Glycopyrrolate 1 MG Oral Tablet

WARNINGS

In the presence of a high environmental temperature, heat prostration (fever and heat stroke due to decreased sweating) can occur with use of glycopyrrolate.

Diarrhea may be an early symptom of incomplete intestinal obstruction, especially in patients with ileostomyor colostomy .

In this instance treatment with this drug would be inappropriate and possibly harmful.

Glycopyrrolate may produce drowsiness and blurred vision.

In this event, 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 this drug.

Theoretically, with overdosage, a curare-like action may occur, i.e., neuro-muscular blockade leading to muscular weakness and possible paralysis.

The safety of this drug during pregnancy has not been established.

The use of any drug during pregnancy requires that potential benefits of the drug be weighed against possible hazards to mother and child.

Reproduction studies in rats revealed no teratogenic effects from glycopyrrolate; however, the potent anticholinergic action of this agent resulted in diminished rates of conception and of survival at weaning, in a dose-related manner.

Other studies in dogs suggest that this may be due to diminished seminal secretion which is evident at high doses of glycopyrrolate.

Information on possible adverse effects in the pregnant female is limited to uncontrolled data derived from marketing experience.

Such experience has revealed no reports of teratogenic or other fetus-damaging potential.

No controlled studies to establish the safety of the drug in pregnancy have been performed.

It is not known whether this drug is excreted in human milk.

As a general rule, nursing should not be undertaken while a patient is on a drug since many drugs are excreted in human milk.

Since there is no adequate experience in pediatric patients who have received this drug, safety and efficacy in pediatric patients have not been established.

DRUG INTERACTIONS

There are no known drug interactions.

OVERDOSAGE

The symptoms of overdosage of glycopyrrolate are peripheral in nature rather than central.

To guard against further absorption of the drug-use gastric lavage, cathartics and/or enemas.

To combat peripheral anticholinergic effects (residual mydriasis, dry mouth, etc.)-utilize a quaternary ammonium anticholinesterase, such as neostigmine methylsulfate.

To combat hypotension-use pressor amines (norepinephrine, metaraminol) i.v.; and supportive care.

To combat respiratory depression-administer oxygen; utilize a respiratory stimulant such as Dopram®* i.v.; artificial respiration.

DESCRIPTION

Glycopyrrolate tablets contain the synthetic anticholinergic glycopyrrolate.

Glycopyrrolate is a quaternary ammonium compound with the following chemical name: 3-[(cyclopentylhydroxyphenylacetyl)oxy]-1,1-dimethylpyrrolidinium bromide.

Its empirical formula is C 19 H 28 BrNO 3 , its molecular weight is 398.33, and its structural formula is: Each 1 mg tablet contains: Glycopyrrolate, USP 1mg Each 2 mg tablet contains: Glycopyrrolate, USP 2mg Inactive Ingredients: Dibasic Calcium Phosphate, Lactose, Magnesium Stearate, Povidone, Sodium Starch Glycolate MM1

HOW SUPPLIED

Glycopyrrolate tablets 1 mg are bisected, compressed white, round tablets debossed “K” above the bisect and “400” below the bisect on one side of the tablet, and plain on the other side.

Available in bottles of 100 (NDC 49884-065-01).

Glycopyrrolate tablets 2 mg are bisected, compressed white, round tablets debossed “K” above the bisect and “401” below the bisect on one side of the tablet, and plain on the other side.

Available in bottles of 100 (NDC 49884-066-01).

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

Dispense in tight container.

Rx only * Dopram ® is a registered trademark of Baxter Healthcare Corporation.

Manufactured by: Par Pharmaceutical Companies, Inc.

Spring Valley, NY 10977 Rev: 07/2010 OS065-01-1-02

INDICATIONS AND USAGE

INDICATIONS & USAGE For use as adjunctive therapy in the treatment of peptic ulcer.

DOSAGE AND ADMINISTRATION

DOSAGE & ADMINISTRATION The dosage of glycopyrrolate should be adjusted to the needs of the individual patient to assure symptomatic control with a minimum of adverse reactions.

The presently recommended maximum daily dosage of glycopyrrolate is 8 mg.

Glycopyrrolate Tablets 1 mg.

The recommended initial dosage of glycopyrrolate for adults is one tablet three times daily (in the morning, early afternoon, and at bedtime).

Some patients may require two tablets at bedtime to assure overnight control of symptoms.

For maintenance, a dosage of one tablet twice a day is frequently adequate.

Glycopyrrolate Tablets 2 mg.

The recommended dosage of glycopyrrolate for adults is one tablet two or three times daily at equally spaced intervals.

Glycopyrrolate tablets are not recommended for use in pediatric patients under the age of 12 years.

lamoTRIgine 200 MG 24HR Extended Release Oral Tablet

DRUG INTERACTIONS

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

Additional details of these drug interaction studies, which were conducted using immediate-release lamotrigine, are provided in the Clinical Pharmacology section [ see Clinical Pharmacology (12.3) ].

Table 5.

Established and Other Potentially Significant Drug Interactions ↓ = Decreased (induces lamotrigine glucuronidation).

↑ = Increased (inhibits lamotrigine glucuronidation).

? = Conflicting data.

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.

Effect of Lamotrigine Extended-Release Tablets 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 extended-release tablets with OCT2 substrates with a narrow therapeutic index (e.g., dofetilide) is not recommended.

OVERDOSAGE

10 10.1 Human Overdose Experience Overdoses involving quantities up to 15 g have been reported for immediate-release 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 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 extended-release tablets.

DESCRIPTION

11 Lamotrigine, 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 is a white or almost white powder and has a pK a of 5.7.

Lamotrigine is very slightly soluble in water and in 0.1M hydrochloric acid.

The structural formula is: Lamotrigine extended-release tablets are supplied for oral administration as 25 mg (yellow, enteric-coated, circular shaped tablet), 50 mg (pink, enteric-coated, circular shaped tablet), 100 mg (light brown, enteric-coated, circular shaped tablet), 200 mg (pink, enteric-coated, circular shaped tablet), and 300 mg (pink, enteric-coated, circular shaped tablet).

Each tablet contains the labeled amount of lamotrigine and the following inactive ingredients: diethyl phthalate, hypromellose, lactose monohydrate, magnesium stearate, methacrylic acid copolymer, polyethylene glycol, talc, titanium dioxide, iron oxide yellow for (25 mg and 100 mg) and iron oxide red for (50 mg, 100 mg, 200 mg, and 300 mg).

Lamotrigine extended-release tablets contain a modified-release eroding formulation as the core.

The tablets are coated with enteric coat to enable a controlled release of drug in the acidic environment of the stomach.

The combination of this and the modified-release core are designed to control the dissolution rate of lamotrigine over a period of approximately 12 to 15 hours, leading to a gradual increase in serum lamotrigine levels.

Structure

CLINICAL STUDIES

14 14.1 Adjunctive Therapy for Primary Generalized Tonic-Clonic Seizures The effectiveness of lamotrigine extended-release tablets as adjunctive therapy in subjects with PGTC seizures was established in a 19-week, international, multicenter, double-blind, randomized, placebo-controlled trial in 143 patients aged 13 years and older (n = 70 on lamotrigine extended-release tablets, n = 73 on placebo).

Patients with at least 3 PGTC seizures during an 8-week baseline phase were randomized to 19 weeks of treatment with lamotrigine extended-release tablets 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 200 to 500 mg/day of lamotrigine extended-release tablets based on concomitant AEDs (target dose = 200 mg for valproate, 300 mg for AEDs not altering plasma lamotrigine levels, and 500 mg for enzyme-inducing AEDs).

The primary efficacy endpoint was percent change from baseline in PGTC seizure frequency during the double-blind treatment phase.

For the intent-to-treat population, the median percent reduction in PGTC seizure frequency was 75% in patients treated with lamotrigine extended-release tablets and 32% in patients treated with placebo, a difference that was statistically significant, defined as a 2-sided P value ≤0.05.

Figure 1 presents the percentage of patients (X-axis) with a percent reduction in PGTC seizure frequency (responder rate) from baseline through the entire treatment period at least as great as that represented on the Y-axis.

A positive value on the Y-axis indicates an improvement from baseline (i.e., a decrease in seizure frequency), while a negative value indicates a worsening from baseline (i.e., an increase in seizure frequency).

Thus, in a display of this type, a curve for an effective treatment is shifted to the left of the curve for placebo.

The proportion of patients achieving any particular level of reduction in PGTC seizure frequency was consistently higher for the group treated with lamotrigine extended-release tablets compared with the placebo group.

For example, 70% of patients randomized to lamotrigine extended-release tablets experienced a 50% or greater reduction in PGTC seizure frequency, compared with 32% of patients randomized to placebo.

Patients with an increase in seizure frequency >100% are represented on the Y-axis as equal to or greater than -100%.

Figure 1.

Proportion of Patients by Responder Rate for Lamotrigine Extended-Release Tablets and Placebo Group (Primary Generalized Tonic-Clonic Seizures Study) Figure 1 14.2 Adjunctive Therapy for Partial-Onset Seizures The effectiveness of immediate-release lamotrigine as adjunctive therapy was initially established in 3 pivotal, multicenter, placebo-controlled, double-blind clinical trials in 355 adults with refractory partial-onset seizures.

The effectiveness of lamotrigine extended-release tablets as adjunctive therapy in partial-onset seizures, with or without secondary generalization, was established in a 19-week, multicenter, double-blind, placebo-controlled trial in 236 patients aged 13 years and older (approximately 93% of patients were aged 16 to 65 years).

Approximately 36% were from the U.S.

and approximately 64% were from other countries including Argentina, Brazil, Chile, Germany, India, Korea, Russian Federation, and Ukraine.

Patients with at least 8 partial-onset seizures during an 8-week prospective baseline phase (or 4-week prospective baseline coupled with a 4-week historical baseline documented with seizure diary data) were randomized to treatment with lamotrigine extended-release tablets (n = 116) or placebo (n = 120) added to their current regimen of 1 or 2 AEDs.

Approximately half of the patients were taking 2 concomitant AEDs at baseline.

Target doses ranged from 200 to 500 mg/day of lamotrigine extended-release tablets based on concomitant AED (target dose = 200 mg for valproate, 300 mg for AEDs not altering plasma lamotrigine, and 500 mg for enzyme-inducing AEDs).

The median partial seizure frequency per week at baseline was 2.3 for lamotrigine extended-release tablets and 2.1 for placebo.

The primary endpoint was the median percent change from baseline in partial-onset seizure frequency during the entire double-blind treatment phase.

The median percent reductions in weekly partial-onset seizures were 47% in patients treated with lamotrigine extended-release tablets and 25% on placebo, a difference that was statistically significant, defined as a 2-sided P value ≤0.05.

Figure 2 presents the percentage of patients (X-axis) with a percent reduction in partial-onset seizure frequency (responder rate) from baseline through the entire treatment period at least as great as that represented on the Y-axis.

The proportion of patients achieving any particular level of reduction in partial-onset seizure frequency was consistently higher for the group treated with lamotrigine extended-release tablets compared with the placebo group.

For example, 44% of patients randomized to lamotrigine extended-release tablets experienced a 50% or greater reduction in partial-onset seizure frequency compared with 21% of patients randomized to placebo.

Figure 2.

Proportion of Patients by Responder Rate for Lamotrigine Extended-Release Tablets and Placebo Group (Partial-Onset Seizure Study) Figure 2 14.3 Conversion to Monotherapy for Partial-Onset Seizures The effectiveness of lamotrigine extended-release tablets as monotherapy for partial-onset seizures was established in a historical control trial in 223 adults with partial-onset seizures.

The historical control methodology is described in a publication by French, et al.

[ see References (15) ].

Briefly, in this study, patients were randomized to ultimately receive either lamotrigine extended-release tablets 300 or 250 mg once a day, and their responses were compared with those of a historical control group.

The historical control consisted of a pooled analysis of the control groups from 8 studies of similar design, which utilized a subtherapeutic dose of an AED as a comparator.

Statistical superiority to the historical control was considered to be demonstrated if the upper 95% confidence interval for the proportion of patients meeting escape criteria in patients receiving lamotrigine extended-release tablets remained below the lower 95% prediction interval of 65.3% derived from the historical control data.

In this study, patients aged 13 years and older experienced at least 4 partial-onset seizures during an 8-week baseline period with at least 1 seizure occurring during each of 2 consecutive 4-week periods while receiving valproate or a non-enzyme-inducing AED.

Lamotrigine extended-release tablet was added to either valproate or a non-enzyme-inducing AED over a 6- to 7-week period followed by the gradual withdrawal of the background AED.

Patients were then continued on monotherapy with lamotrigine extended-release tablets for 12 weeks.

The escape criteria were 1 or more of the following: (1) doubling of average monthly seizure count during any 28 consecutive days, (2) doubling of highest consecutive 2-day seizure frequency during the entire treatment phase, (3) emergence of a new seizure type compared with baseline (4) clinically significant prolongation of generalized tonic-clonic seizures or worsening of seizure considered by the investigator to require intervention.

These criteria were similar to those in the 8 controlled trials from which the historical control group was constituted.

The upper 95% confidence limits of the proportion of subjects meeting escape criteria (40.2% at 300 mg/day and 44.5% at 250 mg/day) were below the threshold of 65.3% derived from the historical control data.

Although the study population was not fully comparable with the historical controlled population and the study was not fully blinded, numerous sensitivity analyses supported the primary results.

Efficacy was further supported by the established effectiveness of the immediate-release formulation as monotherapy.

HOW SUPPLIED

16 /STORAGE AND HANDLING Lamotrigine extended-release tablets 25 mg are available as yellow, enteric-coated, circular shaped tablets, debossed with “W” on one side and “271” on the other side.

NDC 64679-271-01, bottle of 30 tablets NDC 64679-271-02, bottle of 500 tablets NDC 64679-271-04, unit dose of 100 tablets 50 mg are available as pink, enteric-coated, circular shaped tablets, debossed with “W” on one side and “274” on the other side.

NDC 64679-274-01, bottle of 30 tablets NDC 64679-274-02, bottle of 500 tablets NDC 64679-274-03, unit dose of 100 tablets 100 mg are available as light brown, enteric-coated, circular shaped tablets, debossed with “W” on one side and “273” on the other side.

NDC 64679-273-01, bottle of 30 tablets NDC 64679-273-02, bottle of 500 tablets NDC 64679-273-04, unit dose of 100 tablets 200 mg are available as pink, enteric-coated, circular shaped tablets, debossed with “W” on one side and “272” on the other side.

NDC 64679-272-01, bottle of 30 tablets NDC 64679-272-02, bottle of 500 tablets NDC 64679-272-04, unit dose of 100 tablets 300 mg are available as pink, enteric-coated, circular shaped tablets, debossed with “W” on one side and “275” on the other side.

NDC 64679-275-01, bottle of 30 tablets NDC 64679-275-02, bottle of 500 tablets NDC 64679-275-04, unit dose of 100 tablets Storage Store at 20°-25°C (68°-77°F); [see USP Controlled Room Temperature].

RECENT MAJOR CHANGES

Dosage and Administration ( 2.1 , 2.2 ) 12/2014 Warnings and Precautions, Laboratory Tests ( 5.13 ) 3/2015

GERIATRIC USE

8.5 Geriatric Use Clinical trials of lamotrigine extended-release tablets for epilepsy 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 Extended-release tablets: 25 mg, 50 mg, 100 mg, 200 mg, and 300 mg.

( 3.1 , 16 ) 3.1 Extended-Release Tablets 25 mg, yellow, enteric-coated, circular shaped tablet, debossed with “W” on one side and “271” on the other side.

50 mg, pink, enteric-coated, circular shaped tablet, debossed with “W” on one side and “274” on the other side.

100 mg, light brown, enteric-coated, circular shaped tablet, debossed with “W” on one side and “273” on the other side.

200 mg, pink, enteric-coated, circular shaped tablet, debossed with “W” on one side and “272” on the other side.

300 mg, pink, enteric-coated, circular shaped tablet, debossed with “W” on one side and “275” 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 a 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 microM of glycine) in cultured hippocampal neurons exceeded 100 microM.

INDICATIONS AND USAGE

1 Lamotrigine extended-release tablet is an antiepileptic drug (AED) indicated for: Adjunctive therapy for primary generalized tonic-clonic seizures and partial-onset seizures with or without secondary generalization in patients aged 13 years and older.

( 1.1 ) Conversion to monotherapy in patients aged 13 years and older with partial-onset seizures who are receiving treatment with a single AED.

( 1.2 ) Limitation of use: Safety and effectiveness in patients younger than 13 years have not been established.

( 1.3 ) 1.1 Adjunctive Therapy Lamotrigine extended-release tablets are indicated as adjunctive therapy for primary generalized tonic-clonic (PGTC) seizures and partial-onset seizures with or without secondary generalization in patients aged 13 years and older.

1.2 Monotherapy Lamotrigine extended-release tablets are is indicated for conversion to monotherapy in patients aged 13 years and older with partial-onset seizures who are receiving treatment with a single antiepileptic drug (AED).

Safety and effectiveness of lamotrigine extended-release tablets have not been established (1) as initial monotherapy or (2) for simultaneous conversion to monotherapy from 2 or more concomitant AEDs.

1.3 Limitation of Use Safety and effectiveness of lamotrigine extended-release tablets for use in patients younger than 13 years have not been established.

PEDIATRIC USE

8.4 Pediatric Use Lamotrigine extended-release tablet is indicated as adjunctive therapy for PGTC and partial-onset seizures with or without secondary generalization in patients aged 13 years and older.

Safety and effectiveness of lamotrigine extended-release tablets for any use in patients younger than 13 years have not been established.

Immediate-release 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 immediate-release 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).

Immediate-release 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.

In a juvenile animal study in which lamotrigine (oral doses of 5, 15, or 30 mg/kg) was administered to young rats (postnatal days 7 to 62), decreased viability and growth were seen at the highest dose tested and long-term behavioral 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 effects on neurobehavioral development is less than the human dose of 400 mg/day on a mg/m 2 basis.

PREGNANCY

8.1 Pregnancy 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-partum concentrations after delivery.

Dosage adjustments may be necessary to maintain clinical response.

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

In animal studies, lamotrigine was developmentally toxic at doses lower than those administered clinically.

Lamotrigine extended-release tablets should be used during pregnancy only if the potential benefit justifies the potential risk to the fetus.

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 5 or 25 mg/kg) during the period of organogenesis and offspring were evaluated postnatally, behavioral 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 5, 10, or 20 mg/kg) during the latter part of gestation, increased offspring mortality (including stillbirths) was seen at all doses.

The lowest effect dose for peri/postnatal 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.

Lamotrigine decreases fetal folate concentrations in rat, an effect known to be associated with adverse pregnancy outcomes in animals and humans.

Pregnancy Registry To provide information regarding the effects of in utero exposure to lamotrigine extended-release tablets, physicians are advised to recommend that pregnant patients taking lamotrigine extended-release tablets enroll in the North American Antiepileptic Drug (NAAED) Pregnancy Registry.

This can be done by calling the toll-free number 1-888-233-2334 and must be done by patients themselves.

Information on the registry can also be found at the website http://www.aedpregnancyregistry.org.

NUSRING MOTHERS

8.3 Nursing Mothers Lamotrigine is present in milk from lactating women taking lamotrigine extended-release tablets.

Data from multiple small studies indicate that lamotrigine plasma levels in human milk-fed infants have been reported to be as high as 50% of the maternal serum levels.

Neonates and young infants are at risk for high serum levels because maternal serum and milk levels can rise to high levels postpartum if lamotrigine dosage has been increased during pregnancy but not later reduced to the pre-pregnancy dosage.

Lamotrigine exposure is further increased due to the immaturity of the infant glucuronidation capacity needed for drug clearance.

Events including apnea, drowsiness, and poor sucking have been reported in infants who have been human milk-fed by mothers using lamotrigine; whether or not these events were caused by lamotrigine is unknown.

Human milk-fed infants should be closely monitored for adverse events resulting from lamotrigine.

Measurement of infant serum levels should be performed to rule out toxicity if concerns arise.

Human milk-feeding should be discontinued in infants with lamotrigine toxicity.

Caution should be exercised when lamotrigine extended-release tablets are administered to a nursing woman.

BOXED WARNING

WARNING: SERIOUS SKIN RASHES 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 extended-release tablets.

exceeding recommended dose escalation for lamotrigine extended-release tablets.

( 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 extended-release tablets should be discontinued at the first sign of rash, unless the rash is clearly not drug related.

( 5.1 ) Lamotrigine extended-release tablets can cause serious rashes requiring hospitalization and discontinuation of treatment.

The incidence of these rashes, which have included Stevens-Johnson syndrome, is approximately 0.8% (8 per 1,000) in pediatric patients (aged 2 to 16 years) receiving immediate-release lamotrigine as adjunctive therapy for epilepsy and 0.3% (3 per 1,000) in adults on adjunctive therapy for epilepsy.

In a prospectively followed cohort of 1,983 pediatric patients (aged 2 to 16 years) with epilepsy taking adjunctive immediate-release lamotrigine, there was 1 rash-related death.

Lamotrigine extended-release tablet is not approved for patients younger than 13 years.

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.

The risk of serious rash caused by treatment with lamotrigine extended-release tablet is not expected to differ from that with immediate-release lamotrigine.

However, the relatively limited treatment experience with lamotrigine extended-release tablets makes it difficult to characterize the frequency and risk of serious rashes caused by treatment with lamotrigine extended-release tablets.

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 extended-release tablets.

There are suggestions, yet to be proven, that the risk of rash may also be increased by (1) coadministration of lamotrigine extended-release tablets with valproate (includes valproic acid and divalproex sodium), (2) exceeding the recommended initial dose of lamotrigine extended-release tablets, or (3) exceeding the recommended dose escalation for lamotrigine extended-release tablets.

However, cases have occurred in the absence of these factors.

Nearly all cases of life-threatening rashes caused by immediate-release 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 extended-release tablets, it is not possible to predict reliably which rashes will prove to be serious or life threatening.

Accordingly, lamotrigine extended-release tablets 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 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 ) Fatal or life-threatening hypersensitivity reaction: Multiorgan hypersensitivity reactions, also known as drug reaction with eosinophilia and systemic symptoms (DRESS), 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 extended-release tablets should be discontinued if alternate etiology for this reaction is not found.

( 5.2 ) 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.

( 5.3 ) Suicidal behavior and ideation: Monitor for suicidal thoughts or behaviors.

( 5.4 ) Aseptic meningitis: Monitor for signs of meningitis.

( 5.5 ) Medication errors due to product name confusion: Strongly advise patients to visually inspect tablets to verify the received drug is correct.

( 5.6 , 16 , 17 ) 5.1 Serious Skin Rashes [see Boxed Warning] The risk of serious rash caused by treatment with lamotrigine extended-release tablet is not expected to differ from that with immediate-release lamotrigine [ see Boxed Warning ].

However, the relatively limited treatment experience with lamotrigine extended-release tablets makes it difficult to characterize the frequency and risk of serious rashes caused by treatment with lamotrigine extended-release tablets.

Pediatric Population The incidence of serious rash associated with hospitalization and discontinuation of immediate-release lamotrigine in a prospectively followed cohort of pediatric patients (aged 2 to 16 years) with epilepsy receiving adjunctive therapy with immediate-release lamotrigine was approximately 0.8% (16 of 1,983).

When 14 of these cases were reviewed by 3 expert dermatologists, there was considerable disagreement as to their proper classification.

To illustrate, one dermatologist considered none of the cases to be Stevens-Johnson syndrome; another assigned 7 of the 14 to this diagnosis.

There was 1 rash-related death in this 1,983-patient cohort.

Additionally, there have been rare cases of toxic epidermal necrolysis with and without permanent sequelae and/or death in US 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, 1.2% (6 of 482) experienced a serious rash compared with 0.6% (6 of 952) patients not taking valproate.

Lamotrigine extended-release tablet is not approved in patients younger than 13 years.

Adult Population Serious rash associated with hospitalization and discontinuation of immediate-release lamotrigine occurred in 0.3% (11 of 3,348) of adult patients who received immediate-release lamotrigine in premarketing clinical trials of epilepsy.

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 multiorgan hypersensitivity [ see Warnings and Precautions (5.2) ].

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 immediate-release 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 immediate-release 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 extended-release tablets are exceeded and in patients with a history of allergy or rash to other AEDs.

5.2 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 extended-release tablets should be discontinued if an alternative etiology for the signs or symptoms cannot be established.

Prior to initiation of treatment with lamotrigine extended-release tablets, 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 physician immediately.

5.3 Blood Dyscrasias There have been reports of blood dyscrasias with immediate-release lamotrigine that may or may not be associated with multiorgan hypersensitivity (also known as DRESS) [ see Warnings and Precautions (5.2) ].

These have included neutropenia, leukopenia, anemia, thrombocytopenia, pancytopenia, and, rarely, aplastic anemia and pure red cell aplasia.

5.4 Suicidal Behavior and Ideation AEDs, including lamotrigine extended-release tablets, 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 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 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 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 3.4 3.5 2.4 Psychiatric 5.7 8.5 1.5 2.9 Other 1 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 extended-release tablets 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.

5.5 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 (5.2) ].

5.6 Potential Medication Errors Medication errors involving lamotrigine have occurred.

In particular, the names 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 extended-release tablets clearly.

Depictions of the lamotrigine extended-release tablets can be found in the Medication Guide.

Each lamotrigine extended-release tablet has a distinct debossing and is debossed with “W” on one side and “271” on the other side for 25 mg, “W” on one side and “274” on the other side for 50 mg, “W” on one side and “273” on the other side for 100 mg, “W” on one side and “272” on the other side for 200 mg, and “W” on one side and “275” on the other side for 300 mg.

These distinctive features serve to identify the different presentations of the drug and thus may help reduce the risk of medication errors.

Lamotrigine extended-release tablets are supplied in circular, unit-of-use bottles containing 30 tablets and 100 unit dose blister pack.

Additionally, Lamotrigine extended-release tablets are also supplied as 500 count bottle pack.

The label on the bottle includes a depiction of the tablets that further communicates to patients and pharmacists that the medication is lamotrigine extended-release tablets and the specific tablet strength included in the bottle.

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 extended-release tablets each time they fill their prescription.

5.7 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 extended-release tablets [ 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.

5.8 Withdrawal Seizures As with other AEDs, lamotrigine extended-release tablets should not be abruptly discontinued.

In patients with epilepsy there is a possibility of increasing seizure frequency.

Unless safety concerns require a more rapid withdrawal, the dose of lamotrigine extended-release tablets should be tapered over a period of at least 2 weeks (approximately 50% reduction per week) [ see Dosage and Administration (2.1) ].

5.9 Status Epilepticus Valid estimates of the incidence of treatment-emergent status epilepticus among patients treated with immediate-release 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.

5.10 Sudden Unexplained Death in Epilepsy (SUDEP) During the premarketing development of immediate-release 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 immediate-release 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 immediate-release lamotrigine and the accuracy of the estimates provided.

Probably most reassuring is the similarity of estimated SUDEP rates in patients receiving immediate-release lamotrigine and those receiving other AEDs, chemically unrelated to each other, that underwent clinical testing in similar populations.

Importantly, that drug is chemically unrelated to lamotrigine.

This evidence suggests, although it certainly does not prove, that the high SUDEP rates reflect population rates, not a drug effect.

5.11 Addition of Lamotrigine Extended-Release Tablets 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 ( 2.1 , 2.2 ), Drug Interactions (7) ].

5.12 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.

Accordingly, although there are no specific recommendations for periodic ophthalmological monitoring, prescribers should be aware of the possibility of long-term ophthalmologic effects.

5.13 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 extended-release tablets has not been established.

Because of the possible pharmacokinetic interactions between lamotrigine and other drugs, including AEDs (see Table 6), 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.

Effect on Leukocytes Treatment with lamotrigine extended-release tablets caused an increased incidence of subnormal (below the reference range) values in some hematology analytes (e.g., total white blood cells, monocytes).

The treatment effect (Lamotrigine extended-release tablets % – Placebo %) incidence of subnormal counts was 3% for total white blood cells and 4% for monocytes.

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 extended-release tablets, 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 physician 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 physician immediately if they experience any signs or symptoms of these conditions [ see Warnings and Precautions ( 5.2 , 5.3 ) ].

Suicidal Thinking and Behavior Inform patients, their caregivers, and families that AEDs, including lamotrigine extended-release tablets, 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 physician.

Worsening of Seizures Advise patients to notify their physician if worsening of seizure control occurs.

Central Nervous System Adverse Effects Inform patients that lamotrigine extended-release tablets 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 extended-release tablets to gauge whether or not it adversely affects their mental and/or motor performance.

Pregnancy and Nursing Instruct patients to notify their physician 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 extended-release tablet 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 physician 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 (5.7) , Clinical Pharmacology (12.3) ].

Also instruct women to promptly notify their physician if they experience adverse reactions or changes in menstrual pattern (e.g., break-through bleeding) while receiving lamotrigine extended-release tablets in combination with these medications.

Discontinuing Lamotrigine Extended-Release Tablets Instruct patients to notify their physician if they stop taking lamotrigine extended-release tablets for any reason and not to resume lamotrigine extended-release tablets without consulting their physician.

Aseptic Meningitis Inform patients that lamotrigine extended-release tablets may cause aseptic meningitis.

Instruct them to notify their physician 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 extended-release tablets.

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 extended-release tablets each time they fill their prescription [ see Dosage Forms and Strengths (3) , How Supplied/Storage and Handling (16) ].

Refer the patient to the Medication Guide that provides depictions of the lamotrigine extended-release tablets extended-release tablets.

Manufactured by: Wockhardt Limited H-14/2, M.I.D.C.

Area, Waluj, Aurangabad, Maharashtra, India.

Distributed by: Wockhardt USA LLC.

20 Waterview Blvd.

Parsippany, NJ 07054 USA.

