The structural formula is: C 22 H 26 N 2 O 4 S • HCl M.W.

450.99 Diltiazem hydrochloride, USP is a white to off-white crystalline powder with a bitter taste.

It is soluble in water, methanol, and chloroform.

Each tablet, for oral administration, contains 30 mg, 60 mg, 90 mg, or 120 mg diltiazem hydrochloride.

In addition, each tablet contains the following inactive ingredients: ethylcellulose, hypromellose, lactose monohydrate, magnesium stearate, maltodextrin, polyethylene glycol, and sodium lauryl sulfate.

Diltiazem Hydrochloride Tablets, USP 30 mg, 60 mg, 90 mg and 120 mg meet USP Dissolution Test 2 .

Structural Formula – Diltiazem HCL Tablets

HOW SUPPLIED

Diltiazem Hydrochloride Tablets, USP are available containing 30 mg, 60 mg, 90 mg or 120 mg of diltiazem hydrochloride, USP.

The 30 mg tablets are white film-coated, round, unscored, tablets debossed with M over 23 on one side of the tablet and blank on the other side.

They are available as follows: NDC 0615-3548-39 blisterpacks of 30 tablets NDC 0615-3548-31 blisterpacks of 31 tablets The 60 mg tablets are white film-coated, round, tablets debossed with M over 45 on one side of the tablet and scored on the other side.

They are available as follows: NDC 0615-3549-39 blisterpacks of 30 tablets The 90 mg tablets are white film-coated, capsule-shaped tablets debossed with M135 on one side of the tablet and scored on the other side.

They are available as follows: NDC 0615-3550-39 blisterpacks of 30 tablets The 120 mg tablets are white film-coated, capsule-shaped tablets debossed with M525 on one side of the tablet and scored on the other side.

They are available as follows: NDC 0615-3551-39 blisterpacks of 30 tablets Store at 20° to 25°C (68° to 77°F).

[See USP for Controlled Room Temperature.] Protect from light.

Dispense in a tight, light-resistant container as defined in the USP using a child-resistant closure.

GERIATRIC USE

Geriatric Use Clinical studies of diltiazem did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects.

Other reported clinical experience has not identified differences in responses between the elderly and younger patients.

In general, dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range, reflecting the greater frequency of decreased hepatic, renal, or cardiac function, and of concomitant disease or other drug therapy.

MECHANISM OF ACTION

Mechanisms of Action Although precise mechanisms of its antianginal actions are still being delineated, diltiazem is believed to act in the following ways: 1.

Angina Due to Coronary Artery Spasm Diltiazem has been shown to be a potent dilator of coronary arteries both epicardial and subendocardial.

Spontaneous and ergonovine-induced coronary artery spasm are inhibited by diltiazem.

2.

Exertional Angina Diltiazem has been shown to produce increases in exercise tolerance, probably due to its ability to reduce myocardial oxygen demand.

This is accomplished via reductions in heart rate and systemic blood pressure at submaximal and maximal exercise work loads.

In animal models, diltiazem interferes with the slow inward (depolarizing) current in excitable tissue.

It causes excitation-contraction uncoupling in various myocardial tissues without changes in the configuration of the action potential.

Diltiazem produces relaxation of coronary vascular smooth muscle and dilation of both large and small coronary arteries at drug levels which cause little or no negative inotropic effect.

The resultant increases in coronary blood flow (epicardial and subendocardial) occur in ischemic and nonischemic models and are accompanied by dose dependent decreases in systemic blood pressure and decreases in peripheral resistance.

INDICATIONS AND USAGE

Diltiazem hydrochloride tablets are indicated for the management of chronic stable angina and angina due to coronary artery spasm.

PEDIATRIC USE

Pediatric Use Safety and effectiveness in pediatric patients have not been established.

PREGNANCY

Pregnancy Teratogenic Effects Pregnancy Category C Reproduction studies have been conducted in mice, rats, and rabbits.

Administration of doses ranging from 5 to 10 times greater (on a mg/kg basis) than the daily recommended therapeutic dose has resulted in embryo and fetal lethality.

These doses, in some studies, have been reported to cause skeletal abnormalities.

In the perinatal/postnatal studies, there was some reduction in early individual pup weights and survival rates.

There was an increased incidence of stillbirths at doses of 20 times the human dose or greater.

There are no well controlled studies in pregnant women; therefore, use diltiazem in pregnant women only if the potential benefit justifies the potential risk to the fetus.

NUSRING MOTHERS

Nursing Mothers Diltiazem is excreted in human milk.

One report suggests that concentrations in breast milk may approximate serum levels.

If use of diltiazem is deemed essential, an alternative method of infant feeding should be instituted.

DOSAGE AND ADMINISTRATION

Exertional Angina Pectoris Due to Atherosclerotic Coronary Artery Disease or Angina Pectoris at Rest Due to Coronary Artery Spasm Dosage must be adjusted to each patient’s needs.

Starting with 30 mg 4 times daily, before meals and at bedtime, dosage should be increased gradually (given in divided doses 3 or 4 times daily) at one-to two-day intervals until optimum response is obtained.

Although individual patients may respond to any dosage level, the average optimum dosage range appears to be 180 to 360 mg/day.

There are no available data concerning dosage requirements in patients with impaired renal or hepatic function.

If the drug must be used in such patients, titration should be carried out with particular caution.

Concomitant Use with Other Cardiovascular Agents 1.

Sublingual NTG may be taken as required to abort acute anginal attacks during diltiazem therapy.

2.

Prophylactic Nitrate Therapy: Diltiazem may be safely coadministered with short- and long-acting nitrates, but there have been no controlled studies to evaluate the antianginal effectiveness of this combination.

3.

Beta-blockers (See WARNINGS and PRECAUTIONS .)