Rev.020715

DOSAGE AND ADMINISTRATION

2 Do not exceed the recommended initial dosage and subsequent dose escalation.

( 2.1 ) Initiation of adjunctive therapy and conversion to monotherapy requires slow titration dependent on concomitant AEDs; the prescriber must refer to the appropriate algorithm in Dosage and Administration.

( 2.2 , 2.3 ) Adjunctive therapy: Target therapeutic dosage range is 200 to 600 mg daily and is dependent on concomitant AEDs.

( 2.2 ) Conversion to monotherapy: Target therapeutic dosage range is 250 to 300 mg daily.

( 2.3 ) Conversion from immediate-release lamotrigine to lamotrigine extended-release tablets: The initial dose of lamotrigine extended-release tablets should match the total daily dose of the immediate-release lamotrigine.

Patients should be closely monitored for seizure control after conversion.

( 2.4 ) Do not restart lamotrigine extended-release 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 , 5.7 ) Discontinuation: Taper over a period of at least 2 weeks (approximately 50% dose reduction per week).

( 2.1 , 5.8 ) Lamotrigine extended-release tablets are taken once daily, with or without food.

Tablets must be swallowed whole and must not be chewed, crushed, or divided.

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 extended-release tablets with valproate, (2) exceeding the recommended initial dose of lamotrigine extended-release tablets, or (3) exceeding the recommended dose escalation for lamotrigine extended-release tablets.

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 extended-release tablet is exceeded and in patients with a history of allergy or rash to other AEDs.

It is recommended that lamotrigine extended-release 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 extended-release 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 Extended-Release Tablets Added to Drugs Known to Induce or Inhibit Glucuronidation Because lamotrigine is 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 extended-release tablets in patients on estrogen-containing contraceptives and atazanavir/ritonavir, see below and Table 5.

For dosing considerations for lamotrigine extended-release tablets in patients on other drugs known to induce or inhibit glucuronidation, see Table 1 and Table 5.

Target Plasma Levels A therapeutic plasma concentration range has not been established for lamotrigine.

Dosing of lamotrigine extended-release tablets should be based on therapeutic response [ see Clinical Pharmacology (12.3) ].

Women Taking Estrogen-Containing Oral Contraceptives Starting lamotrigine extended-release 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 extended-release 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 extended-release tablets based on the concomitant AED or other concomitant medications (see Table 1).

See below for adjustments to maintenance doses of lamotrigine extended-release tablets in women taking estrogen-containing oral contraceptives.

Adjustments to the Maintenance Dose of lamotrigine extended-release tablets 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 extended-release 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 extended-release 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 Table 1) 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 extended-release 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 extended-release 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 extended-release 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 extended-release 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 extended-release 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 extended-release 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 extended-release 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 extended-release 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 extended-release 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 extended-release tablets based on concomitant AED or other concomitant medications (see Tables 1 and 5).

In patients already taking maintenance doses of lamotrigine extended-release tablets and not taking glucuronidation inducers, the dose of lamotrigine extended-release 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 extended-release tablets should be based on patients’ concomitant medications (see Table 1); 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 immediate-release lamotrigine.

Because there is inadequate experience in this population, lamotrigine extended-release tablets should be used with caution in these patients.

Discontinuation Strategy For patients receiving lamotrigine extended-release 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 extended-release 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 (5.8) ].

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.

2.2 Adjunctive Therapy for Primary Generalized Tonic-Clonic and Partial-Onset Seizures This section provides specific dosing recommendations for patients aged 13 years and older.

Specific dosing recommendations are provided depending upon concomitant AEDs or other concomitant medications.

Table 1.

Escalation Regimen for Lamotrigine Extended-Release Tablets in Patients Aged 13 Years and Older 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) , and Clinical Pharmacology (12.3) ].

c Dose increases at week 8 or later should not exceed 100 mg daily at weekly intervals.

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 every day Weeks 3 and 4 25 mg every day 50 mg every day 100 mg every day Week 5 50 mg every day 100 mg every day 200 mg every day Week 6 100 mg every day 150 mg every day 300 mg every day Week 7 150 mg every day 200 mg every day 400 mg every day Maintenance range (week 8 and onward) 200 to 250 mg every day c 300 to 400 mg every day c 400 to 600 mg every day c 2.3 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 extended-release tablets.

To avoid an increased risk of rash, the recommended maintenance dosage range of lamotrigine extended-release tablets as monotherapy is 250 to 300 mg given once daily.

The recommended initial dose and subsequent dose escalations for lamotrigine extended-release tablets should not be exceeded [ see Boxed Warning ].

Conversion From Adjunctive Therapy With Carbamazepine, Phenytoin, Phenobarbital, or Primidone to Monotherapy With Lamotrigine Extended-Release Tablets After achieving a dose of 500 mg/day of lamotrigine extended-release 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.

Two weeks after completion of withdrawal of the enzyme-inducing AED, the dosage of lamotrigine extended-release tablets may be decreased no faster than 100 mg/day each week to achieve the monotherapy maintenance dosage range of 250 to 300 mg/day.

The regimen for the withdrawal of the concomitant AED is based on experience gained in the controlled monotherapy clinical trial using immediate-release lamotrigine.

Conversion From Adjunctive Therapy With Valproate to Monotherapy With Lamotrigine Extended-Release Tablets The conversion regimen involves the 4 steps outlined in Table 2.

Table 2.

Conversion From Adjunctive Therapy With Valproate to Monotherapy With Lamotrigine Extended-Release Tablets in Patients Aged 13 Years and Older With Epilepsy Lamotrigine Extended – Release Tablets Valproate Step 1 Achieve a dose of 150 mg/day according to guidelines in Table 1.

Maintain established stable dose.

Step 2 Maintain at 150 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 200 mg/day.

Simultaneously decrease to 250 mg/day and maintain for 1 week.

Step 4 Increase to 250 or 300 mg/day.

Discontinue.

Conversion From Adjunctive Therapy With Antiepileptic Drugs Other Than Carbamazepine, Phenytoin, Phenobarbital, Primidone, or Valproate to Monotherapy With Lamotrigine Extended-Release Tablets After achieving a dosage of 250 to 300 mg/day of lamotrigine extended-release tablets using the guidelines in Table 1, the concomitant AED should be withdrawn by 20% decrements each week over a 4-week period.

No adjustment to the monotherapy dose of lamotrigine extended-release tablets is needed.

2.4 Conversion From Immediate-Release Lamotrigine Tablets to Lamotrigine Extended-Release Tablets Patients may be converted directly from immediate-release lamotrigine to lamotrigine extended-release tablets.

The initial dose of lamotrigine extended-release tablets should match the total daily dose of immediate-release lamotrigine.

However, some subjects on concomitant enzyme-inducing agents may have lower plasma levels of lamotrigine on conversion and should be monitored [ see Clinical Pharmacology (12.3) ].

Following conversion to lamotrigine extended-release tablets, all patients (but especially those on drugs that induce lamotrigine glucuronidation) should be closely monitored for seizure control [ see Drug Interactions (7) ].

Depending on the therapeutic response after conversion, the total daily dose may need to be adjusted within the recommended dosing instructions (Table 1).

brimonidine tartrate 0.2 % Ophthalmic Solution

DRUG INTERACTIONS

7 Antihypertensives/cardiac glycosides may lower blood pressure.

( 7.1 ) Use with CNS depressants may result in an additive or potentiating effect.

( 7.2 ) Tricyclic antidepressants may potentially blunt the hypotensive effect of systemic clonidine.

( 7.3 ) Monoamine oxidase inhibitors may result in increased hypotension.

( 7.4 ) 7.1 Antihypertensives/Cardiac Glycosides Because brimonidine tartrate ophthalmic solution may reduce blood pressure, caution in using drugs such as antihypertensives and/or cardiac glycosides with brimonidine tartrate ophthalmic solution is advised.

7.2 CNS Depressants Although specific drug interaction studies have not been conducted with brimonidine tartrate ophthalmic solution, the possibility of an additive or potentiating effect with CNS depressants (alcohol, barbiturates, opiates, sedatives, or anesthetics) should be considered.

7.3 Tricyclic Antidepressants Tricyclic antidepressants have been reported to blunt the hypotensive effect of systemic clonidine.

It is not known whether the concurrent use of these agents with brimonidine tartrate ophthalmic solution in humans can lead to resulting interference with the IOP lowering effect.

Caution is advised in patients taking tricyclic antidepressants which can affect the metabolism and uptake of circulating amines.

7.4 Monoamine Oxidase Inhibitors Monoamine oxidase (MAO) inhibitors may theoretically interfere with the metabolism of brimonidine and potentially result in an increased systemic side-effect such as hypotension.

Caution is advised in patients taking MAO inhibitors which can affect the metabolism and uptake of circulating amines.

OVERDOSAGE

10 Very limited information exists on accidental ingestion of brimonidine in adults; the only adverse reaction reported to date has been hypotension.

Symptoms of brimonidine overdose have been reported in neonates, infants, and children receiving brimonidine tartrate as part of medical treatment of congenital glaucoma or by accidental oral ingestion [ see Use In Specific Populations ( 8.4 ) ].

Treatment of an oral overdose includes supportive and symptomatic therapy; a patent airway should be maintained.

DESCRIPTION

11 Brimonidine tartrate ophthalmic solution 0.2%, sterile, is a relatively selective alpha-2 adrenergic receptor agonist (topical intraocular pressure lowering agent).

The structural formula of brimonidine tartrate is: 5-Bromo-6-(2-imidazolidinylideneamino) quinoxaline L-tartrate; MW= 442.24 In solution, brimonidine tartrate ophthalmic solution 0.2% has a clear, greenish-yellow color.

It has an osmolality of 280 to 330 mOsml/kg and a pH of 5.6 to 6.6.

Each mL of brimonidine tartrate ophthalmic solution contains the active ingredient brimonidine tartrate 0.2% (2 mg/mL) with the inactive ingredients benzalkonium chloride 0.005% (0.05 mg/mL) as a preservative; citric acid; polyvinyl alcohol; sodium chloride; sodium citrate; and water for injection.

Hydrochloric acid and/or sodium hydroxide may be added to adjust pH.

Structural Formula

CLINICAL STUDIES

14 Elevated IOP presents a major risk factor in glaucomatous field loss.

The higher the level of IOP, the greater the likelihood of optic nerve damage and visual field loss.

Brimonidine tartrate has the action of lowering intraocular pressure with minimal effect on cardiovascular and pulmonary parameters.

In comparative clinical studies with timolol 0.5%, lasting up to one year, the IOP lowering effect of brimonidine tartrate ophthalmic solution was approximately 4 to 6 mm Hg compared with approximately 6 mm Hg for timolol.

In these studies, both patient groups were dosed BID; however, due to the duration of action of brimonidine tartrate ophthalmic solution, it is recommended that brimonidine tartrate ophthalmic solution be dosed TID.

Eight percent of subjects were discontinued from studies due to inadequately controlled intraocular pressure, which in 30% of these patients occurred during the first month of therapy.

Approximately 20% were discontinued due to adverse experiences.

HOW SUPPLIED

16 /STORAGE AND HANDLING Brimonidine tartrate ophthalmic solution 0.2% is supplied sterile, in white opaque plastic dropper bottles as follows: 5 mL NDC 17478-715-10 10 mL NDC 17478-715-11 15 mL NDC 17478-715-12 Storage: Store at 20° to 25°C (68° to 77°F) [see USP Controlled Room Temperature].

GERIATRIC USE

8.5 Geriatric Use No overall differences in safety or effectiveness have been observed between elderly and other adult patients.

DOSAGE FORMS AND STRENGTHS

3 Solution containing 2 mg/mL brimonidine tartrate.

Solution containing 2 mg/mL brimonidine tartrate.

( 3 )

MECHANISM OF ACTION

12.1 Mechanism of Action Brimonidine tartrate ophthalmic solution 0.2% is a relatively selective alpha-2 adrenergic receptor agonist with a peak ocular hypotensive effect occurring at two hours post-dosing.

Fluorophotometric studies in animals and humans suggest that brimonidine tartrate has a dual mechanism of action by reducing aqueous humor production and increasing uveoscleral outflow.

INDICATIONS AND USAGE

1 Brimonidine tartrate ophthalmic solution 0.2% is indicated for lowering intraocular pressure (IOP) in patients with open-angle glaucoma or ocular hypertension.

The IOP lowering efficacy of brimonidine tartrate ophthalmic solution diminishes over time in some patients.

This loss of effect appears with a variable time of onset in each patient and should be closely monitored.

Brimonidine tartrate ophthalmic solution 0.2% is an alpha adrenergic agonist indicated for lowering intraocular pressure (IOP) in patients with open-angle glaucoma or ocular hypertension.

( 1 )

PEDIATRIC USE

8.4 Pediatric Use Brimonidine tartrate ophthalmic solution is contraindicated in children under the age of 2 years [ see Contraindications ( 4.1 ) ].

During postmarketing surveillance, apnea, bradycardia, coma, hypotension, hypothermia, hypotonia, lethargy, pallor, respiratory depression, and somnolence have been reported in infants receiving brimonidine.

The safety and effectiveness of brimonidine tartrate have not been studied in children below the age of 2 years.

In a well-controlled clinical study conducted in pediatric glaucoma patients (ages 2 to 7 years) the most commonly observed adverse reactions with brimonidine tartrate ophthalmic solution 0.2% dosed three times daily were somnolence (50% to 83% in patients ages 2 to 6 years) and decreased alertness.

In pediatric patients 7 years of age (greater than 20 kg), somnolence appears to occur less frequently (25%).

Approximately 16% of patients on brimonidine tartrate ophthalmic solution 0.2% discontinued from the study due to somnolence.

PREGNANCY

8.1 Pregnancy Pregnancy Category B: Teratogenicity studies have been performed in animals.

Brimonidine tartrate was not teratogenic when given orally during gestation days 6 through 15 in rats and days 6 through 18 in rabbits.

The highest doses of brimonidine tartrate in rats (2.5 mg/kg/day) and rabbits (5 mg/kg/day) achieved AUC exposure values 375-fold higher or 19-fold higher, respectively, than similar values estimated in humans treated with brimonidine tartrate ophthalmic solution 0.2%, one drop in one eye, twice daily.

There are no adequate and well-controlled studies in pregnant women; however, in animal studies, brimonidine crossed the placenta and entered into the fetal circulation to a limited extent.

Because animal reproduction studies are not always predictive of human response, brimonidine tartrate ophthalmic solution should be used during pregnancy only if the potential benefit to the mother justifies the potential risk to the fetus.

NUSRING MOTHERS

8.3 Nursing Mothers It is not known whether brimonidine tartrate is excreted in human milk, although in animal studies, brimonidine tartrate has been shown to be excreted in breast milk.

Because of the potential for serious adverse reactions from brimonidine tartrate ophthalmic solution in nursing infants, 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.

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS Potentiation of vascular insufficiency.

( 5.1 ) 5.1 Potentiation of Vascular Insufficiency Brimonidine tartrate ophthalmic solution may potentiate syndromes associated with vascular insufficiency.

Brimonidine tartrate ophthalmic solution should be used with caution in patients with depression, cerebral or coronary insufficiency, Raynaud’s phenomenon, orthostatic hypotension, or thromboangiitis obliterans.

5.2 Severe Cardiovascular Disease Although brimonidine tartrate ophthalmic solution had minimal effect on the blood pressure of patients in clinical studies, caution should be exercised in treating patients with severe cardiovascular disease.

5.3 Contamination of Topical Ophthalmic Products After Use There have been reports of bacterial keratitis associated with the use of multiple-dose containers of topical ophthalmic products.

These containers had been inadvertently contaminated by patients who, in most cases, had a concurrent corneal disease or a disruption of the ocular epithelial surface [ see Patient Counseling Information ( 17 ) ].

5.4 Use with Contact Lenses The preservative in brimonidine tartrate ophthalmic solution, benzalkonium chloride, may be absorbed by soft contact lenses.

Patients wearing soft contact lenses should be instructed to wait at least 15 minutes after instilling brimonidine tartrate ophthalmic solution to insert soft contact lenses.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION Handling the Container Instruct patients that ocular solutions, if handled improperly or if the tip of the dispensing container contacts the eye or surrounding structures, can become contaminated by common bacteria known to cause ocular infections.

Serious damage to the eye and subsequent loss of vision may result from using contaminated solutions [ see Warnings and Precautions ( 5.3 ) ].

Always replace the cap after using.

If solution changes color or becomes cloudy, do not use.

Do not use the product after the expiration date marked on the bottle.

When to Seek Physician Advice Advise patients that if they have ocular surgery or develop an intercurrent ocular condition (e.g., trauma or infection), they should immediately seek their physician’s advice concerning the continued use of the present multidose container.

Use with Contact Lenses Advise patients that contact lenses should be removed prior to instillation of brimonidine tartrate ophthalmic solution and may be reinserted 15 minutes following its administration.

Use with Other Ophthalmic Drugs Advise patients that if more than one topical ophthalmic drug is being used, the drugs should be administered at least five minutes apart.

Potential for Decreased Mental Alertness As with other similar medications, brimonidine tartrate ophthalmic solution may cause fatigue and/or drowsiness in some patients.

Caution patients who engage in hazardous activities of the potential for a decrease in mental alertness.

AKORN Distributed by: Akorn Operating Company LLC Gurnee, IL 60031 BT00N Rev.

05/22

DOSAGE AND ADMINISTRATION

2 The recommended dose is one drop of brimonidine tartrate ophthalmic solution 0.2% in the affected eye(s) three times daily, approximately 8 hours apart.

Brimonidine tartrate ophthalmic solution may be used concomitantly with other topical ophthalmic drug products to lower intraocular pressure.

If more than one topical ophthalmic product is to be used, the different products should be instilled at least 5 minutes apart.

One drop in the affected eye(s), three times daily, approximately 8 hours apart.

( 2 )

HCTZ 12.5 MG / valsartan 80 MG Oral Tablet

DRUG INTERACTIONS

7 Valsartan-Hydrochlorothiazide: Lithium: Increases in serum lithium concentrations and lithium toxicity have been reported during concomitant administration of lithium with angiotensin II receptor antagonists or thiazides.

Monitor lithium levels in patients taking valsartan and hydrochlorothiazide.

Valsartan: No clinically significant pharmacokinetic interactions were observed when valsartan was coadministered with amlodipine, atenolol, cimetidine, digoxin, furosemide, glyburide, hydrochlorothiazide or indomethacin.

The valsartan-atenolol combination was more antihypertensive than either component, but it did not lower the heart rate more than atenolol alone.

Coadministration of valsartan and warfarin did not change the pharmacokinetics of valsartan or the time-course of the anticoagulant properties of warfarin.

CYP 450 Interactions: In vitro metabolism studies indicate that CYP 450 mediated drug interactions between valsartan and coadministered drugs are unlikely because of the low extent of metabolism [see Clinical Pharmacology (12.3) ] .

Transporters: The results from an in vitro study with human liver tissue indicate that valsartan is a substrate of the hepatic uptake transporter OATP1B1 and the hepatic efflux transporter MRP2.

Coadministration of inhibitors of the uptake transporter (rifampin, cyclosporine) or efflux transporter (ritonavir) may increase the systemic exposure to valsartan.

Non-Steroidal Anti-Inflammatory Agents including Selective Cyclooxygenase-2 Inhibitors (COX-2 Inhibitors): In patients who are elderly, volume-depleted (including those on diuretic therapy), or with compromised renal function, coadministration of NSAIDs, including selective COX-2 inhibitors, with angiotensin II receptor antagonists, including valsartan, may result in deterioration of renal function, including possible acute renal failure.

These effects are usually reversible.

Monitor renal function periodically in patients receiving valsartan and NSAID therapy.

The antihypertensive effect of angiotensin II receptor antagonists, including valsartan may be attenuated by NSAIDs including selective COX-2 inhibitors.

Potassium : Concomitant use of valsartan with other agents that block the renin-angiotensin system, potassium sparing diuretics (e.g., spironolactone, triamterene, amiloride), potassium supplements, or salt substitutes containing potassium may lead to increases in serum potassium and in heart failure patients to increases in serum creatinine.

If co-medication is considered necessary, monitoring of serum potassium is advisable.

Dual Blockade of the Renin-Angiotensin System (RAS): Dual blockade of the RAS with angiotensin receptor blockers, ACE inhibitors or aliskiren is associated with increased risks of hypotension, hyperkalemia, and changes in renal function (including acute renal failure) compared to monotherapy.

Closely monitor blood pressure, renal function and electrolytes in patients on valsartan and hydrochlorothiazide and other agents that affect the RAS.

Do not coadminister aliskiren with valsartan and hydrochlorothiazide in patients with diabetes.

Avoid use of aliskiren with valsartan and hydrochlorothiazide in patients with renal impairment (GFR < 60 mL/min).

Hydrochlorothiazide: When administered concurrently, the following drugs may interact with thiazide diuretics: Antidiabetic Drugs (oral agents and insulin): Dosage adjustment of the antidiabetic drug may be required.

Nonsteroidal Anti-inflammatory Drugs (NSAIDS and COX-2 selective inhibitors): When valsartan and hydrochlorothiazide and non-steroidal anti-inflammatory agents are used concomitantly, the patient should be observed closely to determine if the desired effect of the diuretic is obtained.

Carbamazepine: May lead to symptomatic hyponatremia.

Ion exchange resins: Staggering the dosage of hydrochlorothiazide and ion exchange resins (e.g., cholestyramine, colestipol) such that hydrochlorothiazide is administered at least 4 hours before or 4 to 6 hours after the administration of resins would potentially minimize the interaction [see Clinical Pharmacology (12.3) ].

Cyclosporine: Concomitant treatment with cyclosporine may increase the risk of hyperuricemia and gout-type complications.

Antidiabetic drugs: Dosage adjustment of antidiabetic may be required.

( 7 ) Cholestyramine and colestipol: Reduced absorption of thiazides.

( 12.3 ) Lithium: Increased risk of lithium toxicity.

Monitor serum lithium concentrations during concurrent use.

( 7 ) Non-Steroidal Anti-Inflammatory Drugs (NSAIDs): May increase risk of renal impairment.

Can reduce diuretic, natriuretic and antihypertensive effects of diuretics.

( 7 ) Dual inhibition of the renin-angiotesin system: Increased risk of renal impairment, hypotension and hyperkalemia.

( 7 )

OVERDOSAGE

10 Valsartan and Hydrochlorothiazide: Limited data are available related to overdosage in humans.

The most likely manifestations of overdosage would be hypotension and tachycardia; bradycardia could occur from parasympathetic (vagal) stimulation.

Depressed level of consciousness, circulatory collapse and shock have been reported.

If symptomatic hypotension should occur, supportive treatment should be instituted.

Valsartan is not removed from the plasma by dialysis.

The degree to which hydrochlorothiazide is removed by hemodialysis has not been established.

The most common signs and symptoms observed in patients are those caused by electrolyte depletion (hypokalemia, hypochloremia, hyponatremia) and dehydration resulting from excessive diuresis.

If digitalis has also been administered, hypokalemia may accentuate cardiac arrhythmias.

In rats and marmosets, single oral doses of valsartan up to 1524 and 762 mg/kg in combination with hydrochlorothiazide at doses up to 476 and 238 mg/kg, respectively, were very well tolerated without any treatment-related effects.

These no adverse effect doses in rats and marmosets, respectively, represent 46.5 and 23 times the maximum recommended human dose (MRHD) of valsartan and 188 and 113 times the MRHD of hydrochlorothiazide on a mg/m 2 basis.

(Calculations assume an oral dose of 320 mg/day valsartan in combination with 25 mg/day hydrochlorothiazide and a 60 kg patient.) Valsartan: Valsartan was without grossly observable adverse effects at single oral doses up to 2000 mg/kg in rats and up to 1000 mg/kg in marmosets, except for salivation and diarrhea in the rat and vomiting in the marmoset at the highest dose (60 and 31 times, respectively, the MRHD on a mg/m 2 basis).

(Calculations assume an oral dose of 320 mg/day and a 60 kg patient.) Hydrochlorothiazide: The oral LD 50 of hydrochlorothiazide is greater than 10 g/kg in both mice and rats, which represents 2,027 and 4,054 times, respectively, the MRHD on a mg/m 2 basis.

(Calculations assume an oral dose of 25 mg/day and a 60 kg patient.)

DESCRIPTION

11 Valsartan and hydrochlorothiazide tablets, USP is a combination of valsartan, an orally active, specific angiotensin II receptor blocker (ARB) acting on the AT 1 receptor subtype, and hydrochlorothiazide, a diuretic.

Valsartan, a nonpeptide molecule, is chemically described as N -[p-(o-1 H -Tetrazol-5-ylphenyl)benzyl- N -Valeryl-L-valine.

Its molecular formula is C 24 H 29 N 5 O 3 , its molecular weight is 435.5, and its structural formula is Valsartan, USP is a white to practically white fine powder.

It is soluble in ethanol and methanol and slightly soluble in water.

Hydrochlorothiazide, USP is a white, or practically white, practically odorless, crystalline powder.

It is slightly soluble in water; freely soluble in sodium hydroxide solution, in n -butylamine, and in dimethylformamide; sparingly soluble in methanol; and insoluble in ether, in chloroform, and in dilute mineral acids.

Hydrochlorothiazide is chemically described as 6-chloro-3,4-dihydro-2 H -1,2,4-benzothiadiazine-7-sulfonamide 1,1-dioxide.

Hydrochlorothiazide is a thiazide diuretic.

Its molecular formula is C 7 H 8 ClN 3 O 4 S 2 , its molecular weight is 297.73, and its structural formula is Valsartan and hydrochlorothiazide tablets are formulated for oral administration to contain valsartan and hydrochlorothiazide 80 mg/12.5 mg, 160 mg/12.5 mg, 160 mg/25 mg, 320 mg/12.5 mg and 320 mg/25 mg.

The inactive ingredients of the tablets are colloidal silicon dioxide, crospovidone, hypromellose, lactose monohydrate, magnesium stearate, microcrystalline cellulose, polydextrose, polyethylene glycol, povidone, pregelatinized starch (corn), sodium lauryl sulfate, titanium dioxide and triacetin.

The 80 mg/12.5 mg, 160 mg/12.5 mg, 160 mg/25 mg and 320 mg/12.5 mg tablets also contain red iron oxide and yellow iron oxide.

The 320 mg/25 mg tablet also contains FD&C Blue No.

2 Aluminum Lake and FD&C Yellow No.

6 Aluminum Lake.

Valsartan Structural Formula Hydrochlorothiazide Structural Formula

CLINICAL STUDIES

14 14.1 Hypertension Valsartan and Hydrochlorothiazide In controlled clinical trials including over 7,600 patients, 4,372 patients were exposed to valsartan (80 mg, 160 mg and 320 mg) and concomitant hydrochlorothiazide (12.5 mg and 25 mg).

Two factorial trials compared various combinations of 80 mg/12.5 mg, 80 mg/25 mg, 160 mg/12.5 mg, 160 mg/25 mg, 320 mg/12.5 mg and 320 mg/25 mg with their respective components and placebo.

The combination of valsartan and hydrochlorothiazide resulted in additive placebo-adjusted decreases in systolic and diastolic blood pressure at trough of 14 to 21/8 to 11 mmHg at 80 mg/12.5 mg to 320 mg/25 mg, compared to 7 to 10/4 to 5 mmHg for valsartan 80 mg to 320 mg and 5 to 11/2 to 5 mmHg for hydrochlorothiazide 12.5 mg to 25 mg, alone.

Three other controlled trials investigated the addition of hydrochlorothiazide to patients who did not respond adequately to valsartan 80 mg to valsartan 320 mg, resulted in the additional lowering of systolic and diastolic blood pressure by approximately 4 to 12/2 to 5 mmHg.

The maximal antihypertensive effect was attained 4 weeks after the initiation of therapy, the first time point at which blood pressure was measured in these trials.

In long-term follow-up studies (without placebo control) the effect of the combination of valsartan and hydrochlorothiazide appeared to be maintained for up to 2 years.

The antihypertensive effect is independent of age or gender.

The overall response to the combination was similar for Black and non-Black patients.

There was essentially no change in heart rate in patients treated with the combination of valsartan and hydrochlorothiazide in controlled trials.

There are no trials of the valsartan and hydrochlorothiazide combination tablet demonstrating reductions in cardiovascular risk in patients with hypertension, but the hydrochlorothiazide component and several ARBs, which are the same pharmacological class as the valsartan component, have demonstrated such benefits.

Valsartan The antihypertensive effects of valsartan were demonstrated principally in seven placebo-controlled, 4- to 12-week trials (one in patients over 65) of dosages from 10 to 320 mg/day in patients with baseline diastolic blood pressures of 95 to 115.

The studies allowed comparison of once daily and twice daily regimens of 160 mg/day; comparison of peak and trough effects; comparison (in pooled data) of response by gender, age and race; and evaluation of incremental effects of hydrochlorothiazide.

Administration of valsartan to patients with essential hypertension results in a significant reduction of sitting, supine, and standing systolic and diastolic blood pressure, usually with little or no orthostatic change.

In most patients, after administration of a single oral dose, onset of antihypertensive activity occurs at approximately 2 hours, and maximum reduction of blood pressure is achieved within 6 hours.

The antihypertensive effect persists for 24 hours after dosing, but there is a decrease from peak effect at lower doses (40 mg) presumably reflecting loss of inhibition of angiotensin II.

At higher doses, however (160 mg), there is little difference in peak and trough effect.

During repeated dosing, the reduction in blood pressure with any dose is substantially present within 2 weeks, and maximal reduction is generally attained after 4 weeks.

In long-term follow-up studies (without placebo control) the effect of valsartan appeared to be maintained for up to 2 years.

The antihypertensive effect is independent of age, gender or race.

The latter finding regarding race is based on pooled data and should be viewed with caution, because antihypertensive drugs that affect the renin-angiotensin system (that is, ACE inhibitors and angiotensin II blockers) have generally been found to be less effective in low-renin hypertensives (frequently Blacks) than in high-renin hypertensives (frequently Whites).

In pooled, randomized, controlled trials of valsartan that included a total of 140 Blacks and 830 Whites, valsartan and an ACE-inhibitor control were generally at least as effective in Blacks as Whites.

The explanation for this difference from previous findings is unclear.

Abrupt withdrawal of valsartan has not been associated with a rapid increase in blood pressure.

The seven studies of valsartan monotherapy included over 2,000 patients randomized to various doses of valsartan and about 800 patients randomized to placebo.

Doses below 80 mg were not consistently distinguished from those of placebo at trough, but doses of 80 mg, 160 mg and 320 mg produced dose related decreases in systolic and diastolic blood pressure, with the difference from placebo of approximately 6 to 9/3 to 5 mmHg at 80 mg to 160 mg and 9/6 mmHg at 320 mg.

Patients with an inadequate response to 80 mg once daily were titrated to either 160 mg once daily or 80 mg twice daily, which resulted in a comparable response in both groups.

In another 4-week study, 1,876 patients randomized to valsartan 320 mg once daily had an incremental blood pressure reduction 3/1 mmHg lower than did 1,900 patients randomized to valsartan 160 mg once daily.

In controlled trials, the antihypertensive effect of once daily valsartan 80 mg was similar to that of once daily enalapril 20 mg or once daily lisinopril 10 mg.

There was essentially no change in heart rate in valsartan-treated patients in controlled trials.

14.2 Initial Therapy – Hypertension The safety and efficacy of valsartan and hydrochlorothiazide as initial therapy for patients with severe hypertension (defined as a sitting diastolic blood pressure ≥ 110 mmHg and systolic blood pressure ≥ 140 mmHg off all antihypertensive therapy) was studied in a 6-week multicenter, randomized, double-blind study.

Patients were randomized to either valsartan and hydrochlorothiazide 160 mg/12.5 mg once daily or to valsartan (160 mg once daily) and followed for blood pressure response.

Patients were force-titrated at 2 week intervals.

Patients on combination therapy were subsequently titrated to 160 mg/25 mg followed by 320 mg/25 mg valsartan and hydrochlorothiazide.

Patients on monotherapy were subsequently titrated to 320 mg valsartan followed by a titration to 320 mg valsartan to maintain the blind.

The study randomized 608 patients, including 261 (43%) females, 147 (24%) Blacks and 75 (12%) ≥ 65 years of age.

The mean blood pressure at baseline for the total population was 168/112 mmHg.

The mean age was 52 years.

After 4 weeks of therapy, reductions in systolic and diastolic blood pressure were 9/5 mmHg greater in the group treated with valsartan and hydrochlorothiazide compared to valsartan.

Similar trends were seen when the patients were grouped according to gender, race or age.

HOW SUPPLIED

16 /STORAGE AND HANDLING Valsartan and Hydrochlorothiazide Tablets, USP are available containing 80 mg/12.5 mg, 160 mg/12.5 mg or 160 mg/25 mg of valsartan, USP and hydrochlorothiazide, USP The 80 mg/12.5 mg tablets are an orange film-coated, round, unscored tablets, debossed with M on one side of the tablet and V21 on the other side.

They are available as follows: NDC 51079-192-03 – Unit dose blister packages of 30 (3 cards of 10 tablets each).

The 160 mg/12.5 mg tablets are an orange film-coated, round, unscored tablets, debossed with M on one side of the tablet and V22 on the other side.

They are available as follows: NDC 51079-193-03 – Unit dose blister packages of 30 (3 cards of 10 tablets each).

The 160 mg/25 mg tablets are an orange film-coated, oval, unscored tablets, debossed with M on one side of the tablet and V23 on the other side.

They are available as follows: NDC 51079-194-03 – Unit dose blister packages of 30 (3 cards of 10 tablets each).

Store at 20° to 25°C (68° to 77°F).

[See USP Controlled Room Temperature.] Protect from light, moisture and heat.

PHARMACIST: Dispense the Patient Information Leaflet with each prescription.

GERIATRIC USE

8.5 Geriatric Use In the controlled clinical trials of valsartan and hydrochlorothiazide, 764 (17.5%) patients treated with valsartan and hydrochlorothiazide were ≥ 65 years and 118 (2.7%) were ≥ 75 years.

No overall difference in the efficacy or safety of valsartan and hydrochlorothiazide was observed between these patients and younger patients, but greater sensitivity of some older individuals cannot be ruled out.

DOSAGE FORMS AND STRENGTHS

3 80 mg/12.5 mg tablets, debossed with M on one side of the tablet and V21 on the other side.

160 mg/12.5 mg tablets, debossed with M on one side of the tablet and V22 on the other side.

160 mg/25 mg tablets, debossed with M on one side of the tablet and V23 on the other side.

320 mg/12.5 mg tablets, debossed with M on one side of the tablet and V24 on the other side.

320 mg/25 mg tablets, debossed with M on one side of the tablet and V25 on the other side.

Tablets (valsartan and HCTZ): 80 mg/12.5 mg, 160 mg/12.5 mg, 160 mg/25 mg, 320 mg/12.5 mg, 320 mg/25 mg ( 3 )

MECHANISM OF ACTION

12.1 Mechanism of Action Angiotensin II is formed from angiotensin I in a reaction catalyzed by angiotensin-converting enzyme (ACE, kininase II).

Angiotensin II is the principal pressor agent of the renin-angiotensin system, with effects that include vasoconstriction, stimulation of synthesis and release of aldosterone, cardiac stimulation, and renal reabsorption of sodium.

Valsartan blocks the vasoconstrictor and aldosterone-secreting effects of angiotensin II by selectively blocking the binding of angiotensin II to the AT 1 receptor in many tissues, such as vascular smooth muscle and the adrenal gland.

Its action is therefore independent of the pathways for angiotensin II synthesis.

There is also an AT 2 receptor found in many tissues, but AT 2 is not known to be associated with cardiovascular homeostasis.

Valsartan has much greater affinity (about 20,000-fold) for the AT 1 receptor than for the AT 2 receptor.

The primary metabolite of valsartan is essentially inactive with an affinity for the AT 1 receptor about one 200 th that of valsartan itself.

Blockade of the renin-angiotensin system with ACE inhibitors, which inhibit the biosynthesis of angiotensin II from angiotensin I, is widely used in the treatment of hypertension.

ACE inhibitors also inhibit the degradation of bradykinin, a reaction also catalyzed by ACE.

Because valsartan does not inhibit ACE (kininase II) it does not affect the response to bradykinin.

Whether this difference has clinical relevance is not yet known.

Valsartan does not bind to or block other hormone receptors or ion channels known to be important in cardiovascular regulation.

Blockade of the angiotensin II receptor inhibits the negative regulatory feedback of angiotensin II on renin secretion, but the resulting increased plasma renin activity and angiotensin II circulating levels do not overcome the effect of valsartan on blood pressure.

Hydrochlorothiazide is a thiazide diuretic.

Thiazides affect the renal tubular mechanisms of electrolyte reabsorption, directly increasing excretion of sodium and chloride in approximately equivalent amounts.

Indirectly, the diuretic action of hydrochlorothiazide reduces plasma volume, with consequent increases in plasma renin activity, increases in aldosterone secretion, increases in urinary potassium loss and decreases in serum potassium.

The renin-aldosterone link is mediated by angiotensin II, so coadministration of an angiotensin II receptor antagonist tends to reverse the potassium loss associated with these diuretics.

The mechanism of the antihypertensive effect of thiazides is unknown.

INDICATIONS AND USAGE

1 Valsartan and hydrochlorothiazide tablets are indicated for the treatment of hypertension, to lower blood pressure.

Lowering blood pressure reduces the risk of fatal and nonfatal cardiovascular events, primarily strokes and myocardial infarctions.

These benefits have been seen in controlled trials of antihypertensive drugs from a wide variety of pharmacologic classes, including hydrochlorothiazide and the ARB class to which valsartan principally belongs.

There are no controlled trials demonstrating risk reduction with valsartan and hydrochlorothiazide tablets.

Control of high blood pressure should be part of comprehensive cardiovascular risk management, including, as appropriate, lipid control, diabetes management, antithrombotic therapy, smoking cessation, exercise and limited sodium intake.

Many patients will require more than one drug to achieve blood pressure goals.

For specific advice on goals and management, see published guidelines, such as those of the National High Blood Pressure Education Program’s Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure (JNC).

Numerous antihypertensive drugs, from a variety of pharmacologic classes and with different mechanisms of action, have been shown in randomized controlled trials to reduce cardiovascular morbidity and mortality, and it can be concluded that it is blood pressure reduction, and not some other pharmacologic property of the drugs, that is largely responsible for those benefits.

The largest and most consistent cardiovascular outcome benefit has been a reduction in the risk of stroke, but reductions in myocardial infarction and cardiovascular mortality have also been seen regularly.

Elevated systolic or diastolic pressure causes increased cardiovascular risk, and the absolute risk increase per mmHg is greater at higher blood pressures, so that even modest reductions of severe hypertension can provide substantial benefit.

Relative risk reduction from blood pressure reduction is similar across populations with varying absolute risk, so the absolute benefit is greater in patients who are at higher risk independent of their hypertension (for example, patients with diabetes or hyperlipidemia), and such patients would be expected to benefit from more aggressive treatment to a lower blood pressure goal.

Some antihypertensive drugs have smaller blood pressure effects (as monotherapy) in black patients, and many antihypertensive drugs have additional approved indications and effects (e.g., on angina, heart failure or diabetic kidney disease).

These considerations may guide selection of therapy.

Add-On Therapy: Valsartan and hydrochlorothiazide tablets may be used in patients whose blood pressure is not adequately controlled on monotherapy.

Replacement Therapy: Valsartan and hydrochlorothiazide tablets may be substituted for the titrated components.

Initial Therapy: Valsartan and hydrochlorothiazide tablets may be used as initial therapy in patients who are likely to need multiple drugs to achieve blood pressure goals.

The choice of valsartan and hydrochlorothiazide tablets as initial therapy for hypertension should be based on an assessment of potential benefits and risks.

Patients with stage 2 hypertension are at a relatively high risk for cardiovascular events (such as strokes, heart attacks and heart failure), kidney failure and vision problems, so prompt treatment is clinically relevant.

The decision to use a combination as initial therapy should be individualized and should be shaped by considerations such as baseline blood pressure, the target goal and the incremental likelihood of achieving goal with a combination compared to monotherapy.

Individual blood pressure goals may vary based upon the patient’s risk.

Data from the high dose multifactorial trial [see Clinical Studies (14.1) ] provides estimates of the probability of reaching a target blood pressure with valsartan and hydrochlorothiazide tablets compared to valsartan or hydrochlorothiazide monotherapy.

The figures below provide estimates of the likelihood of achieving systolic or diastolic blood pressure control with valsartan and hydrochlorothiazide tablets 320 mg/25 mg, based upon baseline systolic or diastolic blood pressure.

The curve of each treatment group was estimated by logistic regression modeling.

The estimated likelihood at the right tail of each curve is less reliable due to small numbers of subjects with high baseline blood pressures.

For example, a patient with a baseline blood pressure of 160/100 mmHg has about a 41% likelihood of achieving a goal of < 140 mmHg (systolic) and 60% likelihood of achieving < 90 mmHg (diastolic) on valsartan alone and the likelihood of achieving these goals on HCTZ alone is about 50% (systolic) or 57% (diastolic).

The likelihood of achieving these goals on valsartan and hydrochlorothiazide tablets rises to about 84% (systolic) or 80% (diastolic).

The likelihood of achieving these goals on placebo is about 23% (systolic) or 36% (diastolic).

Valsartan and hydrochlorothiazide is the combination tablet of valsartan, an angiotensin II receptor blocker (ARB) and hydrochlorothiazide (HCTZ), a diuretic.

Valsartan and hydrochlorothiazide tablets are indicated for the treatment of hypertension, to lower blood pressure: In patients not adequately controlled with monotherapy ( 1 ) As initial therapy in patients likely to need multiple drugs to achieve their blood pressure goals ( 1 ) Lowering blood pressure reduces the risk of fatal and nonfatal cardiovascular events, primarily strokes and myocardial infarctions.

Figure 1.

Probability of Achieving Systolic Blood Pressure  140 mm/Hg at Week 8 Figure 2.

Probability of Achieving Diastolic Blood Pressure  90 mm/Hg at Week 8 Figure 3.

Probability of Achieving Systolic Blood Pressure  130 mm/Hg at Week 8 Figure 4.

Probability of Achieving Diastolic Blood Pressure  80 mm/Hg at Week 8

PEDIATRIC USE

8.4 Pediatric Use Safety and effectiveness of valsartan and hydrochlorothiazide in pediatric patients have not been established.

Neonates with a history of in utero exposure to valsartan and hydrochlorothiazide: If oliguria or hypotension occurs, direct attention toward support of blood pressure and renal perfusion.

Exchange transfusions or dialysis may be required as a means of reversing hypotension and/or substituting for disordered renal function.

PREGNANCY

8.1 Pregnancy Teratogenic Effects.

Pregnancy Category D Use of drugs that act on the renin-angiotensin system during the second and third trimesters of pregnancy reduces fetal renal function and increases fetal and neonatal morbidity and death.

Resulting oligohydramnios can be associated with fetal lung hypoplasia and skeletal deformations.

Potential neonatal adverse effects include skull hypoplasia, anuria, hypotension, renal failure and death.

When pregnancy is detected, discontinue valsartan and hydrochlorothiazide as soon as possible.

These adverse outcomes are usually associated with use of these drugs in the second and third trimester of pregnancy.

Most epidemiologic studies examining fetal abnormalities after exposure to antihypertensive use in the first trimester have not distinguished drugs affecting the renin-angiotensin system from other antihypertensive agents.

Appropriate management of maternal hypertension during pregnancy is important to optimize outcomes for both mother and fetus.

In the unusual case that there is no appropriate alternative to therapy with drugs affecting the renin-angiotensin system for a particular patient, apprise the mother of the potential risk to the fetus.

Perform serial ultrasound examinations to assess the intra-amniotic environment.

If oligohydramnios is observed, discontinue valsartan and hydrochlorothiazide, unless it is considered lifesaving for the mother.

Fetal testing may be appropriate, based on the week of pregnancy.

Patients and physicians should be aware, however, that oligohydramnios may not appear until after the fetus has sustained irreversible injury.

Closely observe infants with histories of in utero exposure to valsartan and hydrochlorothiazide for hypotension, oliguria and hyperkalemia [see Use in Specific Populations (8.4) ] .

Hydrochlorothiazide Thiazides can cross the placenta, and concentrations reached in the umbilical vein approach those in the maternal plasma.

Hydrochlorothiazide, like other diuretics, can cause placental hypoperfusion.

It accumulates in the amniotic fluid, with reported concentrations up to 19 times higher than in umbilical vein plasma.

Use of thiazides during pregnancy is associated with a risk of fetal or neonatal jaundice or thrombocytopenia.

Since they do not prevent or alter the course of EPH (Edema, Proteinuria, Hypertension) gestosis (pre-eclampsia), these drugs should not be used to treat hypertension in pregnant women.

The use of hydrochlorothiazide for other indications (e.g., heart disease) in pregnancy should be avoided.

NUSRING MOTHERS

8.3 Nursing Mothers It is not known whether valsartan is excreted in human milk.

Valsartan was excreted into the milk of lactating rats; however, animal breast milk drug levels may not accurately reflect human breast milk levels.

Hydrochlorothiazide is excreted in human breast milk.

Because many drugs are excreted into human milk and because of the potential for adverse reactions in nursing infants from valsartan and hydrochlorothiazide, a decision should be made whether to discontinue nursing or discontinue the drug, taking into account the importance of the drug to the mother.

BOXED WARNING

WARNING: FETAL TOXICITY When pregnancy is detected, discontinue valsartan and hydrochlorothiazide tablets as soon as possible.

( 5.1 ) Drugs that act directly on the renin-angiotensin system can cause injury and death to the developing fetus.

( 5.1 ) WARNING: FETAL TOXICITY See full prescribing information for complete boxed warning.

When pregnancy is detected, discontinue valsartan and hydrochlorothiazide tablets as soon as possible.

( 5.1 ) Drugs that act directly on the renin-angiotensin system can cause injury and death to the developing fetus.

( 5.1 )

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS Hypotension: Correct volume depletion prior to initiation ( 5.2 ) Observe for signs of fluid or electrolyte imbalance ( 5.9 ) Monitor renal function and potassium in susceptible patients ( 5.3 , 5.7 ) Exacerbation or activation of systemic lupus erythematosus ( 5.5 ) Acute angle-closure glaucoma ( 5.8 ) 5.1 Fetal Toxicity Pregnancy Category D Use of drugs that act on the renin-angiotensin system during the second and third trimesters of pregnancy reduces fetal renal function and increases fetal and neonatal morbidity and death.

Resulting oligohydramnios can be associated with fetal lung hypoplasia and skeletal deformations.

Potential neonatal adverse effects include skull hypoplasia, anuria, hypotension, renal failure and death.

When pregnancy is detected, discontinue valsartan and hydrochlorothiazide as soon as possible [see Use in Specific Populations (8.1) ] .

Intrauterine exposure to thiazide diuretics is associated with fetal or neonatal jaundice, thrombocytopenia, and possibly other adverse reactions that have occurred in adults.

5.2 Hypotension in Volume- and/or Salt-Depleted Patients Excessive reduction of blood pressure was rarely seen (0.7%) in patients with uncomplicated hypertension treated with valsartan and hydrochlorothiazide in controlled trials.

In patients with an activated renin-angiotensin system, such as volume- and/or salt-depleted patients receiving high doses of diuretics, symptomatic hypotension may occur.

This condition should be corrected prior to administration of valsartan and hydrochlorothiazide, or the treatment should start under close medical supervision.

If hypotension occurs, the patient should be placed in the supine position and, if necessary, given an intravenous infusion of normal saline.

A transient hypotensive response is not a contraindication to further treatment, which usually can be continued without difficulty once the blood pressure has stabilized.

5.3 Impaired Renal Function Changes in renal function including acute renal failure can be caused by drugs that inhibit the renin-angiotensin system and by diuretics.

Patients whose renal function may depend in part on the activity of the renin-angiotensin system (e.g., patients with renal artery stenosis, chronic kidney disease, severe congestive heart failure or volume depletion) may be at particular risk of developing acute renal failure on valsartan and hydrochlorothiazide.

Monitor renal function periodically in these patients.

Consider withholding or discontinuing therapy in patients who develop a clinically significant decrease in renal function on valsartan and hydrochlorothiazide [see Drug Interactions (7) ] .

5.4 Hypersensitivity Reaction Hydrochlorothiazide Hypersensitivity reactions to hydrochlorothiazide may occur in patients with or without a history of allergy or bronchial asthma, but are more likely in patients with such a history.

5.5 Systemic Lupus Erythematosus Hydrochlorothiazide Thiazide diuretics have been reported to cause exacerbation or activation of systemic lupus erythematosus.

5.6 Lithium Interaction Increases in serum lithium concentrations and lithium toxicity have been reported with concomitant use of valsartan or thiazide diuretics.

Monitor lithium levels in patients receiving valsartan and hydrochlorothiazide and lithium [see Drug Interactions (7) ] .

5.7 Potassium Abnormalities Valsartan and Hydrochlorothiazide In the controlled trials of various doses of valsartan and hydrochlorothiazide the incidence of hypertensive patients who developed hypokalemia (serum potassium 5.7 mEq/L) was 0.4%.

Hydrochlorothiazide can cause hypokalemia and hyponatremia.

Hypomagnesemia can result in hypokalemia which appears difficult to treat despite potassium repletion.

Drugs that inhibit the renin-angiotensin system can cause hyperkalemia.

Monitor serum electrolytes periodically.

If hypokalemia is accompanied by clinical signs (e.g., muscular weakness, paresis or ECG alterations), valsartan and hydrochlorothiazide should be discontinued.

Correction of hypokalemia and any coexisting hypomagnesemia is recommended prior to the initiation of thiazides.

Some patients with heart failure have developed increases in potassium with valsartan therapy.

These effects are usually minor and transient, and they are more likely to occur in patients with preexisting renal impairment.

Dosage reduction and/or discontinuation of the diuretic and/or valsartan may be required [see Adverse Reactions (6.1) ].

5.8 Acute Myopia and Secondary Angle-Closure Glaucoma Hydrochlorothiazide, a sulfonamide, can cause an idiosyncratic reaction, resulting in acute transient myopia and acute angle-closure glaucoma.

Symptoms include acute onset of decreased visual acuity or ocular pain and typically occur within hours to weeks of drug initiation.

Untreated acute angle-closure glaucoma can lead to permanent vision loss.

The primary treatment is to discontinue hydrochlorothiazide as rapidly as possible.

Prompt medical or surgical treatments may need to be considered if the intraocular pressure remains uncontrolled.

Risk factors for developing acute angle-closure glaucoma may include a history of sulfonamide or penicillin allergy.

5.9 Metabolic Disturbances Hydrochlorothiazide Hydrochlorothiazide may alter glucose tolerance and raise serum levels of cholesterol and triglycerides.

Hydrochlorothiazide may raise the serum uric acid level due to reduced clearance of uric acid and may cause or exacerbate hyperuricemia and precipitate gout in susceptible patients.

Hydrochlorothiazide decreases urinary calcium excretion and may cause elevations of serum calcium.

Monitor calcium levels in patients with hypercalcemia receiving valsartan and hydrochlorothiazide.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION Information for Patients: Pregnancy: Female patients of childbearing age should be told about the consequences of exposure to valsartan and hydrochlorothiazide during pregnancy.

Discuss treatment options with women planning to become pregnant.

Patients should be asked to report pregnancies to their physicians as soon as possible.

Symptomatic Hypotension: A patient receiving valsartan and hydrochlorothiazide should be cautioned that lightheadedness can occur, especially during the first days of therapy, and that it should be reported to the prescribing physician.

The patients should be told that if syncope occurs, valsartan and hydrochlorothiazide should be discontinued until the physician has been consulted.

All patients should be cautioned that inadequate fluid intake, excessive perspiration, diarrhea or vomiting can lead to an excessive fall in blood pressure, with the same consequences of lightheadedness and possible syncope.

Potassium Supplements: A patient receiving valsartan and hydrochlorothiazide should be told not to use potassium supplements or salt substitutes containing potassium without consulting the prescribing physician.

FDA-Approved Patient Labeling PATIENT INFORMATION LEAFLET VALSARTAN AND HYDROCHLOROTHIAZIDE TABLETS, USP (val sar’ tan) (hye” droe klor” oh thye’ a zide) 80 mg/12.5 mg, 160 mg/12.5 mg, 160 mg/25 mg, 320 mg/12.5 mg and 320 mg/25 mg Read the Patient Information that comes with valsartan and hydrochlorothiazide tablets before you start taking it and each time you get a refill.

There may be new information.

This leaflet does not take the place of talking with your doctor about your condition and treatment.

If you have any questions about valsartan and hydrochlorothiazide tablets, ask your doctor or pharmacist.

What is the most important information I should know about valsartan and hydrochlorothiazide tablets? Valsartan and hydrochlorothiazide tablets can cause harm or death to an unborn baby.

Talk to your doctor about other ways to lower your blood pressure if you plan to become pregnant.

If you get pregnant while taking valsartan and hydrochlorothiazide tablets, tell your doctor right away.

What are valsartan and hydrochlorothiazide tablets? Valsartan and hydrochlorothiazide tablets contains two prescription medicines: 1.

valsartan, an angiotensin receptor blocker (ARB) 2.

hydrochlorothiazide (HCTZ), a water pill (diuretic) Valsartan and hydrochlorothiazide tablets may be used to lower high blood pressure (hypertension) in adults- when one medicine to lower your high blood pressure is not enough as the first medicine to lower high blood pressure if your doctor decides you are likely to need more than one medicine.

Valsartan and hydrochlorothiazide tablets has not been studied in children under 18 years of age.

Who should not take valsartan and hydrochlorothiazide tablets? Do not take valsartan and hydrochlorothiazide tablets if you: are allergic to any of the ingredients in valsartan and hydrochlorothiazide tablets.

See the end of this leaflet for a complete list of ingredients in valsartan and hydrochlorothiazide tablets.

make less urine due to kidney problems are allergic to medicines that contain sulfonamides.

What should I tell my doctor before taking valsartan and hydrochlorothiazide tablets? Tell your doctor about all your medical conditions including if you: are pregnant or plan to become pregnant.

See “What is the most important information I should know about valsartan and hydrochlorothiazide tablets?” are breast-feeding.

Valsartan and hydrochlorothiazide passes into breast milk.

You should choose either to take valsartan and hydrochlorothiazide tablets or breast-feed, but not both.

have liver problems have kidney problems have or had gallstones have Lupus have low levels of potassium (with or without symptoms such as muscle weakness, muscle spasms, abnormal heart rhythm) or magnesium in your blood have high levels of calcium in your blood (with or without symptoms such as nausea, vomiting, constipation, stomach pain, frequent urination, thirst, muscle weakness and twitching).

have high levels of uric acid in the blood.

have ever had a reaction called angioedema to another blood pressure medication.

Angioedema causes swelling of the face, lips, tongue, throat, and may cause difficulty breathing.

Tell your doctor about all the medicines you take including prescription and non-prescription medicines, vitamins and herbal supplements.

Some of your other medicines and valsartan and hydrochlorothiazide tablets could affect each other, causing serious side effects.

Especially, tell your doctor if you take: other medicines for high blood pressure or a heart problem water pills (diuretics) potassium supplements.

Your doctor may check the amount of potassium in your blood periodically.

a salt substitute.

Your doctor may check the amount of potassium in your blood periodically.

antidiabetic medicines including insulin narcotic pain medicines sleeping pills lithium, a medicine used in some types of depression (Eskalith ®* , Lithobid ®* , Lithium Carbonate, Lithium Citrate) aspirin or other medicines called Non-Steroidal Anti-Inflammatory Drugs (NSAIDs), like ibuprofen or naproxen digoxin or other digitalis glycosides (a heart medicine) muscle relaxants (medicines used during operations) certain cancer medicines, like cyclophosphamide or methotrexate certain antibiotics (rifamycin group), a drug used to protect against transplant rejection (cyclosporin) or an antiretroviral drug used to treat HIV/AIDS infection (ritonavir).

These drugs may increase the effect of valsartan.

Ask your doctor if you are not sure if you are taking one of these medicines.

Know the medicines you take.

Keep a list of your medicines with you to show to your doctor and pharmacist when a new medicine is prescribed.

Talk to your doctor or pharmacist before you start taking any new medicine.

Your doctor or pharmacist will know what medicines are safe to take together.

How should I take valsartan and hydrochlorothiazide tablets? Take valsartan and hydrochlorothiazide tablets exactly as prescribed by your doctor.

Your doctor may change your dose if needed.

Take valsartan and hydrochlorothiazide tablets once each day.

Valsartan and hydrochlorothiazide tablets can be taken with or without food.

If you miss a dose, take it as soon as you remember.

If it is close to your next dose, do not take the missed dose.

Just take the next dose at your regular time.

If you take too much valsartan and hydrochlorothiazide, call your doctor or Poison Control Center, or go to the nearest hospital emergency room.

What should I avoid while taking valsartan and hydrochlorothiazide tablets? You should not take valsartan and hydrochlorothiazide tablets during pregnancy.

See “What is the most important information I should know about valsartan and hydrochlorothiazide tablets?” What are the possible side effects of valsartan and hydrochlorothiazide tablets? Valsartan and hydrochlorothiazide tablets may cause serious side effects including: Harm to an unborn baby causing injury and even death.

See “What is the most important information I should know about valsartan and hydrochlorothiazide tablets?” Low blood pressure (hypotension).

Low blood pressure is most likely to happen if you: take water pills are on a low salt diet get dialysis treatments have heart problems get sick with vomiting or diarrhea drink alcohol Lie down if you feel faint or dizzy.

Call your doctor right away.

Allergic reactions.

People with and without allergy problems or asthma who take valsartan and hydrochlorothiazide tablets may get allergic reactions.

Worsening of Lupus.

Hydrochlorothiazide, one of the medicines in valsartan and hydrochlorothiazide tablets may cause Lupus to become active or worse.

Fluid and electrolyte (salt) problems.

Tell your doctor about any of the following signs and symptoms of fluid and electrolyte problems: dry mouth thirst lack of energy (lethargic) weakness drowsiness restlessness confusion seizures muscle pain or cramps muscle fatigue very low urine output fast heartbeat nausea and vomiting Kidney problems.

Kidney problems may become worse in people that already have kidney disease.

Some people will have changes on blood tests for kidney function and may need a lower dose of valsartan and hydrochlorothiazide tablets.

Call your doctor if you get swelling in your feet, ankles, or hands, or unexplained weight gain.

If you have heart failure, your doctor should check your kidney function before prescribing valsartan and hydrochlorothiazide tablets.

Skin rash.

Call your doctor right away if you have an unusual skin rash.

Eye Problems.

One of the medicines in valsartan and hydrochlorothiazide tablets can cause eye problems that may lead to vision loss.

Symptoms of eye problems can happen within hours to weeks of starting valsartan and hydrochlorothiazide tablets.

Tell your doctor right away if you have: decrease in vision eye pain Other side effects were generally mild and brief.

They generally have not caused patients to stop taking valsartan and hydrochlorothiazide tablets.

Tell your doctor if you have any side effect that bothers you or that does not go away.

These are not all the possible side effects of valsartan and hydrochlorothiazide tablets.

For a complete list, ask your doctor or pharmacist.

Call your doctor for medical advice about side effects.

You may report side effects to FDA at 1-800-FDA-1088.

How do I store valsartan and hydrochlorothiazide tablets? Store valsartan and hydrochlorothiazide tablets at 20° to 25°C (68° to 77°F).

Keep valsartan and hydrochlorothiazide tablets in a closed container in a dry place.

Keep valsartan and hydrochlorothiazide tablets and all medicines out of the reach of children.

General information about valsartan and hydrochlorothiazide tablets Medicines are sometimes prescribed for conditions that are not mentioned in patient information leaflets.

Do not use valsartan and hydrochlorothiazide tablets for a condition for which it was not prescribed.

Do not give valsartan and hydrochlorothiazide tablets to other people, even if they have the same symptoms you have.

It may harm them.

This leaflet summarizes the most important information about valsartan and hydrochlorothiazide tablets.

If you would like more information, talk with your doctor.

You can ask your doctor or pharmacist for information about valsartan and hydrochlorothiazide tablets that is written for health professionals.

For more information about valsartan and hydrochlorothiazide tablets, call Mylan Pharmaceuticals Inc.

at 1-877-446-3679 (1-877-4-INFO-RX).

What are the ingredients in valsartan and hydrochlorothiazide tablets? Active ingredients: Valsartan and hydrochlorothiazide Inactive ingredients: colloidal silicon dioxide, crospovidone, hypromellose, lactose monohydrate, magnesium stearate, microcrystalline cellulose, polydextrose, polyethylene glycol, povidone, pregelatinized starch(corn), sodium lauryl sulfate, titanium dioxide and triacetin.

The 80 mg/12.5 mg, 160 mg/12.5 mg, 160 mg/25 mg and 320 mg/12.5 mg tablets also contain red iron oxide and yellow iron oxide.

The 320 mg/25 mg tablet also contains FD&C Blue No.

2 Aluminum Lake and FD&C Yellow No.

6 Aluminum Lake.

What is high blood pressure (hypertension)? Blood pressure is the force in your blood vessels when your heart beats and when your heart rests.

You have high blood pressure when the force is too much.

Valsartan and hydrochlorothiazide tablets can help your blood vessels relax and reduce the amount of water in your body so your blood pressure is lower.

Medicines that lower blood pressure lower your risk of having a stroke or heart attack.

High blood pressure makes the heart work harder to pump blood throughout the body and causes damage to the blood vessels.

If high blood pressure is not treated, it can lead to stroke, heart attack, heart failure, kidney failure, and vision problems.

* Registered trademarks are property of their respective owners.

Manufactured by: Mylan Pharmaceuticals Inc.

Morgantown, WV 26505 U.S.A.

Mylan Institutional Inc.

Distributed by: Rockford, IL 61103 U.S.A.

S-11364 R1 9/14

DOSAGE AND ADMINISTRATION

2 Dose once daily.

Titrate as needed to a maximum dose of 320 mg/25 mg ( 2 ) May be used as add-on/switch therapy for patients not adequately controlled on any of the components (valsartan or HCTZ) ( 2 ) May be substituted for titrated components ( 2.3 ) 2.1 General Considerations The usual starting dose is valsartan and hydrochlorothiazide tablets 160 mg/12.5 mg once daily.

The dosage can be increased after 1 to 2 weeks of therapy to a maximum of one 320 mg/25 mg tablet once daily as needed to control blood pressure [see Clinical Studies (14.2) ].

Maximum antihypertensive effects are attained within 2 to 4 weeks after a change in dose.

2.2 Add-On Therapy A patient whose blood pressure is not adequately controlled with valsartan (or another ARB) alone or hydrochlorothiazide alone may be switched to combination therapy with valsartan and hydrochlorothiazide tablets.

A patient who experiences dose-limiting adverse reactions on either component alone may be switched to valsartan and hydrochlorothiazide tablets containing a lower dose of that component in combination with the other to achieve similar blood pressure reductions.

The clinical response to valsartan and hydrochlorothiazide tablets should be subsequently evaluated and if blood pressure remains uncontrolled after 3 to 4 weeks of therapy, the dose may be titrated up to a maximum of 320 mg/25 mg.

2.3 Replacement Therapy Valsartan and hydrochlorothiazide tablets may be substituted for the titrated components.

2.4 Initial Therapy Valsartan and hydrochlorothiazide tablets are not recommended as initial therapy in patients with intravascular volume depletion [see Warnings and Precautions (5.2) ] .

2.5 Use with Other Antihypertensive Drugs Valsartan and hydrochlorothiazide tablets may be administered with other antihypertensive agents.

Atovaquone 250 MG / proguanil hydrochloride 100 MG Oral Tablet [Malarone]

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).]

Insulin Glargine 100 UNT/ML Injectable Solution [Lantus]

DRUG INTERACTIONS

A number of drugs affect glucose metabolism and may require insulin dose adjustment and particularly close monitoring.

The following are examples of drugs that may increase the blood-glucose-lowering effect of insulins including LANTUS and, therefore, increase the susceptibility to hypoglycemia: oral anti-diabetic products, pramlintide, angiotensin converting enzyme (ACE) inhibitors, disopyramide, fibrates, fluoxetine, monoamine oxidase inhibitors, propoxyphene, pentoxifylline, salicylates, somatostatin analogs, and sulfonamide antibiotics.

The following are examples of drugs that may reduce the blood-glucose-lowering effect of insulins including LANTUS: corticosteroids, niacin, danazol, diuretics, sympathomimetic agents (e.g., epinephrine, albuterol, terbutaline), glucagon, isoniazid, phenothiazine derivatives, somatropin, thyroid hormones, estrogens, progestogens (e.g., in oral contraceptives), protease inhibitors and atypical antipsychotic medications (e.g.

olanzapine and clozapine).

Beta-blockers, clonidine, lithium salts, and alcohol may either potentiate or weaken the blood-glucose-lowering effect of insulin.

Pentamidine may cause hypoglycemia, which may sometimes be followed by hyperglycemia.

The signs of hypoglycemia may be reduced or absent in patients taking sympatholytic drugs such as beta-blockers, clonidine, guanethidine, and reserpine.

OVERDOSAGE

An excess of insulin relative to food intake, energy expenditure, or both may lead to severe and sometimes prolonged and life-threatening hypoglycemia.

Mild episodes of hypoglycemia can usually be treated with oral carbohydrates.

Adjustments in drug dosage, meal patterns, or exercise may be needed.

More severe episodes of hypoglycemia with coma, seizure, or neurologic impairment may be treated with intramuscular/subcutaneous glucagon or concentrated intravenous glucose.

After apparent clinical recovery from hypoglycemia, continued observation and additional carbohydrate intake may be necessary to avoid recurrence of hypoglycemia.

DESCRIPTION

LANTUS (insulin glargine [rDNA origin] injection) is a sterile solution of insulin glargine for use as a subcutaneous injection.

Insulin glargine is a recombinant human insulin analog that is a long-acting (up to 24-hour duration of action), parenteral blood-glucose-lowering agent [ See Clinical Pharmacology (12) ].

LANTUS is produced by recombinant DNA technology utilizing a non-pathogenic laboratory strain of Escherichia coli (K12) as the production organism.

Insulin glargine differs from human insulin in that the amino acid asparagine at position A21 is replaced by glycine and two arginines are added to the C-terminus of the B-chain.

Chemically, insulin glargine is 21 A -Gly-30 B a-L-Arg-30 B b-L-Arg-human insulin and has the empirical formula C 267 H 404 N 72 O 78 S 6 and a molecular weight of 6063.

Insulin glargine has the following structural formula: LANTUS consists of insulin glargine dissolved in a clear aqueous fluid.

Each milliliter of LANTUS (insulin glargine injection) contains 100 Units (3.6378 mg) insulin glargine.

The 10 mL vial presentation contains the following inactive ingredients per mL: 30 mcg zinc, 2.7 mg m-cresol, 20 mg glycerol 85%, 20 mcg polysorbate 20, and water for injection.

The 3 mL cartridge presentation contains the following inactive ingredients per mL: 30 mcg zinc, 2.7 mg m-cresol, 20 mg glycerol 85%, and water for injection.

The pH is adjusted by addition of aqueous solutions of hydrochloric acid and sodium hydroxide.

LANTUS has a pH of approximately 4.

MM1

CLINICAL STUDIES

The safety and effectiveness of LANTUS given once-daily at bedtime was compared to that of once-daily and twice-daily NPH insulin in open-label, randomized, active-controlled, parallel studies of 2,327 adult patients and 349 pediatric patients with type 1 diabetes mellitus and 1,563 adult patients with type 2 diabetes mellitus (see Tables 8–11), whose primary efficacy aim was the assessment of effects on blood glucose, as measured by HbA1c.

In general, the reduction in glycated hemoglobin (HbA1c) with LANTUS was similar to that with NPH insulin.

The overall rates of hypoglycemia did not differ between patients with diabetes treated with LANTUS compared to NPH insulin [See Adverse Reactions (6.1) ] .

In the 12,537-participants, 6 to 7 year ORIGIN trial LANTUS was compared to standard glycemic management (largely non-insulin) in reducing cardiovascular (CV) risk as measured by CV outcome events.

Type 1 Diabetes–Adult (see Table 9 ).

In two clinical studies (Studies A and B), patients with type 1 diabetes (Study A; n=585, Study B; n=534) were randomized to 28 weeks of basal-bolus treatment with LANTUS or NPH insulin.

Regular human insulin was administered before each meal.

LANTUS was administered at bedtime.

NPH insulin was administered once daily at bedtime or in the morning and at bedtime when used twice daily.

In another clinical study (Study C), patients with type 1 diabetes (n=619) were randomized to 16 weeks of basal-bolus treatment with LANTUS or NPH insulin.

Insulin lispro was used before each meal.

LANTUS was administered once daily at bedtime and NPH insulin was administered once or twice daily.

In these 3 studies, LANTUS and NPH insulin had similar effects on HbA1c (Table 9) with a similar overall rate of hypoglycemia [See Adverse Reactions (6.1) ] .

Table 9: Type 1 Diabetes Mellitus–Adult Study A Study B Study C Treatment duration Treatment in combination with 28 weeks Regular insulin 28 weeks Regular insulin 16 weeks Insulin lispro LANTUS NPH LANTUS NPH LANTUS NPH Number of subjects treated 292 293 264 270 310 309 HbA1c Baseline HbA1c 8.0 8.0 7.7 7.7 7.6 7.7 Adj.

mean change from baseline +0.2 +0.1 -0.2 -0.2 -0.1 -0.1 LANTUS – NPH +0.1 +0.1 0.0 95% CI for Treatment difference (0.0; +0.2) (-0.1; +0.2) (-0.1; +0.1) Basal insulin dose Baseline mean 21 23 29 29 28 28 Mean change from baseline -2 0 -4 +2 -5 +1 Total insulin dose Baseline mean 48 52 50 51 50 50 Mean change from baseline -1 0 0 +4 -3 0 Fasting blood glucose (mg/dL) Baseline mean 167 166 166 175 175 173 Adj.

mean change from baseline -21 -16 -20 -17 -29 -12 Body weight (kg) Baseline mean 73.2 74.8 75.5 75.0 74.8 75.6 Mean change from baseline 0.1 -0.0 0.7 1.0 0.1 0.5 Type 1 Diabetes–Pediatric (see Table 10 ).

In a randomized, controlled clinical study (Study D), pediatric patients (age range 6 to 15 years) with type 1 diabetes (n=349) were treated for 28 weeks with a basal-bolus insulin regimen where regular human insulin was used before each meal.

LANTUS was administered once daily at bedtime and NPH insulin was administered once or twice daily.

Similar effects on HbA1c (Table 10) and the incidence of hypoglycemia were observed in both treatment groups [See Adverse Reactions (6.1) ] .

Table 10: Type 1 Diabetes Mellitus–Pediatric Study D Treatment duration 28 weeks Treatment in combination with Regular insulin LANTUS NPH Number of subjects treated 174 175 HbA1c Baseline mean 8.5 8.8 Adj.

mean change from baseline +0.3 +0.3 LANTUS – NPH 0.0 95% CI for Treatment difference (-0.2; +0.3) Basal insulin dose Baseline mean 19 19 Mean change from baseline -1 +2 Total insulin dose Baseline mean 43 43 Mean change from baseline +2 +3 Fasting blood glucose (mg/dL) Baseline mean 194 191 Adj.

mean change from baseline -23 -12 Body weight (kg) Baseline mean 45.5 44.6 Mean change from baseline 2.2 2.5 Type 2 Diabetes–Adult (see Table 11 ).

In a randomized, controlled clinical study (Study E) (n=570), LANTUS was evaluated for 52 weeks in combination with oral anti-diabetic medications (a sulfonylurea, metformin, acarbose, or combinations of these drugs).

LANTUS administered once daily at bedtime was as effective as NPH insulin administered once daily at bedtime in reducing HbA1c and fasting glucose (Table 11).

The rate of hypoglycemia was similar in LANTUS and NPH insulin treated patients [See Adverse Reactions (6.1) ] .

In a randomized, controlled clinical study (Study F), in patients with type 2 diabetes not using oral anti-diabetic medications (n=518), a basal-bolus regimen of LANTUS once daily at bedtime or NPH insulin administered once or twice daily was evaluated for 28 weeks.

Regular human insulin was used before meals, as needed.

LANTUS had similar effectiveness as either once- or twice-daily NPH insulin in reducing HbA1c and fasting glucose (Table 11) with a similar incidence of hypoglycemia [See Adverse Reactions (6.1) ] .

In a randomized, controlled clinical study (Study G), patients with type 2 diabetes were randomized to 5 years of treatment with once-daily LANTUS or twice-daily NPH insulin.

For patients not previously treated with insulin, the starting dose of LANTUS or NPH insulin was 10 units daily.

Patients who were already treated with NPH insulin either continued on the same total daily NPH insulin dose or started LANTUS at a dose that was 80% of the total previous NPH insulin dose.

The primary endpoint for this study was a comparison of the progression of diabetic retinopathy by 3 or more steps on the Early Treatment Diabetic Retinopathy Study (ETDRS) scale.

HbA1c change from baseline was a secondary endpoint.

Similar glycemic control in the 2 treatment groups was desired in order to not confound the interpretation of the retinal data.

Patients or study personnel used an algorithm to adjust the LANTUS and NPH insulin doses to a target fasting plasma glucose ≤100 mg/dL.

After the LANTUS or NPH insulin dose was adjusted, other anti-diabetic agents, including pre-meal insulin were to be adjusted or added.

The LANTUS group had a smaller mean reduction from baseline in HbA1c compared to the NPH insulin group, which may be explained by the lower daily basal insulin doses in the LANTUS group (Table 11).

Both treatment groups had a similar incidence of reported symptomatic hypoglycemia.

The incidences of severe symptomatic hypoglycemia are given in Table 6 [See Adverse Reactions (6.1) ] .

Table 11: Type 2 Diabetes Mellitus–Adult Study E Study F Study G Treatment duration 52 weeks 28 weeks 5 years Treatment in combination with Oral agents Regular insulin Regular insulin LANTUS NPH LANTUS NPH LANTUS NPH Number of subjects treated 289 281 259 259 513 504 HbA1c Baseline mean 9.0 8.9 8.6 8.5 8.4 8.3 Adj.

mean change from baseline -0.5 -0.4 -0.4 -0.6 -0.6 -0.8 LANTUS – NPH -0.1 +0.2 +0.2 95% CI for Treatment difference (-0.3; +0.1) (0.0; +0.4) (+0.1, +0.4) Basal insulin dose * Baseline mean 14 15 44.1 45.5 39 44 Mean change from baseline +12 +9 -1 +7 +23 +30 Total insulin dose Baseline mean 14 15 64 67 48 53 Mean change from baseline +12 +9 +10 +13 +41 +40 Fasting blood glucose (mg/dL) Baseline mean 179 180 164 166 190 180 Adj.

mean change from baseline -49 -46 -24 -22 -45 -44 Body weight (kg) Baseline mean 83.5 82.1 89.6 90.7 100 99 Adj.

mean change from baseline 2.0 1.9 0.4 1.4 3.7 4.8 In Study G, the baseline dose of basal or total insulin was the first available on-treatment dose prescribed during the study (on visit month 1.5).

The ORIGIN (Outcome Reduction with Initial Glargine INtervention) trial was a, international, multicenter, randomized, 2×2 factorial design study conducted in 12,537 participants with impaired fasting glucose (IFG), impaired glucose tolerance (IGT) or early type 2 diabetes mellitus and evidence of CV disease.

Participants were randomized to receive LANTUS (n=6264), titrated to a FPG of 95 mg/dL (5.3mM) or less, or Standard Care (n=6273).

At baseline participants had a mean age of 63.5 years, mean duration of diabetes of 5.8 years in those with pre-existing diabetes, and median HbA1c of 6.4%.

Median duration of follow-up was approximately 6.2 years.

At the end of the trial 81% of participants randomized to take LANTUS were still on treatment.

Median on-treatment HbA1c values ranged from 5.9 to 6.4 % in the LANTUS group, and 6.2% to 6.6% in the Standard Care group throughout the duration of follow-up.

Median FPG in the LANTUS group was at target (≤95mg/dL) following dose titration for the duration of the study.

The incidences of severe symptomatic hypoglycemia are given in Table 7 [See Adverse Reactions (6.1) ] .

The median of the change in body weight from baseline to the last on-treatment visit was 2.2 kg greater in the Lantus group than in the Standard Care group.

The primary objective of this trial was to examine the effect of LANTUS on two co-primary composite efficacy outcomes.

The first one was the time to the first occurrence of CV death, nonfatal myocardial infarction (MI), or nonfatal stroke, and the second one was the time to the first occurrence of any of the first co-primary events, or revascularization procedure (cardiac, carotid, or peripheral), or hospitalization for heart failure.

Secondary endpoints were: all-cause mortality a composite microvascular outcome development of type 2 diabetes, in participants with IGT and/or IFG at baseline The primary and secondary outcome results, as well as the results for each component of the coprimary outcomes, are displayed in the two tables (table 12 for the time-to-event analyses, and, for the non-time-to-event analysis of development of diabetes, table 13) below.

Table 12: ORIGIN: Time to Onset of each Primary and Secondary Endpoint LANTUS N=6264 Standard care N=6273 Lantus vs Standard care Participants with Events N (%) Participants with Events N (%) Hazard Ratio (95% CI) Primary endpoints CV death, nonfatal myocardial infarction (MI), or nonfatal stroke 1041 (16.6) 1013 (16.1) 1.02 (0.94, 1.11) CV death, nonfatal myocardial infarction (MI), or nonfatal stroke, or hospitalization for heart failure or revascularization procedure 1792 (28.6) 1727 (27.5) 1.04 (0.97, 1.11) Secondary endpoints All-cause mortality 951 (15.2) 965 (15.4) 0.98 (0.90, 1.08) Composite microvascular outcome * 1323 (21.1) 1363 (21.7) 0.97 (0.90, 1.05) Components of coprimary endpoint CV death 580 (9.3) 576 (9.2) 1.00 (0.89, 1.13) MI (fatal or non-fatal) 336 (5.4) 326 (5.2) 1.03 (0.88, 1.19) stroke(fatal or non-fatal) 331 (5.3) 319 (5.1) 1.03 (0.89, 1.21) Revascularizations 908 (14.5) 860 (13.7) 1.06 (0.96, 1.16) Hospitalization for heart failure 310 (4.9) 343 (5.5) 0.90 (0.77, 1.05) with components of: laser photocoagulation or vitrectomy or blindness for diabetic retinopathy; progression in albuminuria; or doubling of serum creatinine or development of the need for renal replacement therapy Table 13: Incidence Rate of Diabetes by end of study OGTT * Treatment (N) LANTUS (6264) Standard Care (6273) Number of Participants † 737 719 # participants who developed diabetes (%) 182 (24.7) 224 (31.2) Odds Ratio (95% CI) 0.72 (0.58 to 0.91) End of study OGTT was performed 3–4 weeks after discontinuing LANTUS Participants with prediabetes (IFG or IGT) at baseline, based on an OGTT performed then; There were no statistical significant differences between treatment groups in the overall incidence of cancer (all types combined) or death from cancer.

The time to first event of any cancer or new cancer during the study was similar between the two treatment groups with respective hazard ratios of 0.99 (0.88, 1.11) and 0.96 (0.85, 1.09).

Participation in ORIGIN for a median of approximately 6.2 years showed that treatment with Lantus did not alter the risk for cardiovascular outcomes, all-cause mortality or cancer, when compared to standard glucose lowering therapy.

In addition, metabolic control was maintained at a lower level of glycemia, with a decrease in the percentage of participants developing diabetes, at a cost of a modest increase in hypoglycemia and weight gain.

LANTUS Timing of Daily Dosing (see Table 14 ).

The safety and efficacy of LANTUS administered pre-breakfast, pre-dinner, or at bedtime were evaluated in a randomized, controlled clinical study in patients with type 1 diabetes (study H, n=378).

Patients were also treated with insulin lispro at mealtime.

LANTUS administered at different times of the day resulted in similar reductions in HbA1c compared to that with bedtime administration (see Table 11 ).

In these patients, data are available from 8-point home glucose monitoring.

The maximum mean blood glucose was observed just prior to injection of LANTUS regardless of time of administration.

In this study, 5% of patients in the LANTUS-breakfast arm discontinued treatment because of lack of efficacy.

No patients in the other two arms discontinued for this reason.

The safety and efficacy of LANTUS administered pre-breakfast or at bedtime were also evaluated in a randomized, active-controlled clinical study (Study I, n=697) in patients with type 2 diabetes not adequately controlled on oral anti-diabetic therapy.

All patients in this study also received glimepiride 3 mg daily.

LANTUS given before breakfast was at least as effective in lowering HbA1c as LANTUS given at bedtime or NPH insulin given at bedtime (see Table 12 ).

Table 14: LANTUS Timing of Daily Dosing in Type 1 (Study H) and Type 2 (Study I) Diabetes Mellitus Study H Study I Treatment duration 24 weeks 24 weeks Treatment in combination with: Insulin lispro Glimepiride LANTUS Breakfast LANTUS Dinner LANTUS Bedtime LANTUS Breakfast LANTUS Bedtime NPH Bedtime Number of subjects treated * 112 124 128 234 226 227 HbA1c Baseline mean 7.6 7.5 7.6 9.1 9.1 9.1 Mean change from baseline -0.2 -0.1 0.0 -1.3 -1.0 -0.8 Basal insulin dose (U) Baseline mean 22 23 21 19 20 19 Mean change from baseline 5 2 2 11 18 18 Total insulin dose (U) NA † NA NA Baseline mean 52 52 49 Mean change from baseline 2 3 2 Body weight (kg) Baseline mean 77.1 77.8 74.5 80.7 82 81 Mean change from baseline 0.7 0.1 0.4 3.9 3.7 2.9

HOW SUPPLIED

LANTUS solution for injection 100 units per mL (U-100) is available as: Dosage Unit/Strength Package size NDC # 0088 10 mL vials 100 Units/mL Pack of 1 2220-33 3 mL cartridge system * 100 Units/mL package of 5 2220-52 3 mL SoloStar ® disposable insulin device 100 Units/mL package of 5 2219-05 Cartridge systems are for use only in OptiClik Needles are not included in the packs.

BD Ultra-Fine™ needles 1 to be used in conjunction with SoloStar and OptiClik are sold separately and are manufactured by BD.

The brands listed are the registered trademarks of their respective owners and are not trademarks of sanofi-aventis U.S.

LLC LANTUS should not be stored in the freezer and should not be allowed to freeze.

Discard LANTUS if it has been frozen.

Unopened Vial/Cartridge system/SoloStar disposable insulin device: Unopened LANTUS vials, cartridge systems and SoloStar device should be stored in a refrigerator, 36°F – 46°F (2°C – 8°C).

Discard after the expiration date.

Open (In-Use) Vial: Vials must be discarded 28 days after being opened.

If refrigeration is not possible, the open vial can be kept unrefrigerated for up to 28 days away from direct heat and light, as long as the temperature is not greater than 86°F (30°C).

Open (In-Use) Cartridge system: The opened (in-use) cartridge system in OptiClik should NOT be refrigerated but should be kept at room temperature (below 86°F [30°C]) away from direct heat and light.

The opened (in-use) cartridge system in OptiClik must be discarded 28 days after being opened.

Do not store OptiClik , with or without cartridge system, in a refrigerator at any time.

Open (In-Use) SoloStar disposable insulin device: The opened (in-use) SoloStar should NOT be refrigerated but should be kept at room temperature (below 86°F [30°C]) away from direct heat and light.

The opened (in-use) SoloStar device must be discarded 28 days after being opened.

These storage conditions are summarized in the following table: Not in-use (unopened) Refrigerated Not in-use (unopened) Room Temperature In-use (opened) (See Temperature Below) 10 mL Vial Until expiration date 28 days 28 days Refrigerated or room temperature 3 mL Cartridge system Until expiration date 28 days 28 days Refrigerated or room temperature 3 mL Cartridge system inserted into OptiClik ® 28 days Room temperature only (Do not refrigerate) 3 mL SoloStar ® disposable insulin device Until expiration date 28 days 28 days Room temperature only (Do not refrigerate) Parenteral drug products should be inspected visually prior to administration whenever the solution and the container permit.

LANTUS must only be used if the solution is clear and colorless with no particles visible.

Mixing and diluting: LANTUS must NOT be diluted or mixed with any other insulin or solution [S ee Warnings and Precautions (5.2) ] .

Vial: The syringes must not contain any other medicinal product or residue.

Cartridge system/SoloStar : If OptiClik, the Insulin Delivery Device used with the LANTUS cartridge system, or SoloStar disposable insulin device, malfunctions, LANTUS may be drawn from the cartridge system or from SoloStar into a U-100 syringe and injected.

DOSAGE FORMS AND STRENGTHS

DOSAGE FORMS & STRENGTHS LANTUS solution for injection 100 Units per mL is available as: -10 mL Vial (1000 Units/10 mL)-3 mL Cartridge systems for use only in OptiClik ® (300 Units/3 mL)-3 mL SoloStar ® disposable insulin device (300 Units/3 mL)

INDICATIONS AND USAGE

INDICATIONS & USAGE LANTUS is indicated to improve glycemic control in adults and children with type 1 diabetes mellitus and in adults with type 2 diabetes mellitus.

Important Limitations of Use: LANTUS is not recommended for the treatment of diabetic ketoacidosis.

Intravenous short-acting insulin is the preferred treatment for this condition.

WARNING AND CAUTIONS

WARNINGS AND PRECAUTIONS Dose adjustment and monitoring: Monitor blood glucose in all patients treated with insulin.

Insulin regimens should be modified cautiously and only under medical supervision ( 5.1 ) Administration: Do not dilute or mix with any other insulin or solution.

Do not administer subcutaneously via an insulin pump or intravenously because severe hypoglycemia can occur ( 5.2 ) Do not share reusable or disposable insulin devices or needles between patients ( 5.2 ) Hypoglycemia: Most common adverse reaction of insulin therapy and may be life-threatening ( 5.3 , 6.1 ) Allergic reactions: Severe, life-threatening, generalized allergy, including anaphylaxis, can occur ( 5.4 , 6.1 ) Renal or hepatic impairment: May require a reduction in the LANTUS dose ( 5.5 , 5.6 ) Glucose monitoring is essential for all patients receiving insulin therapy.

Changes to an insulin regimen should be made cautiously and only under medical supervision.

Changes in insulin strength, manufacturer, type, or method of administration may result in the need for a change in insulin dose or an adjustment in concomitant oral anti-diabetic treatment.

As with all insulin preparations, the time course of action for LANTUS may vary in different individuals or at different times in the same individual and is dependent on many conditions, including the local blood supply, local temperature, and physical activity.

Do not administer LANTUS intravenously or via an insulin pump.

The intended duration of activity of LANTUS is dependent on injection into subcutaneous tissue Intravenous administration of the usual subcutaneous dose could result in severe hypoglycemia [see Warnings and Precautions (5.3) ] .

Do not dilute or mix LANTUS with any other insulin or solution.

If LANTUS is diluted or mixed, the solution may become cloudy, and the pharmacokinetic or pharmacodynamic profile (e.g., onset of action, time to peak effect) of LANTUS and the mixed insulin may be altered in an unpredictable manner.

When LANTUS and regular human insulin were mixed immediately before injection in dogs, a delayed onset of action and a delayed time to maximum effect for regular human insulin was observed.

The total bioavailability of the mixture was also slightly decreased compared to separate injections of LANTUS and regular human insulin.

The relevance of these observations in dogs to humans is unknown.

Do not share disposable or reusable insulin devices or needles between patients, because doing so carries a risk for transmission of blood-borne pathogens.

Hypoglycemia is the most common adverse reaction of insulin, including LANTUS.

The risk of hypoglycemia increases with intensive glycemic control.

Patients must be educated to recognize and manage hypoglycemia.

Severe hypoglycemia can lead to unconsciousness or convulsions and may result in temporary or permanent impairment of brain function or death.

Severe hypoglycemia requiring the assistance of another person or parenteral glucose infusion or glucagon administration has been observed in clinical trials with insulin, including trials with LANTUS.

The timing of hypoglycemia usually reflects the time-action profile of the administered insulin formulations.

Other factors such as changes in food intake (e.g., amount of food or timing of meals), exercise, and concomitant medications may also alter the risk of hypoglycemia [ See Drug Interactions (7) ].

The prolonged effect of subcutaneous LANTUS may delay recovery from hypoglycemia.

Patients being switched from twice daily NPH insulin to once-daily LANTUS should have their initial LANTUS dose reduced by 20% from the previous total daily NPH dose to reduce the risk of hypoglycemia [see Dosage and Administration (2.3) ].

As with all insulins, use caution in patients with hypoglycemia unawareness and in patients who may be predisposed to hypoglycemia (e.g., the pediatric population and patients who fast or have erratic food intake).

The patient’s ability to concentrate and react may be impaired as a result of hypoglycemia.

This may present a risk in situations where these abilities are especially important, such as driving or operating other machinery.

Early warning symptoms of hypoglycemia may be different or less pronounced under certain conditions, such as longstanding diabetes, diabetic neuropathy, use of medications such as beta-blockers, or intensified glycemic control.

These situations may result in severe hypoglycemia (and, possibly, loss of consciousness) prior to the patient’s awareness of hypoglycemia.

Severe, life-threatening, generalized allergy, including anaphylaxis, can occur with insulin products, including LANTUS.

Due to its long duration of action, Lantus is not recommended during periods of rapidly declining renal function because of the risk for prolonged hypoglycemia.

Although studies have not been performed in patients with diabetes and renal impairment, a reduction in the LANTUS dose may be required in patients with renal impairment because of reduced insulin metabolism, similar to observations found with other insulins.

[See Clinical Pharmacology (12.3) ].

Due to its long duration of action, Lantus is not recommended during periods of rapidly declining hepatic function because of the risk for prolonged hypoglycemia.

Although studies have not been performed in patients with diabetes and hepatic impairment, a reduction in the LANTUS dose may be required in patients with hepatic impairment because of reduced capacity for gluconeogenesis and reduced insulin metabolism, similar to observations found with other insulins.

[See Clinical Pharmacology (12.3) ].

Some medications may alter insulin requirements and subsequently increase the risk for hypoglycemia or hyperglycemia [See Drug Interactions (7) ].

INFORMATION FOR PATIENTS

Patients should be informed that changes to insulin regimens must be made cautiously and only under medical supervision.

Patients should be informed about the potential side effects of insulin therapy, including lipodystrophy (and the need to rotate injection sites within the same body region), weight gain, allergic reactions, and hypoglycemia.

Patients should be informed that the ability to concentrate and react may be impaired as a result of hypoglycemia.

This may present a risk in situations where these abilities are especially important, such as driving or operating other machinery.

Patients who have frequent hypoglycemia or reduced or absent warning signs of hypoglycemia should be advised to use caution when driving or operating machinery.

Accidental mix-ups between LANTUS and other insulins, particularly short-acting insulins, have been reported.

To avoid medication errors between LANTUS and other insulins, patients should be instructed to always check the insulin label before each injection.

LANTUS must only be used if the solution is clear and colorless with no particles visible .

Patients must be advised that LANTUS must NOT be diluted or mixed with any other insulin or solution .

Patients should be advised not to share disposable or reusable insulin devices or needles with other patients, because doing so carries a risk for transmission of blood-borne pathogens.

Patients should be instructed on self-management procedures including glucose monitoring, proper injection technique, and management of hypoglycemia and hyperglycemia.

Patients must be instructed on handling of special situations such as intercurrent conditions (illness, stress, or emotional disturbances), an inadequate or skipped insulin dose, inadvertent administration of an increased insulin dose, inadequate food intake, and skipped meals.

Patients with diabetes should be advised to inform their health care professional if they are pregnant or are contemplating pregnancy.

Refer patients to the LANTUS “Patient Information” for additional information.

See attached document at end of Full Prescribing Information.

DOSAGE AND ADMINISTRATION

DOSAGE & ADMINISTRATION The starting dose should be individualized based on the type of diabetes and whether the patient is insulin-naïve ( 2.1 , 2.2 , 2.3 ) Administer subcutaneously once daily at any time of day, but at the same time every day.

( 2.1 ) Rotate injection sites within an injection area (abdomen, thigh, or deltoid) to reduce the risk of lipodystrophy.

( 2.1 ) Converting from other insulin therapies may require adjustment of timing and dose of LANTUS.

Closely monitor glucoses especially upon converting to LANTUS and during the initial weeks thereafter.

( 2.3 ) LANTUS is a recombinant human insulin analog for once daily subcutaneous administration with potency that is approximately the same as the potency of human insulin.

LANTUS exhibits a relatively constant glucose-lowering profile over 24 hours that permits once-daily dosing.

LANTUS may be administered at any time during the day.

LANTUS should be administered subcutaneously once a day at the same time every day.

The dose of LANTUS must be individualized based on clinical response.

Blood glucose monitoring is essential in all patients receiving insulin therapy.

Patients adjusting the amount or timing of dosing with LANTUS, should only do so under medical supervision with appropriate glucose monitoring [see Warnings and Precautions (5.1) .] In patients with type 1 diabetes, LANTUS must be used in regimens with short-acting insulin.

The intended duration of activity of LANTUS is dependent on injection into subcutaneous tissue [see Clinical pharmacology (12.2) ] .

LANTUS should not be administered intravenously or via an insulin pump.

Intravenous administration of the usual subcutaneous dose could result in severe hypoglycemia [see Warnings and Precautions (5.3) ] .

As with all insulins, injection sites should be rotated within the same region (abdomen, thigh, or deltoid) from one injection to the next to reduce the risk of lipodystrophy [See Adverse Reactions (6.1) ].

In clinical studies, there was no clinically relevant difference in insulin glargine absorption after abdominal, deltoid, or thigh subcutaneous administration.

As for all insulins, the rate of absorption, and consequently the onset and duration of action, may be affected by exercise and other variables, such as stress, intercurrent illness, or changes in co-administered drugs or meal patterns .

The recommended starting dose of LANTUS in patients with type 1 diabetes should be approximately one-third of the total daily insulin requirements.

Short-acting, premeal insulin should be used to satisfy the remainder of the daily insulin requirements.

The recommended starting dose of LANTUS in patients with type 2 diabetes who are not currently treated with insulin is 10 units (or 0.2 Units/kg) once daily, which should subsequently be adjusted to the patient’s needs.

The dose of LANTUS should be adjusted according to blood glucose measurements.

The dosage of LANTUS should be individualized under the supervision of a healthcare provider in accordance with the needs of the patient.

If changing from a treatment regimen with an intermediate- or long-acting insulin to a regimen with LANTUS, the amount and timing of shorter-acting insulins and doses of any oral anti-diabetic drugs may need to be adjusted.

If transferring patients from once-daily NPH insulin to once-daily LANTUS, the recommended initial LANTUS dose is the same as the dose of NPH that is being discontinued.

If transferring patients from twice-daily NPH insulin to once-daily LANTUS, the recommended initial LANTUS dose is 80% of the total NPH dose that is being discontinued.

This dose reduction will lower the likelihood of hypoglycemia [ see Warnings and Precautions (5.3) ].

nifedipine 90 MG Osmotic 24 HR Extended Release Oral Tablet

WARNINGS

Excessive Hypotension Although in most angina patients the hypotensive effect of nifedipine is modest and well tolerated, occasional patients have had excessive and poorly tolerated hypotension.

These responses have usually occurred during initial titration or at the time of subsequent upward dosage adjustment, and may be more likely in patients on concomitant beta-blockers.

Severe hypotension and/or increased fluid volume requirements have been reported in patients receiving nifedipine together with a beta-blocking agent who underwent coronary artery bypass surgery using high dose fentanyl anesthesia.

The interaction with high dose fentanyl appears to be due to the combination of nifedipine and a beta-blocker, but the possibility that it may occur with nifedipine alone, with low doses of fentanyl, in other surgical procedures, or with other narcotic analgesics cannot be ruled out.

In nifedipine-treated patients where surgery using high dose fentanyl anesthesia is contemplated, the physician should be aware of these potential problems and if the patient’s condition permits, sufficient time (at least 36 hours) should be allowed for nifedipine to be washed out of the body prior to surgery.

The following information should be taken into account in those patients who are being treated for hypertension as well as angina: Increased Angina and/or Myocardial Infarction Rarely, patients, particularly those who have severe obstructive coronary artery disease, have developed well documented increased frequency, duration and/or severity of angina or acute myocardial infarction on starting nifedipine or at the time of dosage increase.

The mechanism of this effect is not established.

Beta-Blocker Withdrawal It is important to taper beta-blockers if possible, rather than stopping them abruptly before beginning nifedipine.

Patients recently withdrawn from beta-blockers may develop a withdrawal syndrome with increased angina, probably related to increased sensitivity to catecholamines.

Initiation of nifedipine treatment will not prevent this occurrence and on occasion has been reported to increase it.

Congestive Heart Failure Rarely, patients, usually receiving a beta-blocker, have developed heart failure after beginning nifedipine.

Patients with tight aortic stenosis may be at greater risk for such an event, as the unloading effect of nifedipine would be expected to be of less benefit to those patients, owing to their fixed impedance to flow across the aortic valve in these patients.

Gastointestinal Obstruction Requiring Surgery There have been rare reports of obstructive symptoms in patients with known strictures in association with the ingestion of Nifedipine Extended-release Tablets.

Bezoars can occur in very rare cases and may require surgical intervention.

Cases of serious gastrointestinal obstruction have been identified in patients with no known gastrointestinal disease, including the need for hospitalization and surgical intervention.

Risk factors for a gastrointestinal obstruction identified from post-marketing reports of Nifedipine Extended-release Tablets include alteration in gastrointestinal anatomy (e.g., severe gastrointestinal narrowing, colon cancer, small bowel obstruction, bowel resection, gastric bypass, vertical banded gastroplasty, colostomy, diverticulitis, diverticulosis, and inflammatory bowel disease), hypomotility disorders (e.g., constipation, gastroesophageal reflux disease, ileus, obesity, hypothyroidism, and diabetes) and concomitant medications (e.g., H2-histamine blockers, opiates, nonsteroidal anti-inflammatory drugs, laxatives, anticholinergic agents, levothyroxine, and neuromuscular blocking agents).

Gastrointestinal Ulcers Cases of tablet adherence to the gastrointestinal wall with ulceration have been reported, some requiring hospitalization and intervention.

DRUG INTERACTIONS

Drug Interactions Beta-adrenergic blocking agents (See INDICATIONS AND USAGE and WARNINGS .) Experience in over 1400 patients with nifedipine capsules in a noncomparative clinical trial has shown that concomitant administration of nifedipine and beta-blocking agents is usually well tolerated, but there have been occasional literature reports suggesting that the combination may increase the likelihood of congestive heart failure, severe hypotension, or exacerbation of angina.

Long-acting Nitrates Nifedipine may be safely coadministered with nitrates, but there have been no controlled studies to evaluate the antianginal effectiveness of this combination.

Digitalis Administration of nifedipine with digoxin increased digoxin levels in nine of twelve normal volunteers.

The average increase was 45%.

Another investigator found no increase in digoxin levels in thirteen patients with coronary artery disease.

In an uncontrolled study of over two hundred patients with congestive heart failure during which digoxin blood levels were not measured, digitalis toxicity was not observed.

Since there have been isolated reports of patients with elevated digoxin levels, it is recommended that digoxin levels be monitored when initiating, adjusting, and discontinuing nifedipine to avoid possible over- or under-digitalization.

Coumarin Anticoagulants There have been rare reports of increased prothrombin time in patients taking coumarin anticoagulants to whom nifedipine was administered.

However, the relationship to nifedipine therapy is uncertain.

Cimetidine A study in six healthy volunteers has shown a significant increase in peak nifedipine plasma levels (80%) and area-under-the-curve (74%), after a one week course of cimetidine at 1000 mg per day and nifedipine at 40 mg per day.

Ranitidine produced smaller, non-significant increases.

The effect may be mediated by the known inhibition of cimetidine on hepatic cytochrome P-450, the enzyme system probably responsible for the first-pass metabolism of nifedipine.

If nifedipine therapy is initiated in a patient currently receiving cimetidine, cautious titration is advised.

OVERDOSAGE

Experience with nifedipine overdosage is limited.

Generally, overdosage with nifedipine leading to pronounced hypotension calls for active cardiovascular support including monitoring of cardiovascular and respiratory function, elevation of extremities, judicious use of calcium infusion, pressor agents and fluids.

Clearance of nifedipine would be expected to be prolonged in patients with impaired liver function.

Since nifedipine is highly protein-bound, dialysis is not likely to be of any benefit.

There has been one reported case of massive overdosage with Nifedipine Extended-release Tablets.

The main effects of ingestion of approximately 4800 mg of Nifedipine Extended-release Tablets in a young man attempting suicide as a result of cocaine-induced depression was initial dizziness, palpitations, flushing, and nervousness.

Within several hours of ingestion, nausea, vomiting, and generalized edema developed.

No significant hypotension was apparent at presentation, 18 hours post-ingestion.

Electrolyte abnormalities consisted of a mild, transient elevation of serum creatinine, and modest elevations of LDH and CPK, but normal SGOT.

Vital signs remained stable, no electrocardiographic abnormalities were noted and renal function returned to normal within 24 to 48 hours with routine supportive measures alone.

No prolonged sequelae were observed.

The effect of a single 900 mg ingestion of nifedipine capsules in a depressed anginal patient also on tricyclic antidepressants was loss of consciousness within 30 minutes of ingestion, and profound hypotension, which responded to calcium infusion, pressor agents, and fluid replacement.

A variety of ECG abnormalities were seen in this patient with a history of bundle branch block, including sinus bradycardia and varying degrees of AV block.

These dictated the prophylactic placement of a temporary ventricular pacemaker, but otherwise resolved spontaneously.

Significant hyperglycemia was seen initially in this patient, but plasma glucose levels rapidly normalized without further treatment.

A young hypertensive patient with advanced renal failure ingested 280 mg of nifedipine capsules at one time, with resulting marked hypotension responding to calcium infusion and fluids.

No AV conduction abnormalities, arrhythmias, or pronounced changes in heart rate were noted, nor was there any further deterioration in renal function.

DESCRIPTION

Nifedipine is a drug belonging to a class of pharmacological agents known as the calcium channel blockers.

Nifedipine is 3, 5-pyridinedicarboxylic acid, 1, 4-dihydro-2, 6-dimethyl-4-(2-nitrophenyl)-, dimethyl ester, C 17 H 18 N 2 O 6, and has the structural formula: Nifedipine is a yellow crystalline substance, practically insoluble in water but soluble in ethanol.

It has a molecular weight of 346.33.

Nifedipine Extended-release Tablet is formulated as a once-a-day extended-release tablet for oral administration designed to deliver 30, 60, or 90 mg of nifedipine.

Inert ingredients in the formulations are: black iron oxide; cellulose acetate; colloidal silicon dioxide; hypromellose; lactose monohydrate; magnesium stearate; microcrystalline cellulose; polyethylene glycol; polyethylene oxide; polysorbate; povidone; propylene glycol; red ferric oxide; sodium chloride; titanium dioxide; triacetin.

Chemical Structure System Components and Performance Nifedipine Extended-release Tablet is similar in appearance to a conventional tablet.

It consists, however, of a semipermeable membrane surrounding an osmotically active drug core.

As water from the gastrointestinal tract enters the tablet, pressure increases in the core of the tablet, releasing drug through the precision laser-drilled tablet orifice in the one side of the tablet.

Nifedipine Extended-release Tablet is designed to provide nifedipine at an approximately constant rate over 24 hours.

This controlled rate of drug delivery into the gastrointestinal lumen is independent of pH or gastrointestinal motility.

Nifedipine Extended-release Tablet depends for its action on the existence of an osmotic gradient between the contents of the tablet core and fluid in the GI tract.

Drug delivery is essentially constant as long as the osmotic gradient remains constant, and then gradually falls to zero.

Upon swallowing, the biologically inert components of the tablet remain intact during GI transit and are eliminated in the feces as an insoluble shell.

USP Drug Release Test 5.

HOW SUPPLIED

Nifedipine Extended-release Tablets 90 mg are round, biconvex, pink coated tablets imprinted with “KU 262 in black ink.

They are supplied as follows: Bottles of 30 Tablets NDC 33261-0840-30 Store at 20°-25°C (68°-77°F) (See USP Controlled Room Temperature).

Protect from moisture, humidity, and light.

For Medical Information Contact: Medical Affairs Department Phone: (800) 477-7877 Distributed by: Kremers, Urban Pharmaceuticals, Inc.

Princeton, NJ 08540, USA for: logo Wilmington, NC 28405, USA L4759N Repackaged By : Aidarex Pharmaceuticals LLC, Corona, CA 92880 Rev.6E 10/2011 logo

MECHANISM OF ACTION

Mechanism of Action A) Angina The precise mechanisms by which inhibition of calcium influx relieves angina has not been fully determined, but includes at least the following two mechanisms: 1) Relaxation and Prevention of Coronary Artery Spasm Nifedipine dilates the main coronary arteries and coronary arterioles, both in normal and ischemic regions, and is a potent inhibitor of coronary artery spasm, whether spontaneous or ergonovine-induced.

This property increases myocardial oxygen delivery in patients with coronary artery spasm, and is responsible for the effectiveness of nifedipine in vasospastic (Prinzmetal’s or variant) angina.

Whether this effect plays any role in classical angina is not clear, but studies of exercise tolerance have not shown an increase in the maximum exercise rate-pressure product, a widely accepted measure of oxygen utilization.

This suggests that, in general, relief of spasm or dilation of coronary arteries is not an important factor in classical angina.

2) Reduction of Oxygen Utilization Nifedipine regularly reduces arterial pressure at rest and at a given level of exercise by dilating peripheral arterioles and reducing the total peripheral vascular resistance (afterload) against which the heart works.

This unloading of the heart reduces myocardial energy consumption and oxygen requirements, and probably accounts for the effectiveness of nifedipine in chronic stable angina.

B) Hypertension The mechanism by which nifedipine reduces arterial blood pressure involves peripheral arterial vasodilatation and the resulting reduction in peripheral vascular resistance.

The increased peripheral vascular resistance that is an underlying cause of hypertension results from an increase in active tension in the vascular smooth muscle.

Studies have demonstrated that the increase in active tension reflects an increase in cytosolic free calcium.

Nifedipine is a peripheral arterial vasodilator which acts directly on vascular smooth muscle.

The binding of nifedipine to voltage-dependent and possibly receptor-operated channels in vascular smooth muscle results in an inhibition of calcium influx through these channels.

Stores of intracellular calcium in vascular smooth muscle are limited and thus dependent upon the influx of extracellular calcium for contraction to occur.

The reduction in calcium influx by nifedipine causes arterial vasodilation and decreased peripheral vascular resistance which results in reduced arterial blood pressure.

INDICATIONS AND USAGE

I.

Vasospastic Angina Nifedipine Extended-release Tablet is indicated for the management of vasospastic angina confirmed by any of the following criteria: 1) classical pattern of angina at rest accompanied by ST segment elevation, 2) angina or coronary artery spasm provoked by ergonovine, or 3) angiographically demonstrated coronary artery spasm.

In those patients who have had angiography, the presence of significant fixed obstructive disease is not incompatible with the diagnosis of vasospastic angina, provided that the above criteria are satisfied.

Nifedipine Extended-release Tablet may also be used where the clinical presentation suggests a possible vasospastic component but where vasospasm has not been confirmed, e.g., where pain has a variable threshold on exertion or in unstable angina where electrocardiographic findings are compatible with intermittent vasospasm, or when angina is refractory to nitrates and/or adequate doses of beta-blockers.

II.

Chronic Stable Angina (Classical Effort-Associated Angina) Nifedipine Extended-release Tablet is indicated for the management of chronic stable angina (effort-associated angina) without evidence of vasospasm in patients who remain symptomatic despite adequate doses of beta-blockers and/or organic nitrates or who cannot tolerate those agents.

In chronic stable angina (effort-associated angina) nifedipine has been effective in controlled trials of up to eight weeks duration in reducing angina frequency and increasing exercise tolerance, but confirmation of sustained effectiveness and evaluation of long-term safety in these patients is incomplete.

Controlled studies in small numbers of patients suggest concomitant use of nifedipine and beta-blocking agents may be beneficial in patients with chronic stable angina, but available information is not sufficient to predict with confidence the effects of concurrent treatment, especially in patients with compromised left ventricular function or cardiac conduction abnormalities.

When introducing such concomitant therapy, care must be taken to monitor blood pressure closely since severe hypotension can occur from the combined effects of the drugs.

(See WARNINGS .) III.

Hypertension Nifedipine Extended-release Tablet is indicated for the treatment of hypertension.

It may be used alone or in combination with other antihypertensive agents.

PEDIATRIC USE

Pediatric Use Safety and effectiveness in pediatric patients have not been established.

PREGNANCY

Pregnancy Pregnancy Category C Nifedipine has been shown to produce teratogenic findings in rats and rabbits, including digital anomalies similar to those reported for phenytoin.

Digital anomalies have been reported to occur with other members of the dihydropyridine class and are possibly a result of compromised uterine blood flow.

Nifedipine administration was associated with a variety of embryotoxic, placentotoxic, and fetotoxic effects, including stunted fetuses (rats, mice, rabbits), rib deformities (mice), cleft palate (mice), small placentas and underdeveloped chorionic villi (monkeys), embryonic and fetal deaths (rats, mice, rabbits), and prolonged pregnancy/decreased neonatal survival (rats; not evaluated in other species).

On a mg/kg basis, all of the doses associated with the teratogenic embryotoxic or fetotoxic effects in animals were higher (3.5 to 42 times) than the maximum recommended human dose of 120 mg/day.

On a mg/m 2 basis, some doses were higher and some were lower than the maximum recommended human dose but all are within an order of magnitude of it.

The doses associated with placentotoxic effects in monkeys were equivalent to or lower than the maximum recommended human dose on a mg/m 2 basis.

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

Nifedipine Extended-release Tablets should be used during pregnancy only if the potential benefit justifies the potential risk to the fetus.

INFORMATION FOR PATIENTS

Information for Patients Nifedipine Extended-release Tablets should be swallowed whole.

Do not chew, divide or crush tablets.

Do not be concerned if you occasionally notice in your stool something that looks like a tablet.

In Nifedipine Extended-release Tablet, the medication is contained within a nonabsorbable shell that has been specially designed to slowly release the drug for your body to absorb.

When this process is completed, the empty tablet is eliminated from your body.

DOSAGE AND ADMINISTRATION

Dosage must be adjusted according to each patient’s needs.

Therapy for either hypertension or angina should be initiated with 30 or 60 mg once daily.

Nifedipine Extended-release Tablets should be swallowed whole and should not be bitten or divided.

In general, titration should proceed over a 7-14 day period so that the physician can fully assess the response to each dose level and monitor blood pressure before proceeding to higher doses.

Since steady-state plasma levels are achieved on the second day of dosing, titration may proceed more rapidly, if symptoms so warrant, provided the patient is assessed frequently.

Titration to doses above 120 mg are not recommended.

Angina patients controlled on nifedipine capsules alone or in combination with other antianginal medications may be safely switched to Nifedipine Extended-release Tablets at the nearest equivalent total daily dose (e.g., 30 mg t.i.d.

of nifedipine capsules may be changed to 90 mg once daily of Nifedipine Extended-release Tablets).

Subsequent titration to higher or lower doses may be necessary and should be initiated as clinically warranted.

Experience with doses greater than 90 mg in patients with angina is limited.

Therefore, doses greater than 90 mg should be used with caution and only when clinically warranted.

No “rebound effect” has been observed upon discontinuation of Nifedipine Extended-release Tablets.

However, if discontinuation of nifedipine is necessary, sound clinical practice suggests that the dosage should be decreased gradually with close physician supervision.

Care should be taken when dispensing Nifedipine Extended-release Tablets to assure that the extended release dosage form has been prescribed.

Coadministration with Other Antianginal Drugs Sublingual nitroglycerin may be taken as required for the control of acute manifestations of angina, particularly during nifedipine titration.

See PRECAUTIONS, Drug Interactions , for information on coadministration of nifedipine with beta-blockers or long-acting nitrates.

Basaglar KwikPen 100 UNT/ML in 3 ML Pen Injector

Generic Name: INSULIN GLARGINE
Brand Name: BASAGLAR Tempo Pen
  • Substance Name(s):
  • INSULIN GLARGINE

DRUG INTERACTIONS

7 Table 6 includes clinically significant drug interactions with BASAGLAR Table 6: Clinically Significant Drug Interactions with BASAGLAR Drugs That May Increase the Risk of Hypoglycemia Drugs: Antidiabetic agents, ACE inhibitors, angiotensin II receptor blocking agents, disopyramide, fibrates, fluoxetine, monoamine oxidase inhibitors, pentoxifylline, pramlintide, salicylates, somatostatin analogs (e.g., octreotide), and sulfonamide antibiotics.

Intervention: Dose reductions and increased frequency of glucose monitoring may be required when BASAGLAR is co-administered with these drugs.

Drugs That May Decrease the Blood Glucose Lowering Effect of BASAGLAR Drugs: Atypical antipsychotics (e.g., olanzapine and clozapine), corticosteroids, danazol, diuretics, estrogens, glucagon, isoniazid, niacin, oral contraceptives, phenothiazines, progestogens (e.g., in oral contraceptives), protease inhibitors, somatropin, sympathomimetic agents (e.g., albuterol, epinephrine, terbutaline), and thyroid hormones Intervention: Dose increases and increased frequency of glucose monitoring may be required when BASAGLAR is co-administered with these drugs.

Drugs That May Increase or Decrease the Blood Glucose Lowering Effect of BASAGLAR Drugs: Alcohol, beta-blockers, clonidine, and lithium salts.

Pentamidine may cause hypoglycemia, which may sometimes be followed by hyperglycemia.

Intervention: Dose adjustment and increased frequency of glucose monitoring may be required when BASAGLAR is co-administered with these drugs.

Drugs That May Blunt Signs and Symptoms of Hypoglycemia Drugs: beta-blockers, clonidine, guanethidine, and reserpine Intervention: Increased frequency of glucose monitoring may be required when BASAGLAR is co-administered with these drugs.

Drugs that may increase the risk of hypoglycemia: antidiabetic agents, ACE inhibitors, angiotensin II receptor blocking agents, disopyramide, fibrates, fluoxetine, monoamine oxidase inhibitors, pentoxifylline, pramlintide, salicylates, somatostatin analog (e.g., octreotide), and sulfonamide antibiotics ( 7 ).

Drugs that may decrease the blood glucose lowering effect: atypical antipsychotics, corticosteroids, danazol, diuretics, estrogens, glucagon, isoniazid, niacin, oral contraceptives, phenothiazines, progestogens (e.g., in oral contraceptives), protease inhibitors, somatropin, sympathomimetic agents (e.g., albuterol, epinephrine, terbutaline), and thyroid hormones ( 7 ).

Drugs that may increase or decrease the blood glucose lowering effect: alcohol, beta-blockers, clonidine, lithium salts, and pentamidine ( 7 ).

Drugs that may blunt the signs and symptoms of hypoglycemia: beta-blockers, clonidine, guanethidine, and reserpine ( 7 ).

OVERDOSAGE

10 Excess insulin administration relative to food intake, energy expenditure, or both may lead to severe and sometimes prolonged and life-threatening hypoglycemia and hypokalemia [see Warnings and Precautions ( 5.3 , 5.6 )] .

Mild episodes of hypoglycemia can be treated with oral glucose.

Adjustments in drug dosage, meal patterns, or physical activity level may be needed.

More severe episodes with coma, seizure, or neurologic impairment may be treated with a glucagon product for emergency use or concentrated intravenous glucose.

Sustained carbohydrate intake and observation may be necessary because hypoglycemia may recur after apparent clinical recovery.

Hypokalemia must be corrected appropriately.

DESCRIPTION

11 Insulin glargine is a long-acting human insulin analog produced by recombinant DNA technology utilizing a non-pathogenic laboratory strain of Escherichia coli (K12) as the production organism.

Insulin glargine differs from human insulin in that the amino acid asparagine at position A21 is replaced by glycine and two arginines are added to the C-terminus of the B-chain.

Chemically, insulin glargine is 21 A -Gly-30 B -a-L-Arg-30 B b-L-Arg-human insulin and has the empirical formula C 267 H 404 N 72 O 78 S 6 and a molecular weight of 6.063 kDa.

Insulin glargine has the following structural formula: BASAGLAR (insulin glargine) injection is a sterile clear and colorless aqueous solution for subcutaneous use.

Each mL contains 100 units of insulin glargine (3.6378 mg).

The 3 mL BASAGLAR prefilled pen presentations contain the following inactive ingredients per mL: glycerin (17 mg), metacresol (2.7 mg), zinc oxide (content adjusted to provide 30 mcg zinc ion), and Water for Injection, USP.

The pH is adjusted by addition of aqueous solutions of hydrochloric acid 10% and/or sodium hydroxide 10%.

BASAGLAR has a pH of approximately 4.

Structural Formula

CLINICAL STUDIES

14 14.1 Overview of Clinical Studies The safety and effectiveness of another insulin glargine product, 100 units/mL, given once-daily at bedtime was compared to that of once-daily and twice-daily NPH insulin in open-label, randomized, active-controlled, parallel studies of 2,327 adults and 349 pediatric patients with type 1 diabetes mellitus and 1,563 adult patients with type 2 diabetes mellitus ( see Tables 8 , 9 , 11 , and 12 ).

In general, the reduction in glycated hemoglobin (HbA 1c ) with this other insulin glargine product was similar to that with NPH insulin.

14.2 Clinical Studies in Adult and Pediatric Patients with Type 1 Diabetes Patients with inadequately controlled type 1 diabetes participated in a 24-week open-label, active-controlled study with a 28 week extension to evaluate the glucose lowering effect of once-daily BASAGLAR compared to that of once-daily administration of another insulin glargine product, 100 units/mL, or a non-U.S.-licensed insulin glargine, 100 units/mL, (comparator insulin glargine products, 100 units/mL) both in combination with mealtime insulin lispro.

Randomized were 535 adults with type 1 diabetes.

Mean age was 41.2 years and mean duration of diabetes was 16.39 years.

57.9% were male.

74.5% were Caucasian, 2.1% Black or African American and 4.3% American Indian or Alaskan native.

3.9% were Hispanic.

73.5 percent of patients had GFR>90 mL/min/1.73m 2 .

The mean BMI was approximately 25.54 kg/m 2 .

At week 24, treatment with BASAGLAR provided a mean reduction in HbA 1c that was non-inferior to that achieved with comparator insulin glargine products, 100 units/mL ( see Table 7 ).

Table 7: Type 1 Diabetes Mellitus – Adult (BASAGLAR plus Mealtime insulin versus Comparator Insulin Glargine Products, 100 units/mL, plus Mealtime Insulin) a One patient randomized to the BASAGLAR group was not included in the Full Analysis Set.

b “Comparator insulin glargine products, 100 units/mL” refers to another insulin glargine product, 100 units/mL, and a non-U.S.-licensed insulin glargine, 100 units/mL, used in this study.

c ANCOVA Model includes treatment, country and time of baseline basal insulin injection (daytime or evening/bedtime) as fixed effects and baseline HbA 1c as covariate.

d The results were calculated based on the number of patients in the Full Analysis Set using their last observed post-baseline value of HbA 1c .

Observed HbA 1c data at 24 weeks were available from 256 (95.5%) and 258 (96.6%) subjects randomized to the BASAGLAR and comparator insulin glargine products, 100 units/mL, groups, respectively.

Efficacy Parameter BASAGLAR + insulin lispro (N=268 a ) Comparator Insulin Glargine Products, 100 units/mL b + insulin lispro (N=267) HbA 1c (%) Baseline (mean) 7.75 7.79 Change from baseline (adjusted mean c,d ) -0.35 -0.46 Difference from comparator (adjusted mean c,d ) (95% CI) 0.11 (-0.002, 0.219) Proportion of patients achieving HbA 1c <7% d 34.5% 32.2% In two clinical studies (Studies A and B), patients with type 1 diabetes (Study A; n=585, Study B; n=534) were randomized to 28 weeks of basal-bolus treatment with another insulin glargine product, 100 units/mL, or NPH insulin.

Regular human insulin was administered before each meal.

This other insulin glargine product was administered at bedtime.

NPH insulin was administered once daily at bedtime or in the morning and at bedtime when used twice daily.

In Study A, the average age was 39.2 years.

The majority of patients were Caucasian (99%) and 55.7% were male.

The mean BMI was approximately 24.9 kg/m 2 .

The mean duration of diabetes was 15.5 years.

In Study B, the average age was 38.5 years.

The majority of patients were Caucasian (95.3%) and 50.6% were male.

The mean BMI was approximately 25.8 kg/m 2 .

The mean duration of diabetes was 17.4 years.

In another clinical study (Study C), patients with type 1 diabetes (n=619) were randomized to 16 weeks of basal-bolus treatment with another insulin glargine product, 100 units/mL, or NPH insulin.

Insulin lispro was used before each meal.

This other insulin glargine product was administered once daily at bedtime and NPH insulin was administered once or twice daily.

The average age was 39.2 years.

The majority of patients were Caucasian (96.9%) and 50.6% were male.

The mean BMI was approximately 25.6 kg/m 2 .

The mean duration of diabetes was 18.5 years.

In these 3 studies, another insulin glargine product, 100 units/mL, and NPH insulin had similar effects on HbA 1c ( see Table 8 ) with a similar overall rate of hypoglycemia [see Adverse Reactions ( 6.1 )] .

Table 8: Type 1 Diabetes Mellitus – Adult (Another Insulin Glargine Product, 100 units/mL, versus NPH) Treatment duration Treatment in combination with Study A 28 weeks Regular insulin Study B 28 weeks Regular insulin Study C 16 weeks Insulin lispro Another Insulin Glargine Product NPH Another Insulin Glargine Product NPH Another Insulin Glargine Product NPH Number of subject treated 292 293 264 270 310 309 HbA 1c (%) Baseline (mean) 8.0 8.0 7.7 7.7 7.6 7.7 Adjusted mean change at trial end +0.2 +0.1 -0.2 -0.2 -0.1 -0.1 Treatment Difference (95% CI) +0.1 (0.0; + 0.2) +0.1(-0.1; + 0.2) 0.0 (+0.1; + 0.1) Fasting blood glucose (mg/dL) Baseline (mean) 167 166 166 175 175 173 Adjusted mean change at trial end -21 -16 -20 -17 -29 -12 Type 1 Diabetes – Pediatric ( see Table 9) The efficacy of BASAGLAR to improve glycemic control in pediatric patients with type 1 diabetes mellitus is based on an adequate and well-controlled trial of another insulin glargine product, 100 units/mL, in pediatric patients with type 1 diabetes mellitus (Study D).

In this randomized, active-controlled clinical study (Study D), pediatric patients (age range 6 to 15 years) with type 1 diabetes (n=349) were treated for 28 weeks with a basal-bolus insulin regimen where regular human insulin was used before each meal.

Patients were randomized to either this other insulin glargine product administered once daily at bedtime or NPH insulin administered once or twice daily.

The average age was 11.7 years.

The majority of patients were Caucasian (96.8%) and 51.9% were male.

The mean BMI was approximately 18.9 kg/m 2 .

The mean duration of diabetes was 4.8 years.

Similar effects on HbA 1c ( see Table 9 ) were observed in both treatment groups.

Table 9: Type 1 Diabetes Mellitus – Pediatric (Another Insulin Glargine Product, 100 units/mL, plus Regular Insulin versus NPH plus Regular Insulin) Study D Another Insulin Glargine Product + Regular Insulin NPH + Regular Insulin Number of subjects treated 174 175 HbA 1c Baseline mean 8.5 8.8 Change from baseline (adjusted mean) +0.3 +0.3 Difference from NPH (adjusted mean) (95% CI) 0.0 (-0.2; +0.3) Fasting blood glucose (mg/dL) Baseline mean 194 191 Mean change from baseline -23 -12 14.3 Clinical Studies in Adults with Type 2 Diabetes Patients with type 2 diabetes participated in a double-blind, active-controlled study to evaluate the glucose lowering effect of once-daily BASAGLAR plus oral antidiabetic medication (OAM) compared to that of another insulin glargine product, 100 units/mL, or a non-U.S.-licensed insulin glargine, 100 units/mL (comparator insulin glargine products, 100 units/mL) administered once-daily along with OAMs.

Patients were either insulin naïve (approximately 60%) and had failed to achieve adequate glycemic control on at least 2 OAMs, or were already on another insulin glargine product, 100 units/mL, or a non-U.S.-licensed insulin glargine, 100 units/mL, along with at least 2 OAMs with adequate or inadequate glycemic control (approximately 40%).

A total of 759 patients were randomized.

Three patients randomized to BASAGLAR did not receive study drug and were not included in efficacy analysis.

The average age was approximately 59 years.

The majority of patients were White (78%) and 50% of the patients were male.

Sixty-eight percent of patients had GFR>90 mL/min/1.73m 2 .

The mean BMI was approximately 32 kg/m 2 .

At week 24, treatment with BASAGLAR provided a mean reduction in HbA 1c that was non-inferior to that achieved with comparator insulin glargine products, 100 units/mL ( see Table 10 ).

Table 10: Type 2 Diabetes Mellitus – Adult (BASAGLAR plus Oral Antidiabetic Medications versus Comparator Insulin Glargine Products, 100 units/mL, plus Oral Antidiabetic Medications) a Three patients randomized to BASAGLAR did not receive study drug and were not included in the Full Analysis Set.

b “Comparator insulin glargine products, 100 units/mL” refers to another insulin glargine product, 100 units/mL, and a non-U.S.-licensed insulin glargine, 100 units/mL, used in this study.

c ANCOVA Model includes treatment, country, sulfonylurea use and time of baseline basal insulin injection (daytime or evening/bedtime) as fixed effects and baseline HbA 1c as covariate.

d The results were calculated based on the number of patients in the Full Analysis Set using their last observed post-baseline value of HbA 1c .

Observed HbA 1c data at 24 weeks were available from 331 (88%) and 329 (87%) subjects randomized to the BASAGLAR and comparator insulin glargine products, 100 units/mL, groups, respectively.

BASAGLAR + Oral Antidiabetic Medication (N=376) a Comparator Insulin Glargine Products, 100 units/mL b + Oral Antidiabetic Medication (N=380) HbA 1c (%) Baseline (mean) 8.35 8.31 Change from baseline (adjusted mean c,d ) -1.3 -1.3 Difference from comparator (adjusted mean c,d ) (95% CI) 0.05 (-0.07, 0.17) Proportion of patients achieving HbA 1c <7% d 48.8% 52.5% In a randomized, controlled clinical study (Study E) (n=570), another insulin glargine product, 100 units/mL, was evaluated for 52 weeks in combination with oral anti-diabetic medications (a sulfonylurea, metformin, acarbose, or combination of these drugs).

The average age was 59.5 years.

The majority of patients were Caucasian (92.8%) and 53.7% were male.

The mean BMI was approximately 29.1 kg/m 2 .

The mean duration of diabetes was 10.3 years.

This other insulin glargine product administered once daily at bedtime was as effective as NPH insulin administered once daily at bedtime in reducing HbA 1c and fasting glucose ( see Table 11 ).

The rate of hypoglycemia was similar in this other insulin glargine product and NPH insulin treated patients [see Adverse Reactions ( 6.1 )] .

In a randomized, controlled clinical study (Study F), in patients with type 2 diabetes not using oral anti-diabetic medications (n=518), a basal-bolus regimen of another insulin glargine product, 100 units/mL, once daily at bedtime or NPH insulin administered once or twice daily was evaluated for 28 weeks.

Regular human insulin was used before meals, as needed.

The average age was 59.3 years.

The majority of patients were Caucasian (80.7%) and 60% were male.

The mean BMI was approximately 30.5 kg/m 2 .

The mean duration of diabetes was 13.7 years.

This other insulin glargine product had similar effectiveness as either once- or twice daily NPH insulin in reducing HbA 1c and fasting glucose ( see Table 11 ) with a similar incidence of hypoglycemia [see Adverse Reactions ( 6.1 )] .

In a randomized, controlled clinical study (Study G), patients with type 2 diabetes were randomized to 5 years of treatment with another insulin glargine product, 100 units/mL, once-daily or twice-daily NPH insulin.

For patients not previously treated with insulin, the starting dose of this other insulin glargine product or NPH insulin was 10 units daily.

Patients who were already treated with NPH insulin either continued on the same total daily NPH insulin dose or started this other insulin glargine product at a dose that was 80% of the total previous NPH insulin dose.

The primary endpoint for this study was a comparison of the progression of diabetic retinopathy by 3 or more steps on the ETDRS scale.

HbA 1c change from baseline was a secondary endpoint.

Similar glycemic control in the 2 treatment groups was desired in order to not confound the interpretation of the retinal data.

Patients or study personnel used an algorithm to adjust this other insulin glargine product and NPH insulin doses to a target fasting plasma glucose ≤100 mg/dL.

After this other insulin glargine product or NPH insulin dose was adjusted, other anti-diabetic agents, including pre-meal insulin were to be adjusted or added.

The average age was 55.1 years.

The majority of patients were Caucasian (85.3%) and 53.9% were male.

The mean BMI was approximately 34.3 kg/m 2 .

The mean duration of diabetes was 10.8 years.

This other insulin glargine product group had a smaller mean reduction from baseline in HbA 1c compared to the NPH insulin group, which may be explained by the lower daily basal insulin doses in this other insulin glargine product group ( see Table 11 ).

Both treatment groups had a similar incidence of reported symptomatic hypoglycemia.

The incidence of severe symptomatic hypoglycemia in the ORIGIN Trial is given in Table 5 [see Adverse Reactions ( 6.1 )] .

Table 11: Type 2 Diabetes Mellitus – Adult (Another Insulin Glargine Product, 100 units/mL, versus NPH) Treatment duration Treatment in combination with Study E 52 weeks Oral agents Study F 28 weeks Regular insulin Study G 5 years Regular insulin Another Insulin Glargine Product NPH Another Insulin Glargine Product NPH Another Insulin Glargine Product NPH Number of subjects treated 289 281 259 259 513 504 HbA 1c Baseline mean 9.0 8.9 8.6 8.5 8.4 8.3 Adjusted mean change from baseline -0.5 -0.4 -0.4 -0.6 -0.6 -0.8 Another insulin glargine product, 100 units/mL – NPH -0.1 +0.2 +0.2 95% CI for Treatment difference (-0.3; +0.1) (0.0; +0.4) (+0.1; +0.4) Fasting blood glucose (mg/dL) Baseline mean 179 180 164 166 190 180 Adjusted mean change from baseline -49 -46 -24 -22 -45 -44 Another Insulin Glargine Product, 100 units/mL, Timing of Daily Dosing ( see Table 12) The safety and efficacy of this other insulin glargine product administered pre-breakfast, pre-dinner, or at bedtime were evaluated in a randomized, controlled clinical study in patients with type 1 diabetes (Study H; n=378).

Patients were also treated with insulin lispro at mealtime.

The average age was 40.9 years.

All patients were Caucasian (100%) and 53.7% were male.

The mean BMI was approximately 25.3 kg/m 2 .

The mean duration of diabetes was 17.3 years.

This other insulin glargine product administered at different times of the day resulted in similar reductions in HbA 1c compared to that with bedtime administration ( see Table 12 ).

In these patients, data are available from 8-point home glucose monitoring.

The maximum mean blood glucose was observed just prior to injection of this other insulin glargine product regardless of time of administration.

In this study, 5% of patients in this other insulin glargine product-breakfast arm discontinued treatment because of lack of efficacy.

No patients in the other two arms discontinued for this reason.

The safety and efficacy of this other insulin glargine product administered pre-breakfast or at bedtime were also evaluated in a randomized, active-controlled clinical study (Study I, n=697) in patients with type 2 diabetes not adequately controlled on oral anti-diabetic therapy.

All patients in this study also received glimepiride 3 mg daily.

The average age was 60.8 years.

The majority of patients were Caucasian (96.6%) and 53.7% were male.

The mean BMI was approximately 28.7 kg/m 2 .

The mean duration of diabetes was 10.1 years.

This other insulin glargine product given before breakfast was at least as effective in lowering HbA 1c as this other insulin glargine product given at bedtime or NPH insulin given at bedtime ( see Table 12 ).

Table 12: Type 1 Diabetes Mellitus – Adults (Another Insulin Glargine Product, 100 units/mL, plus Insulin Lispro) and Type 2 Diabetes Mellitus – Adults (Another Insulin Glargine Product, 100 units/mL, plus Glimepiride versus NPH plus Glimepiride) a Intent to treat.

b Total number of patients evaluable for safety.

c Not applicable.

Treatment duration Treatment in combination with Study H 24 weeks Insulin lispro Study I 24 weeks Glimepiride Another Insulin Glargine Product Breakfast Another Insulin Glargine Product Dinner Another Insulin Glargine Product Bedtime Another Insulin Glargine Product Breakfast Another Insulin Glargine Product Bedtime NPH Bedtime Number of subjects treated a 112 124 128 234 226 227 HbA 1c Baseline mean 7.6 7.5 7.6 9.1 9.1 9.1 Mean change from baseline -0.2 -0.1 0.0 -1.3 -1.0 -0.8 Five-year Trial Evaluating the Progression of Retinopathy Retinopathy was evaluated in clinical studies with another insulin glargine product, 100 units/mL, by analysis of reported retinal adverse events and fundus photography.

The numbers of retinal adverse events reported for this other insulin glargine product and NPH insulin treatment groups were similar for patients with type 1 and type 2 diabetes.

Another insulin glargine product, 100 units/mL, was compared to NPH insulin in a 5-year randomized clinical trial that evaluated the progression of retinopathy as assessed with fundus photography using a grading protocol derived from the Early Treatment Diabetic Retinopathy Scale (ETDRS).

Patients had type 2 diabetes (mean age 55 years) with no (86%) or mild (14%) retinopathy at baseline.

Mean baseline HbA 1c was 8.4%.

The primary outcome was progression by 3 or more steps on the ETDRS scale at study endpoint.

Patients with pre-specified post-baseline eye procedures (pan-retinal photocoagulation for proliferative or severe nonproliferative diabetic retinopathy, local photocoagulation for new vessels, and vitrectomy for diabetic retinopathy) were also considered as 3-step progressions regardless of actual change in ETDRS score from baseline.

Retinopathy graders were blinded to treatment group assignment.

The results for the primary endpoint are shown in Table 13 for both the per-protocol and Intent-to-Treat populations, and indicate similarity of this other insulin glargine product to NPH in the progression of diabetic retinopathy as assessed by this outcome.

Table 13: Number (%) of Patients with 3 or More Step Progression on ETDRS Scale at Endpoint a Difference = another insulin glargine product, 100 units/mL – NPH.

b Using a generalized linear model (SAS GENMOD) with treatment and baseline HbA 1c strata (cutoff 9.0%) as the classified independent variables, and with binomial distribution and identity link function.

Another Insulin Glargine Product, 100 units/mL (%) NPH (%) Difference a,b (SE) 95% CI for difference Per-protocol 53/374 (14.2%) 57/363 (15.5%) -2.0% (2.6%) -7.0% to +3.1% Intent-to-Treat 63/502 (12.5%) 71/487 (14.6%) -2.1% (2.1%) -6.3% to +2.1% The ORIGIN Study The Outcome Reduction with Initial Glargine Intervention trial (i.e., ORIGIN) was an open-label, randomized, 2-by-2, factorial design study.

One intervention in ORIGIN compared the effect of another insulin glargine product, 100 units/mL, to standard care on major adverse cardiovascular outcomes in 12,537 participants ≥50 years of age with abnormal glucose levels [i.e., impaired fasting glucose (IFG) and/or impaired glucose tolerance (IGT)] or early type 2 diabetes mellitus and established cardiovascular (i.e., CV) disease or CV risk factors at baseline.

The objective of the trial was to demonstrate that use of this other insulin glargine product could significantly lower the risk of major cardiovascular outcomes compared to standard care.

Two co-primary composite cardiovascular endpoints were used in ORIGIN.

The first co-primary endpoint was the time to first occurrence of a major adverse cardiovascular event defined as the composite of CV death, nonfatal myocardial infarction and nonfatal stroke.

The second co-primary endpoint was the time to the first occurrence of CV death or nonfatal myocardial infarction or nonfatal stroke or revascularization procedure or hospitalization for heart failure.

Participants were randomized to either this other insulin glargine product (N=6264) titrated to a goal fasting plasma glucose of ≤95 mg/dL or to standard care (N=6273).

Anthropometric and disease characteristics were balanced at baseline.

The mean age was 64 years and 8% of participants were 75 years of age or older.

The majority of participants were male (65%).

Fifty nine percent were Caucasian, 25% were Latin, 10% were Asian and 3% were Black.

The median baseline BMI was 29 kg/m 2 .

Approximately 12% of participants had abnormal glucose levels (IGT and/or IFG) at baseline and 88% had type 2 diabetes.

For patients with type 2 diabetes, 59% were treated with a single oral antidiabetic drug, 23% had known diabetes but were on no antidiabetic drug and 6% were newly diagnosed during the screening procedure.

The mean HbA 1c (SD) at baseline was 6.5% (1.0).

Fifty nine percent of participants had had a prior cardiovascular event and 39% had documented coronary artery disease or other cardiovascular risk factors.

Vital status was available for 99.9% and 99.8% of participants randomized to this other insulin glargine product and standard care respectively at end of trial.

The median duration of follow-up was 6.2 years [range: 8 days to 7.9 years].

The mean HbA 1c (SD) at the end of the trial was 6.5% (1.1) and 6.8% (1.2) in this other insulin glargine product and standard group respectively.

The median dose of this other insulin glargine product at end of trial was 0.45 U/kg.

Eighty-one percent of patients randomized to this other insulin glargine product were using this other insulin glargine product at end of the study.

The mean change in body weight from baseline to the last treatment visit was 2.2 kg greater in this other insulin glargine group than in the standard care group.

Overall, the incidence of major adverse cardiovascular outcomes was similar between groups ( see Table 14 ).

All-cause mortality was also similar between groups.

Table 14: Cardiovascular Outcomes in ORIGIN – Time to First Event Analyses Another Insulin Glargine Product, 100 units/mL N=6264 Standard Care N=6273 Another Insulin Glargine Product, 100 units/mL vs.

Standard Care n (Events per 100 PY) n (Events per 100 PY) Hazard Ratio (95% CI) Co-primary endpoints CV death, nonfatal myocardial infarction, or nonfatal stroke 1041 (2.9) 1013 (2.9) 1.02 (0.94, 1.11) CV death, nonfatal myocardial infarction, nonfatal stroke, hospitalization for heart failure or revascularization procedure 1792 (5.5) 1727 (5.3) 1.04 (0.97, 1.11) Components of co-primary endpoints CV death 580 576 1.00 (0.89, 1.13) Myocardial Infarction (fatal or nonfatal) 336 326 1.03 (0.88, 1.19) Stroke (fatal or nonfatal) 331 319 1.03 (0.89, 1.21) Revascularizations 908 860 1.06 (0.96, 1.16) Hospitalization for heart failure 310 343 0.90 (0.77, 1.05) In the ORIGIN trial, the overall incidence of cancer (all types combined) or death from cancer in the ORIGIN trial ( see Table 15 ) was similar between treatment groups.

Table 15: Cancer Outcomes in ORIGIN – Time to First Event Analyses Another Insulin Glargine Product, 100 units/mL N=6264 Standard Care N=6273 Another Insulin Glargine Product, 100 units/mL vs.

Standard Care n (Events per 100 PY) n (Events per 100 PY) Hazard Ratio (95% CI) Cancer endpoints Any cancer event (new or recurrent) 559 (1.56) 561 (1.56) 0.99 (0.88, 1.11) New cancer events 524 (1.46) 535 (1.49) 0.96 (0.85, 1.09) Death due to Cancer 189 (0.51) 201 (0.54) 0.94 (0.77, 1.15)

HOW SUPPLIED

16 /STORAGE AND HANDLING 16.1 How Supplied BASAGLAR (insulin glargine) injection is a clear, colorless solution, 100 units/mL (U-100), available as: a Tempo Pen contains a component that allows for data connectivity when used with a compatible transmitter.

BASAGLAR Total Volume NDC Number Package Size BASAGLAR single-patient-use KwikPen 3 mL 0002-7715-59 5 pens BASAGLAR single-patient-use Tempo Pen a 3 mL 0002-8214-05 5 pens The BASAGLAR KwikPen and Tempo Pen dial in 1 unit increments.

Needles are not included.

This device is recommended for use with Becton, Dickinson & Company’s insulin pen needles which are sold separately.

16.2 Storage and Handling Dispense in the original sealed carton with the enclosed Instructions for Use.

Protect BASAGLAR from heat and light.

Do not freeze BASAGLAR.

In-use BASAGLAR prefilled pens must be used within 28 days or be discarded, even if they still contain BASAGLAR.

Storage conditions are summarized in the following table: Not In-Use (Unopened) Room Temperature (up to 86°F [30°C]) Not In-Use (Unopened) Refrigerated (36°F to 46°F [2°C to 8°C]) In-Use (Opened) Room Temperature, (up to 86°F [30°C]) 3 mL single-patient-use BASAGLAR KwikPen 28 days Until expiration date 28 days, Do not refrigerate.

3 mL single-patient-use BASAGAR Tempo Pen 28 days Until expiration date 28 days, Do not refrigerate.

GERIATRIC USE

8.5 Geriatric Use Of the total number of subjects in clinical studies of patients with type 2 diabetes who were treated with BASAGLAR or another insulin glargine product, 100 units/mL, each in combination with oral agents in a controlled clinical trial environment, 28.3% were 65 and over, while 4.5% were 75 and over.

No overall differences in safety or effectiveness were observed between these subjects and younger subjects, and other reported clinical experience has not identified differences in responses between the elderly and younger patients, but greater sensitivity of some older individuals cannot be ruled out.

Nevertheless, caution should be exercised when BASAGLAR is administered to geriatric patients.

In elderly patients with diabetes, the initial dosing, dose increments, and maintenance dosage should be conservative to avoid hypoglycemic reactions.

Hypoglycemia may be difficult to recognize in the elderly.

DOSAGE FORMS AND STRENGTHS

3 Injection: 100 units/mL (U-100) clear and colorless solution available as: 3 mL single-patient-use BASAGLAR KwikPen 3 mL single-patient-use BASAGLAR Tempo Pen Injection: 100 units/mL (U-100) is available as: ( 3 ) 3 mL single-patient-use BASAGLAR KwikPen ® 3 mL single-patient-use BASAGLAR Tempo Pen TM

MECHANISM OF ACTION

12.1 Mechanism of Action The primary activity of insulin, including insulin glargine, is regulation of glucose metabolism.

Insulin and its analog lower blood glucose by stimulating peripheral glucose uptake, especially by skeletal muscle and fat, and by inhibiting hepatic glucose production.

Insulin inhibits lipolysis and proteolysis, and enhances protein synthesis.

INDICATIONS AND USAGE

1 BASAGLAR ® is indicated to improve glycemic control in adults and pediatric patients with type 1 diabetes mellitus and in adults with type 2 diabetes mellitus.

BASAGLAR ® is a long-acting human insulin analog indicated to improve glycemic control in adults and pediatric patients with type 1 diabetes mellitus and in adults with type 2 diabetes mellitus.

( 1 ) Limitations of Use : Not recommended for treating diabetic ketoacidosis.

( 1 ) Limitations of Use BASAGLAR is not recommended for the treatment of diabetic ketoacidosis.

PEDIATRIC USE

8.4 Pediatric Use The safety and effectiveness of BASAGLAR have been established in pediatric patients (age 6 to 15 years) with type 1 diabetes based on an adequate and well-controlled trial of another insulin glargine product, 100 units/mL, in pediatric patients (age 6 to 15 years) with type 1 diabetes and additional data in adults with type 1 diabetes [see Clinical Studies ( 14.2 )] .

The safety and effectiveness of BASAGLAR in pediatric patients younger than 6 years of age with type 1 diabetes and pediatric patients with type 2 diabetes has not been established.

In the pediatric clinical trial, pediatric patients (age 6 to 15 years) with type 1 diabetes had a higher incidence of severe symptomatic hypoglycemia compared to the adults in trials with type 1 diabetes [see Adverse Reactions ( 6.1 )] .

PREGNANCY

8.1 Pregnancy Risk Summary Published studies with use of insulin glargine products during pregnancy have not reported a clear association with insulin glargine products and adverse developmental outcomes (see Data).

There are risks to the mother and fetus associated with poorly controlled diabetes in pregnancy (see Clinical Considerations).

In animal reproduction studies, another insulin glargine product was administered to rats before, during and throughout pregnancy at doses up to 7 times the clinical dose of 10 units/day and to rabbits during organogenesis at doses approximately 2 times the clinical dose of 10 units/day.

The effects of this other insulin glargine product did not generally differ from those observed with regular human insulin in rats or rabbits (see Data).

The estimated background risk of major birth defects is 6-10% in women with pre-gestational diabetes with a HbA1c >7 and has been reported to be as high as 20-25% in women with a HbA1c >10.

The estimated background risk of miscarriage for the indicated population is unknown.

In the US general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2-4% and 15-20%, respectively.

Clinical Considerations Disease-associated maternal and/or embryo/fetal risk Poorly controlled diabetes in pregnancy increases the maternal risk for diabetic ketoacidosis, pre-eclampsia, spontaneous abortions, preterm delivery, and delivery complications.

Poorly controlled diabetes increases the fetal risk for major birth defects, stillbirth, and macrosomia related morbidity.

Data Human Data Published data do not report a clear association with insulin glargine products and major birth defects, miscarriage, or adverse maternal or fetal outcomes when insulin glargine products are used during pregnancy.

However, these studies cannot definitely establish the absence of any risk because of methodological limitations including small sample size and some with no comparative group.

Animal Data Subcutaneous reproduction and teratology studies have been performed with another insulin glargine product and with regular human insulin in rats and Himalayan rabbits.

This other insulin glargine product was given to female rats before mating, during mating, and throughout pregnancy at dose up to 0.36 mg/kg/day, which is approximately 7 times the recommended human subcutaneous starting dose of 10 units/day (0.008 mg/kg/day) based on mg/m 2 .

In rabbits, doses of 0.072 mg/kg/day, which is approximately 2 times the recommended human subcutaneous starting dose of 10 units/day (0.008 mg/kg/day), based on mg/m 2 , were administered during organogenesis.

The effects of this other insulin glargine product did not generally differ from those observed with regular human insulin in rats and rabbits.

However, in rabbits, five fetuses from two litters of the high-dose group exhibited dilation of the cerebral ventricles.

Fertility and early embryonic development appeared normal.

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS Never share a BASAGLAR prefilled pen between patients, even if the needle is changed.

( 5.1 ) Hyperglycemia or hypoglycemia with changes in insulin regimen: Make changes to a patient’s insulin regimen (e.g., insulin strength, manufacturer, type, injection site or method of administration) under close medical supervision with increased frequency of blood glucose monitoring.

( 5.2 ) Hypoglycemia: May be life-threatening.

Increase frequency of glucose monitoring with changes to: insulin dosage, co-administered glucose lowering medications, meal pattern, physical activity; and in patients with renal or hepatic impairment and hypoglycemia unawareness.

( 5.3 , 6.1 ) Hypoglycemia due to medication errors: Accidental mix-ups between insulin products can occur.

Instruct patients to check insulin labels before injection.

( 5.4 ) Hypersensitivity reactions: Severe, life-threatening, generalized allergy, including anaphylaxis, can occur.

Discontinue BASAGLAR, monitor and treat if indicated.

( 5.5 , 6.1 ) Hypokalemia: May be life-threatening.

Monitor potassium levels in patients at risk of hypokalemia and treat if indicated.

( 5.6 ) Fluid retention and heart failure with concomitant use of thiazolidinediones (TZDs): Observe for signs and symptoms of heart failure; consider dosage reduction or discontinuation if heart failure occurs.

( 5.7 ) 5.1 Never Share a BASAGLAR Prefilled Pen Between Patients BASAGLAR prefilled pens must never be shared between patients, even if the needle is changed.

Sharing poses a risk for transmission of blood-borne pathogens.

5.2 Hyperglycemia or Hypoglycemia with Changes in Insulin Regimen Changes in an insulin regimen (e.g., insulin strength, manufacturer, type, injection site or method of administration) may affect glycemic control and predispose to hypoglycemia [see Warnings and Precautions ( 5.3 )] or hyperglycemia.

Repeated insulin injections into areas of lipodystrophy or localized cutaneous amyloidosis have been reported to result in hyperglycemia; and a sudden change in the injection site (to an unaffected area) has been reported to result in hypoglycemia [see Adverse Reactions ( 6 )] .

Make any changes to a patient’s insulin regimen under close medical supervision with increased frequency of blood glucose monitoring.

Advise patients who have repeatedly injected into areas of lipodystrophy or localized cutaneous amyloidosis to change the injection site to unaffected areas and closely monitor for hypoglycemia.

For patients with type 2 diabetes, dosage adjustments of concomitant anti-diabetic products may be needed.

5.3 Hypoglycemia Hypoglycemia is the most common adverse reaction associated with insulins, including BASAGLAR [see Adverse Reactions ( 6.1 )] .

Severe hypoglycemia can cause seizures, may be life-threatening or cause death.

Hypoglycemia can impair concentration ability and reaction time; this may place an individual and others at risk in situations where these abilities are important (e.g., driving or operating other machinery).

BASAGLAR, or any insulin, should not be used during episodes of hypoglycemia [see Contraindications ( 4 )] .

Hypoglycemia can happen suddenly and symptoms may differ in each individual and change over time in the same individual.

Symptomatic awareness of hypoglycemia may be less pronounced in patients with longstanding diabetes, in patients with diabetic nerve disease, in patients using medications that block the sympathetic nervous system (e.g., beta-blockers) [see Drug Interactions ( 7 )] , or in patients who experience recurrent hypoglycemia.

Risk Factors for Hypoglycemia The risk of hypoglycemia after an injection is related to the duration of action of the insulin and, in general, is highest when the glucose lowering effect of the insulin is maximal.

As with all insulins, the glucose lowering effect time course of BASAGLAR may vary in different individuals or at different times in the same individual and depends on many conditions, including the area of injection as well as the injection site blood supply and temperature [see Clinical Pharmacology ( 12.2 )] .

The risk of hypoglycemia generally increases with intensity of glycemic control.

Other factors which may increase the risk of hypoglycemia include changes in meal pattern (e.g., macronutrient content or timing of meals), changes in level of physical activity, or changes to co-administered medication [see Drug Interactions ( 7 )] .

Patients with renal or hepatic impairment may be at higher risk of hypoglycemia [see Use in Specific Populations ( 8.6 , 8.7 )] .

Risk Mitigation Strategies for Hypoglycemia Patients and caregivers must be educated to recognize and manage hypoglycemia.

Self-monitoring of blood glucose plays an essential role in the prevention and management of hypoglycemia.

In patients at higher risk for hypoglycemia and patients who have reduced symptomatic awareness of hypoglycemia, increased frequency of blood glucose monitoring is recommended.

The long-acting effect of BASAGLAR may delay recovery from hypoglycemia.

5.4 Hypoglycemia Due to Medication Errors Accidental mix-ups between insulin products have been reported.

To avoid medication errors between BASAGLAR and other insulins, instruct patients to always check the insulin label before each injection.

5.5 Hypersensitivity Reactions Severe, life-threatening, generalized allergy, including anaphylaxis, can occur with insulins, including BASAGLAR.

If hypersensitivity reactions occur, discontinue BASAGLAR; treat per standard of care and monitor until symptoms and signs resolve [see Adverse Reactions ( 6.1 )] .

BASAGLAR is contraindicated in patients who have had hypersensitivity reactions to insulin glargine or one of the excipients [see Contraindications ( 4 )] .

5.6 Hypokalemia All insulins, including BASAGLAR, cause a shift in potassium from the extracellular to intracellular space, possibly leading to hypokalemia.

Untreated hypokalemia may cause respiratory paralysis, ventricular arrhythmia, and death.

Monitor potassium levels in patients at risk for hypokalemia if indicated (e.g., patients using potassium-lowering medications, patients taking medications sensitive to serum potassium concentrations).

5.7 Fluid Retention and Heart Failure with Concomitant Use of PPAR-gamma Agonists Thiazolidinediones (TZDs), which are peroxisome proliferator-activated receptor (PPAR)-gamma agonists, can cause dose-related fluid retention, particularly when used in combination with insulin.

Fluid retention may lead to or exacerbate heart failure.

Patients treated with insulin, including BASAGLAR, and a PPAR-gamma agonist should be observed for signs and symptoms of heart failure.

If heart failure develops, it should be managed according to current standards of care, and discontinuation or dose reduction of the PPAR-gamma agonist must be considered.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION See FDA-approved patient labeling (Patient Information and Instructions for Use).

Never Share a BASAGLAR Prefilled Pen Between Patients Advise patients that they must never share a BASAGLAR prefilled pen with another person, even if the needle is changed, because doing so carries a risk for transmission of blood-borne pathogens [see Warnings and Precautions ( 5.1 )] .

Hyperglycemia or Hypoglycemia Inform patients that hypoglycemia is the most common adverse reaction with insulin.

Inform patients of the symptoms of hypoglycemia.

Inform patients that the ability to concentrate and react may be impaired as a result of hypoglycemia.

This may present a risk in situations where these abilities are especially important, such as driving or operating other machinery.

Advise patients who have frequent hypoglycemia or reduced or absent warning signs of hypoglycemia to use caution when driving or operating machinery.

Advise patients that changes in insulin regimen can predispose to hyperglycemia or hypoglycemia and that changes in insulin regimen should be made under close medical supervision [see Warnings and Precautions ( 5.2 )] .

Medication errors Inform patients to always check the insulin label before each injection [see Warnings and Precautions ( 5.4 )].

Literature revised July 2021 Manufactured by: Eli Lilly and Company Indianapolis, IN 46285, USA US License Number 1891 Copyright © 2015, 2021, Eli Lilly and Company.

All rights reserved.

BAS-0007-USPI-20210727

DOSAGE AND ADMINISTRATION

2 Individualize dosage based on metabolic needs, blood glucose monitoring, glycemic control, type of diabetes, prior insulin use.

( 2.2 , 2.3 , 2.4 ) Administer subcutaneously once daily at any time of day, but at the same time every day.

( 2.2 ) Rotate injection sites into the abdominal area, thigh, or deltoid to reduce the risk of lipodystrophy and localized cutaneous amyloidosis.

( 2.1 ) Closely monitor glucose when converting to BASAGLAR and during initial weeks thereafter.

( 2.2 ) Do not dilute or mix with any other insulin or solution.

( 2.1 ) 2.1 Important Administration Instructions Always check insulin labels before administration [see Warnings and Precautions ( 5.4 )] .

Visually inspect BASAGLAR prefilled pens for particulate matter and discoloration prior to administration.

Only use if the solution is clear and colorless with no visible particles.

Administer BASAGLAR subcutaneously into the abdominal area, thigh, or deltoid, and rotate injection sites within the same region from one injection to the next to reduce the risk of lipodystrophy and localized cutaneous amyloidosis.

Do not inject into areas of lipodystrophy or localized cutaneous amyloidosis [see Warnings and Precautions ( 5.2 ) and Adverse Reactions ( 6 )] .

During changes to a patient’s insulin regimen, increase the frequency of blood glucose monitoring [see Warnings and Precautions ( 5.2 )] .

Use BASAGLAR with caution in patients with visual impairment that may rely on audible clicks to dial their dose.

Do not dilute or mix BASAGLAR with any other insulin or solution.

Do not administer intravenously or via an insulin pump.

2.2 General Dosing Instructions In patients with type 1 diabetes, BASAGLAR must be used concomitantly with short-acting insulin.

Inject BASAGLAR subcutaneously once daily at any time of day but at the same time every day.

Individualize and adjust the dosage of BASAGLAR based on the individual’s metabolic needs, blood glucose monitoring results and glycemic control goal.

Dosage adjustments may be needed with changes in physical activity, changes in meal patterns (i.e., macronutrient content or timing of food intake), during acute illness, or changes in renal or hepatic function.

Dosage adjustments should only be made under medical supervision with appropriate glucose monitoring [see Warnings and Precautions ( 5.2 )] .

The BASAGLAR prefilled pens each dials in 1 unit increments and delivers a maximum dose of 80 units per injection.

2.3 Initiation of BASAGLAR Therapy The recommended starting dose of BASAGLAR in patients with type 1 diabetes should be approximately one-third of the total daily insulin requirements.

Short- or rapid-acting, pre-meal insulin should be used to satisfy the remainder of the daily insulin requirements.

The recommended starting dose of BASAGLAR in patients with type 2 diabetes is 0.2 units/kg or up to 10 units once daily.

2.4 Changing to BASAGLAR from Other Insulin Therapies If changing patients from another insulin glargine product, 100 units/mL, to BASAGLAR, the dose of BASAGLAR should be the same as the other insulin glargine product, 100 units/mL.

If changing patients from a once-daily insulin glargine product, 300 units/mL, to once-daily BASAGLAR, the recommended initial BASAGLAR dosage is 80% of the insulin glargine product, 300 units/mL [see Warnings and Precautions ( 5.2 )] .

If changing from a treatment regimen with an intermediate- or long-acting insulin to a regimen with BASAGLAR, a change in the dose of the basal insulin may be required.

If changing patients from twice-daily NPH insulin to once-daily BASAGLAR, the recommended initial BASAGLAR dosage is 80% of the total daily NPH dosage [see Warnings and Precautions ( 5.2 )] .

ARIPiprazole 2 MG Oral Tablet

Generic Name: ARIPIPRAZOLE
Brand Name: Aripiprazole
  • Substance Name(s):
  • ARIPIPRAZOLE

DRUG INTERACTIONS

7 Dosage adjustment due to drug interactions ( 7.1 ): Factors Dosage Adjustments for Aripiprazole Known CYP2D6 Poor Metabolizers Administer half of usual dose Known CYP2D6 Poor Metabolizers and strong CYP3A4 inhibitors Administer a quarter of usual dose Strong CYP2D6 or CYP3A4 inhibitors Administer half of usual dose Strong CYP2D6 and CYP3A4 inhibitors Administer a quarter of usual dose Strong CYP3A4 inducers Double usual dose over 1 to 2 weeks 7.1 Drugs Having Clinically Important Interactions with Aripiprazole Table 25: Clinically Important Drug Interactions with Aripiprazole: Concomitant Drug Name or Drug Class Clinical Rationale Clinical Recommendation Strong CYP3A4 Inhibitors (e.g., itraconazole, clarithromycin) or strong CYP2D6 inhibitors (e.g., quinidine, fluoxetine, paroxetine) The concomitant use of aripiprazole with strong CYP 3A4 or CYP2D6 inhibitors increased the exposure of aripiprazole compared to the use of aripiprazole alone [see Clinical Pharmacology (12.3) ] .

With concomitant use of aripiprazole with a strong CYP3A4 inhibitor or CYP2D6 inhibitor, reduce the aripiprazole dosage [see Dosage and Administration (2.7) ] .

Strong CYP3A4 Inducers (e.g., carbamazepine, rifampin) The concomitant use of aripiprazole and carbamazepine decreased the exposure of aripiprazole compared to the use of aripiprazole alone [see Clinical Pharmacology (12.3) ] .

With concomitant use of aripiprazole with a strong CYP3A4 inducer, consider increasing the aripiprazole dosage [see Dosage and Administration (2.7) ] .

Antihypertensive Drugs Due to its alpha adrenergic antagonism, aripiprazole has the potential to enhance the effect of certain antihypertensive agents.

Monitor blood pressure and adjust dose accordingly [see Warnings and Precautions (5.8) ] .

Benzodiazepines (e.g., lorazepam) The intensity of sedation was greater with the combination of oral aripiprazole and lorazepam as compared to that observed with aripiprazole alone.

The orthostatic hypotension observed was greater with the combination as compared to that observed with lorazepam alone [see Warnings and Precautions (5.8) ].

Monitor sedation and blood pressure.

Adjust dose accordingly.

7.2 Drugs Having No Clinically Important Interactions with Aripiprazole Based on pharmacokinetic studies, no dosage adjustment of aripiprazole is required when administered concomitantly with famotidine, valproate, lithium, lorazepam.

In addition, no dosage adjustment is necessary for substrates of CYP2D6 (e.g., dextromethorphan, fluoxetine, paroxetine, or venlafaxine), CYP2C9 (e.g., warfarin), CYP2C19 (e.g., omeprazole, warfarin, escitalopram), or CYP3A4 (e.g., dextromethorphan) when co-administered with aripiprazole.

Additionally, no dosage adjustment is necessary for valproate, lithium, lamotrigine, lorazepam, or sertraline when co-administered with aripiprazole [see Clinical Pharmacology (12.3) ] .

OVERDOSAGE

10 MedDRA terminology has been used to classify the adverse reactions.

10.1 Human Experience In clinical trials and in post-marketing experience, adverse reactions of deliberate or accidental overdosage with oral aripiprazole have been reported worldwide.

These include overdoses with oral aripiprazole alone and in combination with other substances.

No fatality was reported with aripiprazole alone.

The largest known dose with a known outcome involved acute ingestion of 1260 mg of oral aripiprazole (42 times the maximum recommended daily dose) by a patient who fully recovered.

Deliberate or accidental overdosage was also reported in children (age 12 years and younger) involving oral aripiprazole ingestions up to 195 mg with no fatalities.

Common adverse reactions (reported in at least 5% of all overdose cases) reported with oral aripiprazole overdosage (alone or in combination with other substances) include vomiting, somnolence, and tremor.

Other clinically important signs and symptoms observed in one or more patients with aripiprazole overdoses (alone or with other substances) include acidosis, aggression, aspartate aminotransferase increased, atrial fibrillation, bradycardia, coma, confusional state, convulsion, blood creatine phosphokinase increased, depressed level of consciousness, hypertension, hypokalemia, hypotension, lethargy, loss of consciousness, QRS complex prolonged, QT prolonged, pneumonia aspiration, respiratory arrest, status epilepticus, and tachycardia.

10.2 Management of Overdosage No specific information is available on the treatment of overdose with aripiprazole.

An electrocardiogram should be obtained in case of overdosage and if QT interval prolongation is present, cardiac monitoring should be instituted.

Otherwise, management of overdose should concentrate on supportive therapy, maintaining an adequate airway, oxygenation and ventilation, and management of symptoms.

Close medical supervision and monitoring should continue until the patient recovers.

Charcoal: In the event of an overdose of aripiprazole, an early charcoal administration may be useful in partially preventing the absorption of aripiprazole.

Administration of 50 g of activated charcoal, one hour after a single 15 mg oral dose of aripiprazole, decreased the mean AUC and C max of aripiprazole by 50%.

Hemodialysis: Although there is no information on the effect of hemodialysis in treating an overdose with aripiprazole, hemodialysis is unlikely to be useful in overdose management since aripiprazole is highly bound to plasma proteins.

DESCRIPTION

11 Aripiprazole, USP is an atypical antipsychotic drug that is available as aripiprazole tablets, USP.

Aripiprazole, USP is 7-[4-[4-(2,3-dichlorophenyl)-1-piperazinyl]butoxy]-3,4-dihydrocarbostyril.

The molecular formula is C 23 H 27 Cl 2 N 3 O 2 and its molecular weight is 448.38.

The chemical structure is: Aripiprazole tablets, USP are available in 2 mg, 5 mg, 10 mg, 15 mg, 20 mg, and 30 mg strengths.

Inactive ingredients include croscarmellose sodium, magnesium stearate, microcrystalline cellulose and tartaric acid.

The 2 mg and 5 mg tablets contain FD&C Blue No.

2 indigo carmine aluminum lake (30% to 36%).

The 2 mg and 15 mg tablets contain ferric oxide yellow.

The 10 mg and 30 mg tablets contain FD&C Red No.

40 allura red AC aluminum lake (15% to 17%).

1

CLINICAL STUDIES

14 Efficacy of the oral formulations of aripiprazole was established in the following adequate and well-controlled trials: Four short-term trials and one maintenance trial in adult patients and one short-term trial in adolescents (ages 13 to 17 years) with schizophrenia [see Clinical Studies (14.1) ] One maintenance monotherapy trial and in one maintenance adjunctive trial in adult patients with bipolar I disorder [see Clinical Studies (14.2) ] Two short-term trials in pediatric patients (ages 6 to 17 years) for the treatment of irritability associated with autistic disorder [see Clinical Studies (14.4) ] Two short-term trials in pediatric patients (ages 6 to 18 years) with Tourette’s disorder [see Clinical Studies (14.5) ] 14.1 Schizophrenia Adults The efficacy of aripiprazole in the treatment of schizophrenia was evaluated in five short-term (4-week and 6-week), placebo-controlled trials of acutely relapsed inpatients who predominantly met DSM-III/IV criteria for schizophrenia.

Four of the five trials were able to distinguish aripiprazole from placebo, but one study, the smallest, did not.

Three of these studies also included an active control group consisting of either risperidone (one trial) or haloperidol (two trials), but they were not designed to allow for a comparison of aripiprazole and the active comparators.

In the four positive trials for aripiprazole, four primary measures were used for assessing psychiatric signs and symptoms.

Efficacy was evaluated using the total score on the Positive and Negative Syndrome Scale (PANSS).

The PANSS is a 30 item scale that measures positive symptoms of schizophrenia (7 items), negative symptoms of schizophrenia (7 items), and general psychopathology (16 items), each rated on a scale of 1 (absent) to 7 (extreme); total PANSS scores range from 30 to 210.

The Clinical Global Impression (CGI) assessment reflects the impression of a skilled observer, fully familiar with the manifestations of schizophrenia, about the overall clinical state of the patient.

In a 4-week trial (n=414) comparing two fixed doses of aripiprazole (15 or 30 mg/day) to placebo, both doses of aripiprazole were superior to placebo in the PANSS total score (Study 1 in Table 26), PANSS positive subscale, and CGI-severity score.

In addition, the 15 mg dose was superior to placebo in the PANSS negative subscale.

In a 4-week trial (n=404) comparing two fixed doses of aripiprazole (20 or 30 mg/day) to placebo, both doses of aripiprazole were superior to placebo in the PANSS total score (Study 2 in Table 26), PANSS positive subscale, PANSS negative subscale, and CGI-severity score.

In a 6-week trial (n=420) comparing three fixed doses of aripiprazole (10, 15, or 20 mg/day) to placebo, all three doses of aripiprazole were superior to placebo in the PANSS total score (Study 3 in Table 26), PANSS positive subscale, and the PANSS negative subscale.

In a 6-week trial (n=367) comparing three fixed doses of aripiprazole (2, 5, or 10 mg/day) to placebo, the 10 mg dose of aripiprazole was superior to placebo in the PANSS total score (Study 4 in Table 26), the primary outcome measure of the study.

The 2 and 5 mg doses did not demonstrate superiority to placebo on the primary outcome measure.

Thus, the efficacy of 10 mg, 15 mg, 20 mg, and 30 mg daily doses was established in two studies for each dose.

Among these doses, there was no evidence that the higher dose groups offered any advantage over the lowest dose group of these studies.

An examination of population subgroups did not reveal any clear evidence of differential responsiveness on the basis of age, gender, or race.

A longer-term trial enrolled 310 inpatients or outpatients meeting DSM-IV criteria for schizophrenia who were, by history, symptomatically stable on other antipsychotic medications for periods of 3 months or longer.

These patients were discontinued from their antipsychotic medications and randomized to aripiprazole 15 mg/day or placebo for up to 26 weeks of observation for relapse.

Relapse during the double-blind phase was defined as CGI-Improvement score of ≥5 (minimally worse), scores ≥5 (moderately severe) on the hostility or uncooperativeness items of the PANSS, or ≥20% increase in the PANSS total score.

Patients receiving aripiprazole 15 mg/day experienced a significantly longer time to relapse over the subsequent 26 weeks compared to those receiving placebo (Study 5 in Figure 6).

Pediatric Patients The efficacy of aripiprazole in the treatment of schizophrenia in pediatric patients (13 to 17 years of age) was evaluated in one 6-week, placebo-controlled trial of outpatients who met DSM-IV criteria for schizophrenia and had a PANSS score ≥70 at baseline.

In this trial (n=302) comparing two fixed doses of aripiprazole (10 mg/day or 30 mg/day) to placebo, aripiprazole was titrated starting from 2 mg/day to the target dose in 5 days in the 10 mg/day treatment arm and in 11 days in the 30 mg/day treatment arm.

Both doses of aripiprazole were superior to placebo in the PANSS total score (Study 6 in Table 26), the primary outcome measure of the study.

The 30 mg/day dosage was not shown to be more efficacious than the 10 mg/day dose.

Although maintenance efficacy in pediatric patients has not been systematically evaluated, maintenance efficacy can be extrapolated from adult data along with comparisons of aripiprazole pharmacokinetic parameters in adult and pediatric patients.

Table 26: Schizophrenia Studies Study Number Treatment Group Primary Efficacy Measure: PANSS Mean Baseline Score (SD) LS Mean Change from Baseline (SE) Placebo-subtracted Difference * (95% CI) Study 1 Aripiprazole (15 mg/day) † 98.5 (17.2) -15.5 (2.40) -12.6 (-18.9, -6.2) Aripiprazole (30 mg/day) † 99.0 (19.2) -11.4 (2.39) -8.5 (-14.8, -2.1) Placebo 100.2 (16.5) -2.9 (2.36) — Study 2 Aripiprazole (20 mg/day) † 92.6 (19.5) -14.5 (2.23) -9.6 (-15.4, -3.8) Aripiprazole (30 mg/day) † 94.2 (18.5) -13.9 (2.24) -9.0 (-14.8, -3.1) Placebo 94.3 (18.5) -5.0 (2.17) — Study 3 Aripiprazole (10 mg/day) † 92.7 (19.5) -15.0 (2.38) -12.7 (-19.00, -6.41) Aripiprazole (15 mg/day) † 93.2 (21.6) -11.7 (2.38) -9.4 (-15.71, -3.08) Aripiprazole (20 mg/day) † 92.5 (20.9) -14.4 (2.45) -12.1 (-18.53, -5.68) Placebo 92.3 (21.8) -2.3 (2.35) — Study 4 Aripiprazole (2 mg/day) 90.7 (14.5) -8.2 (1.90) -2.9 (-8.29, 2.47) Aripiprazole (5 mg/day) 92.0 (12.6) -10.6 (1.93) -5.2 (-10.7, 0.19) Aripiprazole (10 mg/day) † 90.0 (11.9) -11.3 (1.88) -5.9 (-11.3, -0.58) Placebo 90.8 (13.3) -5.3 (1.97) — Study 6 (Pediatric, 13 to 17 years) Aripiprazole (10 mg/day) † 93.6 (15.7) -26.7 (1.91) -5.5 (-10.7, -0.21) Aripiprazole (30 mg/day) † 94.0 (16.1) -28.6 (1.92) -7.4 (-12.7, -2.13) Placebo 94.6 (15.6) -21.2 (1.93) — SD: standard deviation; SE: standard error; LS Mean: least-squares mean; CI: unadjusted confidence interval.

*Difference (drug minus placebo) in least-squares mean change from baseline.

† Doses statistically significantly superior to placebo.

Figure 6: Kaplan-Meier Estimation of Cumulative Proportion of Patients with Relapse (Schizophrenia Study 5) 1 14.2 Bipolar Disorder Maintenance Treatment of Bipolar I Disorder Monotherapy Maintenance Therapy A maintenance trial was conducted in adult patients meeting DSM-IV criteria for bipolar I disorder with a recent manic or mixed episode who had been stabilized on open-label aripiprazole and who had maintained a clinical response for at least 6 weeks.

The first phase of this trial was an open-label stabilization period in which inpatients and outpatients were clinically stabilized and then maintained on open-label aripiprazole (15 or 30 mg/day, with a starting dose of 30 mg/day) for at least 6 consecutive weeks.

One hundred sixty-one outpatients were then randomized in a double-blind fashion, to either the same dose of aripiprazole they were on at the end of the stabilization and maintenance period or placebo and were then monitored for manic or depressive relapse.

During the randomization phase, aripiprazole was superior to placebo on time to the number of combined affective relapses (manic plus depressive), the primary outcome measure for this study (Study 7 in Figure 7).

A total of 55 mood events were observed during the double-blind treatment phase.

Nineteen were from the aripiprazole group and 36 were from the placebo group.

The number of observed manic episodes in the aripiprazole group (6) were fewer than that in the placebo group (19), while the number of depressive episodes in the aripiprazole group (9) was similar to that in the placebo group (11).

An examination of population subgroups did not reveal any clear evidence of differential responsiveness on the basis of age and gender; however, there were insufficient numbers of patients in each of the ethnic groups to adequately assess inter-group differences.

Figure 7: Kaplan-Meier Estimation of Cumulative Proportion of Patients with Relapse (Bipolar Study 7) Adjunctive Maintenance Therapy An adjunctive maintenance trial was conducted in adult patients meeting DSM-IV criteria for bipolar I disorder with a recent manic or mixed episode.

Patients were initiated on open-label lithium (0.6 mEq/L to 1.0 mEq/L) or valproate (50 mcg/mL to 125 mcg/mL) at therapeutic serum levels, and remained on stable doses for 2 weeks.

At the end of 2 weeks, patients demonstrating inadequate response (Y-MRS total score ≥16 and ≤35% improvement on the Y-MRS total score) to lithium or valproate received aripiprazole with a starting dose of 15 mg/day with the option to increase to 30 mg or reduce to 10 mg as early as day 4, as adjunctive therapy with open-label lithium or valproate.

Prior to randomization, patients on the combination of single-blind aripiprazole and lithium or valproate were required to maintain stability (Y-MRS and MADRS total scores ≤12) for 12 consecutive weeks.

Three hundred thirty-seven patients were then randomized in a double-blind fashion, to either the same dose of aripiprazole they were on at the end of the stabilization period or placebo plus lithium or valproate and were then monitored for manic, mixed, or depressive relapse for a maximum of 52 weeks.

Aripiprazole was superior to placebo on the primary endpoint, time from randomization to relapse to any mood event (Study 8 in Figure 8).

A mood event was defined as hospitalization for a manic, mixed, or depressive episode, study discontinuation due to lack of efficacy accompanied by Y-MRS score >16 and/or a MADRS >16, or an SAE of worsening disease accompanied by Y-MRS score >16 and/or a MADRS >16.

A total of 68 mood events were observed during the double-blind treatment phase.

Twenty-five were from the aripiprazole group and 43 were from the placebo group.

The number of observed manic episodes in the aripiprazole group (7) were fewer than that in the placebo group (19), while the number of depressive episodes in the aripiprazole group (14) was similar to that in the placebo group (18).

The Kaplan-Meier curves of the time from randomization to relapse to any mood event during the 52-week, double-blind treatment phase for aripiprazole and placebo groups are shown in Figure 8.

An examination of population subgroups did not reveal any clear evidence of differential responsiveness on the basis of age and gender; however, there were insufficient numbers of patients in each of the ethnic groups to adequately assess inter-group differences.

Figure 8: Kaplan-Meier Estimation of Cumulative Proportion of Patients with Relapse to Any Mood Event (Bipolar Study 8) 1 1 14.4 Irritability Associated with Autistic Disorder Pediatric Patients The efficacy of aripiprazole in the treatment of irritability associated with autistic disorder was established in two 8-week, placebo-controlled trials in pediatric patients (6 to 17 years of age) who met the DSM-IV criteria for autistic disorder and demonstrated behaviors such as tantrums, aggression, self-injurious behavior, or a combination of these problems.

Over 75% of these patients were under 13 years of age.

Efficacy was evaluated using two assessment scales: The Aberrant Behavior Checklist (ABC) and the Clinical Global Impression-Improvement (CGI-I) scale.

The primary outcome measure in both trials was the change from baseline to endpoint in the Irritability subscale of the ABC (ABC-I).

The ABC-I subscale measured symptoms of irritability in autistic disorder.

The results of these trials are as follows: In one of the 8-week, placebo-controlled trials, children and adolescents with autistic disorder (n=98), aged 6 to 17 years, received daily doses of placebo or aripiprazole 2 mg/day to 15 mg/day.

Aripiprazole, starting at 2 mg/day with increases allowed up to 15 mg/day based on clinical response, significantly improved scores on the ABC-I subscale and on the CGI-I scale compared with placebo.

The mean daily dose of aripiprazole at the end of 8-week treatment was 8.6 mg/day (Study 1 in Table 29).

In the other 8-week, placebo-controlled trial in children and adolescents with autistic disorder (n=218), aged 6 to 17 years, three fixed doses of aripiprazole (5 mg/day, 10 mg/day, or 15 mg/day) were compared to placebo.

Aripiprazole dosing started at 2 mg/day and was increased to 5 mg/day after one week.

After a second week, it was increased to 10 mg/day for patients in the 10 and 15 mg dose arms, and after a third week, it was increased to 15 mg/day in the 15 mg/day treatment arm (Study 2 in Table 29).

All three doses of aripiprazole significantly improved scores on the ABC-I subscale compared with placebo.

Table 29: Irritability Associated with Autistic Disorder Studies (Pediatric) Study Number Treatment Group Primary Efficacy Measure: ABC-I Mean Baseline Score (SD) LS Mean Change from Baseline (SE) Placebo-subtracted Difference * (95% CI) Study 1 Aripiprazole (2 mg/day to 15 mg/day) † 29.6 (6.37) -12.9 (1.44) -7.9 (-11.7, -4.1) Placebo 30.2 (6.52) -5.0 (1.43) — Study 2 Aripiprazole (5 mg/day) † Aripiprazole (10 mg/day) † Aripiprazole (15 mg/day) 28.6 (7.56) 28.2 (7.36) 28.9 (6.41) -12.4 (1.36) -13.2 (1.25) -14.4 (1.31) -4.0 (-7.7, -0.4) -4.8 (-8.4, -1.3) -6.0 (-9.6, -2.3) Placebo 28.0 (6.89) -8.4 (1.39) — SD: standard deviation; SE: standard error; LS Mean: least-squares mean; CI: unadjusted confidence interval.

* Difference (drug minus placebo) in least-squares mean change from baseline.

† Doses statistically significantly superior to placebo.

14.5 Tourette’s Disorder Pediatric Patients The efficacy of aripiprazole in the treatment of Tourette’s disorder was established in one 8-week (7 to 17 years of age) and one 10-week (6 to 18 years of age), placebo-controlled trials in pediatric patients (6 to 18 years of age) who met the DSM-IV criteria for Tourette’s disorder and had a Total Tic score (TTS) ≥ 20 to 22 on the Yale Global Tic Severity Scale (YGTSS).

The YGTSS is a fully validated scale designed to measure current tic severity.

Efficacy was evaluated using two assessment scales: 1) the Total Tic score (TTS) of the YGTSS and 2) the Clinical Global Impressions Scale for Tourette’s Syndrome (CGI-TS), a clinician-determined summary measure that takes into account all available patient information.

Over 65% of these patients were under 13 years of age.

The primary outcome measure in both trials was the change from baseline to endpoint in the TTS of the YGTSS.

Ratings for the TTS are made along 5 different dimensions on a scale of 0 to 5 for motor and vocal tics each.

Summation of these 10 scores provides a TTS (i.e., 0 to 50).

The results of these trials are as follows: In the 8-week, placebo-controlled, fixed-dose trial, children and adolescents with Tourette’s disorder (n=133), aged 7 to 17 years, were randomized 1:1:1 to low dose aripiprazole, high dose aripiprazole, or placebo.

The target doses for the low and high dose aripiprazole groups were based on weight.

Patients <50 kg in the low dose aripiprazole group started at 2 mg per day with a target dose of 5 mg per day after 2 days.

Patients ≥50 kg in the low dose aripiprazole group, started at 2 mg per day increased to 5 mg per day after 2 days, with a subsequent increase to a target dose of 10 mg per day at day 7.

Patients < 50 kg in the high dose aripiprazole group started at 2 mg per day increased to 5 mg per day after 2 days, with a subsequent increase to a target dose of 10 mg per day at day 7.

Patients ≥ 50 kg in the high dose aripiprazole group, started at 2 mg per day increased to 5 mg per day after 2 days, with a subsequent increase to a dose of 10 mg per day at day 7 and were allowed weekly increases of 5 mg per day up to a target dose 20 mg per day at Day 21.

Aripiprazole (both high and low dose groups) demonstrated statistically significantly improved scores on the YGTSS TTS (Study 1 in Table 30) and on the CGI-TS scale compared with placebo.

The estimated improvements on the YGTSS TTS over the course of the study are displayed in Figure 9.

Figure 9: Least Square Means of Change from Baseline in YGTSS TTS by Week (Tourette’s Disorder Study 1) In the 10-week, placebo-controlled, flexible-dose trial in children and adolescents with Tourette’s disorder (n=61), aged 6 to 18 years, patients received daily doses of placebo or aripiprazole, starting at 2 mg/day with increases allowed up to 20 mg/day based on clinical response.

Aripiprazole demonstrated statistically significantly improved scores on the YGTSS TTS scale compared with placebo (Study 2 in Table 30).

The mean daily dose of aripiprazole at the end of 10-week treatment was 6.54 mg/day.

Table 30: Tourette’s Disorder Studies (Pediatric) Study Number Treatment Group Primary Efficacy Measure: YGTSS TTS Mean Baseline Score (SD) LS Mean Change from Baseline (SE) Placebo-subtracted Difference * (95% CI) Study 1 Aripiprazole (low dose) † 29.2 (5.63) -13.4 (1.59) -6.3 (-10.2, -2.3) Aripiprazole (high dose) † 31.2 (6.40) -16.9 (1.61) -9.9 (-13.8, -5.9) Placebo 30.7 (5.95) -7.1 (1.55) — Study 2 Aripiprazole (2 to 20 mg/day) † 28.3 (5.51) -15.0 (1.51) -5.3 (-9.8, -0.9) Placebo 29.5 (5.60) -9.6 (1.64) — SD: standard deviation; SE: standard error; LS Mean: least-squares mean; CI: unadjusted confidence interval.

*Difference (drug minus placebo) in least-squares mean change from baseline.

† Doses statistically significantly superior to placebo.

1

HOW SUPPLIED

16 /STORAGE AND HANDLING 16.1 How Supplied Aripiprazole tablets USP, 2 mg, are supplied as green to light green colored, mosaic appearance, modified rectangle shaped, uncoated tablet with debossing “AN896” and “2” on one side and plain on the other side.

They are available as follows: Bottles of 30: NDC 65162-896-03 Bottles of 90: NDC 65162-896-09 Bottles of 500: NDC 65162-896-50 Bottles of 1000: NDC 65162-896-11 Aripiprazole tablets USP, 5 mg, are supplied as blue to light blue colored, mosaic appearance, modified rectangle shaped, uncoated tablet with debossing “AN897” and “5” on one side and plain on the other side.

They are available as follows: Bottles of 30: NDC 65162-897-03 Bottles of 90: NDC 65162-897-09 Bottles of 500: NDC 65162-897-50 Bottles of 1000: NDC 65162-897-11 Aripiprazole tablets USP, 10 mg, are supplied as pink to light pink colored, mosaic appearance, modified rectangle shaped, uncoated tablet with debossing “AN898” and “10” on one side and plain on the other side.

They are available as follows: Bottles of 30: NDC 65162-898-03 Bottles of 90: NDC 65162-898-09 Bottles of 500: NDC 65162-898-50 Bottles of 1000: NDC 65162-898-11 Aripiprazole tablets USP, 15 mg, are supplied as yellow to light yellow colored, mosaic appearance, round shaped, uncoated tablet with debossing “AN899” and “15” on one side and plain on the other side.

They are available as follows: Bottles of 30: NDC 65162-899-03 Bottles of 90: NDC 65162-899-09 Bottles of 500: NDC 65162-899-50 Bottles of 1000: NDC 65162-899-11 Aripiprazole tablets USP, 20 mg, are supplied as white to off-white, round shaped, uncoated tablet with debossing “AN901” and “20” on one side and plain on the other side.

They are available as follows: Bottles of 30: NDC 65162-901-03 Bottles of 90: NDC 65162-901-09 Bottles of 500: NDC 65162-901-50 Bottles of 1000: NDC 65162-901-11 Aripiprazole tablets USP, 30 mg, are supplied as pink to light pink colored, mosaic appearance, round shaped, uncoated tablet with debossing “AN902” and “30” on one side and plain on the other side.

They are available as follows: Bottles of 30: NDC 65162-902-03 Bottles of 90: NDC 65162-902-09 Bottles of 500: NDC 65162-902-50 Bottles of 1000: NDC 65162-902-11 16.2 Storage Tablets Store at 20° to 25°C (68° to 77°F); excursions permitted between 15° to 30°C (59° to 86°F) [see USP Controlled Room Temperature] in tightly closed containers.

GERIATRIC USE

8.5 Geriatric Use No dosage adjustment is recommended for elderly patients [see Boxed Warning , Warnings and Precautions (5.1) , and Clinical Pharmacology (12.3) ] .

Of the 13,543 patients treated with oral aripiprazole in clinical trials, 1,073 (8%) were ≥65 years old and 799 (6%) were ≥75 years old.

Placebo-controlled studies of oral aripiprazole in schizophrenia, or other indications did not include sufficient numbers of patients aged 65 and over to determine whether they respond differently from younger patients.

Aripiprazole is not approved for the treatment of patients with psychosis associated with Alzheimer’s disease [see Boxed Warning and Warnings and Precautions (5.1) ] .

DOSAGE FORMS AND STRENGTHS

3 Aripiprazole Tablets, USP are available as described in Table 3.

Table 3: Aripiprazole Tablet Presentations TabletStrength TabletColor/Shape TabletMarkings 2 mg green to light green mosaic appearance, modified rectangle shaped “AN896” and “2” 5 mg blue to light blue mosaic appearance, modified rectangle shaped “AN897” and “5” 10 mg pink to light pink mosaic appearance, modified rectangle shaped “AN898” and “10” 15 mg yellow to light yellow mosaic appearance, round shaped “AN899” and “15” 20 mg white to off-white, round shaped “AN901” and “20” 30 mg pink to light pink mosaic appearance, round shaped “AN902” and “30” Tablets: 2 mg, 5 mg, 10 mg, 15 mg, 20 mg, and 30 mg.

( 3 )

MECHANISM OF ACTION

12.1 Mechanism of Action The mechanism of action of aripiprazole in schizophrenia, is unclear.

However, the efficacy of aripiprazole in the listed indications could be mediated through a combination of partial agonist activity at D 2 and 5-HT 1A receptors and antagonist activity at 5-HT 2A receptors.

INDICATIONS AND USAGE

1 Aripiprazole Oral Tablets are indicated for the treatment of: Schizophrenia Irritability Associated with Autistic Disorder Treatment of Tourette’s Disorder Aripiprazole tablets are an atypical antipsychotic.

The oral formulations are indicated for: Schizophrenia.

( 14.1 ) Irritability Associated with Autistic Disorder.

( 14.4 ) Treatment of Tourette’s disorder.

( 14.5 )

PEDIATRIC USE

8.4 Pediatric Use The pharmacokinetics of aripiprazole and dehydro-aripiprazole in pediatric patients, 10 to 17 years of age, were similar to those in adults after correcting for the differences in body weight [see Clinical Pharmacology (12.3) ] .

Schizophrenia Safety and effectiveness in pediatric patients with schizophrenia were established in a 6-week, placebo-controlled clinical trial in 202 pediatric patients aged 13 to 17 years [see Dosage and Administration (2.1) , Adverse Reactions (6.1) , and Clinical Studies (14.1) ] .

Although maintenance efficacy in pediatric patients has not been systematically evaluated, maintenance efficacy can be extrapolated from adult data along with comparisons of aripiprazole pharmacokinetic parameters in adult and pediatric patients.

Irritability Associated with Autistic Disorder Safety and effectiveness in pediatric patients demonstrating irritability associated with autistic disorder were established in two 8-week, placebo-controlled clinical trials in 212 pediatric patients aged 6 to 17 years [see Indications and Usage (1) , Dosage and Administration (2.4) , Adverse Reactions (6.1) , and Clinical Studies (14.4) ] .

A maintenance trial was conducted in pediatric patients (6 to 17 years of age) with irritability associated with autistic disorder.

The first phase of this trial was an open-label, flexibly dosed (aripiprazole 2 to 15 mg/day) phase in which patients were stabilized (defined as > 25% improvement on the ABC-I subscale, and a CGI-I rating of “much improved” or “very much improved”) on aripiprazole for 12 consecutive weeks.

Overall, 85 patients were stabilized and entered the second, 16-week, double-blind phase where they were randomized to either continue aripiprazole treatment or switch to placebo.

In this trial, the efficacy of aripiprazole for the maintenance treatment of irritability associated with autistic disorder was not established.

Tourette’s Disorder Safety and effectiveness of aripiprazole in pediatric patients with Tourette’s Disorder were established in one 8-week (aged 7 to 17 years) and one 10-week trial (aged 6 to 18 years) in 194 pediatric patients [see Dosage and Administration (2.5) , Adverse Reactions (6.1) , and Clinical Studies (14.5) ] .

Maintenance efficacy in pediatric patients has not been systematically evaluated.

Juvenile Animal Studies Aripiprazole in juvenile rats caused mortality, CNS clinical signs, impaired memory and learning, and delayed sexual maturation when administered at oral doses of 10, 20, 40 mg/kg/day from weaning (21 days old) through maturity (80 days old).

At 40 mg/kg/day, mortality, decreased activity, splayed hind limbs, hunched posture, ataxia, tremors and other CNS signs were observed in both genders.

In addition, delayed sexual maturation was observed in males.

At all doses and in a dose-dependent manner, impaired memory and learning, increased motor activity, and histopathology changes in the pituitary (atrophy), adrenals (adrenocortical hypertrophy), mammary glands (hyperplasia and increased secretion), and female reproductive organs (vaginal mucification, endometrial atrophy, decrease in ovarian corpora lutea) were observed.

The changes in female reproductive organs were considered secondary to the increase in prolactin serum levels.

A No Observed Adverse Effect Level (NOAEL) could not be determined and, at the lowest tested dose of 10 mg/kg/day, there is no safety margin relative to the systemic exposures (AUC 0-24 ) for aripiprazole or its major active metabolite in adolescents at the maximum recommended pediatric dose of 15 mg/day.

All drug-related effects were reversible after a 2-month recovery period, and most of the drug effects in juvenile rats were also observed in adult rats from previously conducted studies.

Aripiprazole in juvenile dogs (2 months old) caused CNS clinical signs of tremors, hypoactivity, ataxia, recumbency and limited use of hind limbs when administered orally for 6 months at 3, 10, 30 mg/kg/day.

Mean body weight and weight gain were decreased up to 18% in females in all drug groups relative to control values.

A NOAEL could not be determined and, at the lowest tested dose of 3 mg/kg/day, there is no safety margin relative to the systemic exposures (AUC 0-24 ) for aripiprazole or its major active metabolite in adolescents at the maximum recommended pediatric dose of 15 mg/day.

All drug-related effects were reversible after a 2-month recovery period.

BOXED WARNING

WARNING: INCREASED MORTALITY IN ELDERLY PATIENTS WITH DEMENTIA-RELATED PSYCHOSIS and SUICIDAL THOUGHTS AND BEHAVIORS WITH ANTIDEPRESSANT DRUGS Elderly patients with dementia-related psychosis treated with antipsychotic drugs are at an increased risk of death.

Aripiprazole is not approved for the treatment of patients with dementia-related psychosis [see Warnings and Precautions (5.1) ] .

Antidepressants increased the risk of suicidal thoughts and behavior in children, adolescents, and young adults in short-term studies.

These studies did not show an increase in the risk of suicidal thoughts and behavior with antidepressant use in patients over age 24 years; there was a reduction in risk with antidepressant use in patients aged 65 years and older [see Warnings and Precautions (5.3) ] .

In patients of all ages who are started on antidepressant therapy, monitor closely for worsening, and for emergence of suicidal thoughts and behaviors.

Advise families and caregivers of the need for close observation and communication with the prescriber [see Warnings and Precautions (5.3) ] .

WARNING: INCREASED MORTALITY IN ELDERLY PATIENTS WITH DEMENTIA-RELATED PSYCHOSIS and SUICIDAL THOUGHTS AND BEHAVIORS WITH ANTIDEPRESSANT DRUGS See full prescribing information for complete boxed warning.

Elderly patients with dementia-related psychosis treated with antipsychotic drugs are at an increased risk of death.

Aripiprazole is not approved for the treatment of patients with dementia-related psychosis.

( 5.1 ) Increased risk of suicidal thinking and behavior in children, adolescents, and young adults taking antidepressants.

Monitor for worsening and emergence of suicidal thoughts and behaviors.

( 5.3 )

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS Cerebrovascular Adverse Reactions in Elderly Patients with Dementia-Related Psychosis: Increased incidence of cerebrovascular adverse reactions (e.g., stroke, transient ischemic attack, including fatalities).

( 5.2 ) Neuroleptic Malignant Syndrome: Manage with immediate discontinuation and close monitoring.

( 5.4 ) Tardive Dyskinesia: Discontinue if clinically appropriate.

( 5.5 ) Metabolic Changes: Atypical antipsychotic drugs have been associated with metabolic changes that include hyperglycemia/diabetes mellitus, dyslipidemia, and body weight gain.

( 5.6 ) Hyperglycemia/Diabetes Mellitus: Monitor glucose regularly in patients with and at risk for diabetes.

( 5.6 ) Dyslipidemia: Undesirable alterations in lipid levels have been observed in patients treated with atypical antipsychotics.

( 5.6 ) Weight Gain: Weight gain has been observed with atypical antipsychotic use.

Monitor weight.

( 5.6 ) Pathological Gambling and Other Compulsive Behaviors: Consider dose reduction or discontinuation.

( 5.7 ) Orthostatic Hypotension: Monitor heart rate and blood pressure and warn patients with known cardiovascular or cerebrovascular disease, and risk of dehydration or syncope.

( 5.8 ) Leukopenia, Neutropenia, and Agranulocytosis: have been reported with antipsychotics including aripiprazole.

Patients with a history of a clinically significant low white blood cell count (WBC) or a drug-induced leukopenia/neutropenia should have their complete blood count (CBC) monitored frequently during the first few months of therapy and discontinuation of aripiprazole should be considered at the first sign of a clinically significant decline in WBC in the absence of other causative factors.

( 5.10 ) Seizures/Convulsions: Use cautiously in patients with a history of seizures or with conditions that lower the seizure threshold.

( 5.11 ) Potential for Cognitive and Motor Impairment: Use caution when operating machinery.

( 5.12 ) Suicide: The possibility of a suicide attempt is inherent in schizophrenia.

Closely supervise high-risk patients.

( 5.14 ) 5.1 Increased Mortality in Elderly Patients with Dementia-Related Psychosis Increased Mortality Elderly patients with dementia-related psychosis treated with antipsychotic drugs are at an increased risk of death.

Aripiprazole is not approved for the treatment of patients with dementia-related psychosis [see Boxed Warning ] .

Safety Experience in Elderly Patients with Psychosis Associated with Alzheimer’s Disease In three, 10-week, placebo-controlled studies of aripiprazole in elderly patients with psychosis associated with Alzheimer’s disease (n=938; mean age: 82.4 years; range: 56 to 99 years), the adverse reactions that were reported at an incidence of ≥3% and aripiprazole incidence at least twice that for placebo were lethargy [placebo 2%, aripiprazole 5%], somnolence (including sedation) [placebo 3%, aripiprazole 8%], and incontinence (primarily, urinary incontinence) [placebo 1%, aripiprazole 5%], excessive salivation [placebo 0%, aripiprazole 4%], and lightheadedness [placebo 1%, aripiprazole 4%].

The safety and efficacy of aripiprazole in the treatment of patients with psychosis associated with dementia have not been established.

If the prescriber elects to treat such patients with aripiprazole, assess for the emergence of difficulty swallowing or excessive somnolence, which could predispose to accidental injury or aspiration [see Boxed Warning ] .

5.2 Cerebrovascular Adverse Events, Including Stroke In placebo-controlled clinical studies (two flexible dose and one fixed dose study) of dementia-related psychosis, there was an increased incidence of cerebrovascular adverse events (e.g., stroke, transient ischemic attack), including fatalities, in aripiprazole-treated patients (mean age: 84 years; range: 78 to 88 years).

In the fixed-dose study, there was a statistically significant dose response relationship for cerebrovascular adverse events in patients treated with aripiprazole.

Aripiprazole is not approved for the treatment of patients with dementia-related psychosis [see Boxed Warning ] .

5.3 Suicidal Thoughts and Behaviors in Children, Adolescents, and Young Adults Patients with major depressive disorder (MDD), both adult and pediatric, may experience worsening of their depression and/or the emergence of suicidal ideation and behavior (suicidality) or unusual changes in behavior, whether or not they are taking antidepressant medications, and this risk may persist until significant remission occurs.

Suicide is a known risk of depression and certain other psychiatric disorders, and these disorders themselves are the strongest predictors of suicide.

There has been a long-standing concern, however, that antidepressants may have a role in inducing worsening of depression and the emergence of suicidality in certain patients during the early phases of treatment.

Pooled analyses of short-term, placebo-controlled trials of antidepressant drugs (SSRIs and others) showed that these drugs increase the risk of suicidal thinking and behavior (suicidality) in children, adolescents, and young adults (ages 18 to 24 years) with MDD and other psychiatric disorders.

Short-term studies did not show an increase in the risk of suicidality with antidepressants compared to placebo in adults beyond age 24 years; there was a reduction with antidepressants compared to placebo in adults aged 65 years and older.

The pooled analyses of placebo-controlled trials in children and adolescents with MDD, Obsessive Compulsive Disorder (OCD), or other psychiatric disorders included a total of 24 short-term trials of 9 antidepressant drugs in over 4400 patients.

The pooled analyses of placebo-controlled trials in adults with MDD or other psychiatric disorders included a total of 295 short-term trials (median duration of 2 months) of 11 antidepressant drugs in over 77,000 patients.

There was considerable variation in risk of suicidality among drugs, but a tendency toward an increase in the younger patients for almost all drugs studied.

There were differences in absolute risk of suicidality across the different indications, with the highest incidence in MDD.

The risk differences (drug vs.

placebo), however, were relatively stable within age strata and across indications.

These risk differences (drug-placebo difference in the number of cases of suicidality per 1000 patients treated) are provided in Table 5.

Table 5: Age Range Drug-Placebo Difference in Number of Cases of Suicidality per 1000 Patients Treated Increases Compared to Placebo < 18 14 additional cases 18 to 24 5 additional cases Decreases Compared to Placebo 25 to 64 1 fewer case ≥ 65 6 fewer cases No suicides occurred in any of the pediatric trials.

There were suicides in the adult trials, but the number was not sufficient to reach any conclusion about drug effect on suicide.

It is unknown whether the suicidality risk extends to longer-term use, i.e.

beyond several months.

However, there is substantial evidence from placebo-controlled maintenance trials in adults with depression that the use of antidepressants can delay the recurrence of depression.

All patients being treated with antidepressants for any indication should be monitored appropriately and observed closely for clinical worsening, suicidality, and unusual changes in behavior, especially during the initial few months of a course of drug therapy, or at times of dose changes, either increases or decreases.

The following symptoms, anxiety, agitation, panic attacks, insomnia, irritability, hostility, aggressiveness, impulsivity, akathisia (psychomotor restlessness), hypomania, and mania, have been reported in adult and pediatric patients being treated with antidepressants for MDD as well as for other indications, both psychiatric and nonpsychiatric.

Although a causal link between the emergence of such symptoms and either the worsening of depression and/or the emergence of suicidal impulses has not been established, there is concern that such symptoms may represent precursors to emerging suicidality.

Consideration should be given to changing the therapeutic regimen, including possibly discontinuing the medication, in patients whose depression is persistently worse, or who are experiencing emergent suicidality or symptoms that might be precursors to worsening depression or suicidality, especially if these symptoms are severe, abrupt in onset, or were not part of the patient’s presenting symptoms.

Families and caregivers of patients being treated with antidepressants for major depressive disorder or other indications, both psychiatric and nonpsychiatric, should be alerted about the need to monitor patients for the emergence of agitation, irritability, unusual changes in behavior, and the other symptoms described above, as well as the emergence of suicidality, and to report such symptoms immediately to healthcare providers.

Such monitoring should include daily observation by families and caregivers.

Prescriptions for aripiprazole should be written for the smallest quantity of tablets consistent with good patient management, in order to reduce the risk of overdose.

Screening Patients for Bipolar Disorder: A major depressive episode may be the initial presentation of bipolar disorder.

It is generally believed (though not established in controlled trials) that treating such an episode with an antidepressant alone may increase the likelihood of precipitation of a mixed/manic episode in patients at risk for bipolar disorder.

Whether any of the symptoms described above represent such a conversion is unknown.

However, prior to initiating treatment with an antidepressant, patients with depressive symptoms should be adequately screened to determine if they are at risk for bipolar disorder; such screening should include a detailed psychiatric history, including a family history of suicide, bipolar disorder, and depression.

It should be noted that aripiprazole is not approved for use in treating depression in the pediatric population.

5.4 Neuroleptic Malignant Syndrome (NMS) A potentially fatal symptom complex sometimes referred to as Neuroleptic Malignant Syndrome (NMS) may occur with administration of antipsychotic drugs, including aripiprazole.

Rare cases of NMS occurred during aripiprazole treatment in the worldwide clinical database.

Clinical manifestations of NMS are hyperpyrexia, muscle rigidity, altered mental status, and evidence of autonomic instability (irregular pulse or blood pressure, tachycardia, diaphoresis, and cardiac dysrhythmia).

Additional signs may include elevated creatine phosphokinase, myoglobinuria (rhabdomyolysis), and acute renal failure.

The diagnostic evaluation of patients with this syndrome is complicated.

In arriving at a diagnosis, it is important to exclude cases where the clinical presentation includes both serious medical illness (e.g., pneumonia, systemic infection) and untreated or inadequately treated extrapyramidal signs and symptoms (EPS).

Other important considerations in the differential diagnosis include central anticholinergic toxicity, heat stroke, drug fever, and primary central nervous system pathology.

The management of NMS should include: 1) immediate discontinuation of antipsychotic drugs and other drugs not essential to concurrent therapy; 2) intensive symptomatic treatment and medical monitoring; and 3) treatment of any concomitant serious medical problems for which specific treatments are available.

There is no general agreement about specific pharmacological treatment regimens for uncomplicated NMS.

If a patient requires antipsychotic drug treatment after recovery from NMS, the potential reintroduction of drug therapy should be carefully considered.

The patient should be carefully monitored, since recurrences of NMS have been reported.

5.5 Tardive Dyskinesia A syndrome of potentially irreversible, involuntary, dyskinetic movements may develop in patients treated with antipsychotic drugs.

Although the prevalence of the syndrome appears to be highest among the elderly, especially elderly women, it is impossible to rely upon prevalence estimates to predict, at the inception of antipsychotic treatment, which patients are likely to develop the syndrome.

Whether antipsychotic drug products differ in their potential to cause tardive dyskinesia is unknown.

The risk of developing tardive dyskinesia and the likelihood that it will become irreversible are believed to increase as the duration of treatment and the total cumulative dose of antipsychotic drugs administered to the patient increase.

However, the syndrome can develop, although much less commonly, after relatively brief treatment periods at low doses.

Tardive dyskinesia may remit, partially or completely, if antipsychotic treatment is withdrawn.

Antipsychotic treatment, itself, however, may suppress (or partially suppress) the signs and symptoms of the syndrome and, thereby, may possibly mask the underlying process.

The effect that symptomatic suppression has upon the long-term course of the syndrome is unknown.

Given these considerations, aripiprazole should be prescribed in a manner that is most likely to minimize the occurrence of tardive dyskinesia.

Chronic antipsychotic treatment should generally be reserved for patients who suffer from a chronic illness that (1) is known to respond to antipsychotic drugs and (2) for whom alternative, equally effective, but potentially less harmful treatments are not available or appropriate.

In patients who do require chronic treatment, the smallest dose and the shortest duration of treatment producing a satisfactory clinical response should be sought.

The need for continued treatment should be reassessed periodically.

If signs and symptoms of tardive dyskinesia appear in a patient on aripiprazole, drug discontinuation should be considered.

However, some patients may require treatment with aripiprazole despite the presence of the syndrome.

5.6 Metabolic Changes Atypical antipsychotic drugs have been associated with metabolic changes that include hyperglycemia/diabetes mellitus, dyslipidemia, and body weight gain.

While all drugs in the class have been shown to produce some metabolic changes, each drug has its own specific risk profile.

Hyperglycemia/Diabetes Mellitus Hyperglycemia, in some cases extreme and associated with ketoacidosis or hyperosmolar coma or death, has been reported in patients treated with atypical antipsychotics.

There have been reports of hyperglycemia in patients treated with aripiprazole [see Adverse Reactions (6.1 , 6.2 )] .

Assessment of the relationship between atypical antipsychotic use and glucose abnormalities is complicated by the possibility of an increased background risk of diabetes mellitus in patients with schizophrenia and the increasing incidence of diabetes mellitus in the general population.

Given these confounders, the relationship between atypical antipsychotic use and hyperglycemia-related adverse events is not completely understood.

However, epidemiological studies suggest an increased risk of hyperglycemia-related adverse reactions in patients treated with the atypical antipsychotics.

Because aripiprazole was not marketed at the time these studies were performed, it is not known if aripiprazole is associated with this increased risk.

Precise risk estimates for hyperglycemia-related adverse reactions in patients treated with atypical antipsychotics are not available.

Patients with an established diagnosis of diabetes mellitus who are started on atypical antipsychotics should be monitored regularly for worsening of glucose control.

Patients with risk factors for diabetes mellitus (e.g., obesity, family history of diabetes) who are starting treatment with atypical antipsychotics should undergo fasting blood glucose testing at the beginning of treatment and periodically during treatment.

Any patient treated with atypical antipsychotics should be monitored for symptoms of hyperglycemia including polydipsia, polyuria, polyphagia, and weakness.

Patients who develop symptoms of hyperglycemia during treatment with atypical antipsychotics should undergo fasting blood glucose testing.

In some cases, hyperglycemia has resolved when the atypical antipsychotic was discontinued; however, some patients required continuation of anti-diabetic treatment despite discontinuation of the suspect drug.

Adults In an analysis of 13 placebo-controlled monotherapy trials in adults, primarily with schizophrenia or another disorder, the mean change in fasting glucose in aripiprazole-treated patients (+4.4 mg/dL; median exposure 25 days; N=1057) was not significantly different than in placebo-treated patients (+2.5 mg/dL; median exposure 22 days; N=799).

Table 6 shows the proportion of aripiprazole-treated patients with normal and borderline fasting glucose at baseline (median exposure 25 days) that had treatment-emergent high fasting glucose measurements compared to placebo-treated patients (median exposure 22 days).

Table 6: Changes in Fasting Glucose from Placebo-Controlled Monotherapy Trials in Adult Patients Category Change (at least once) from Baseline Treatment Arm n/N % Fasting Glucose Normal to High (< 100 mg/dL to ≥ 126 mg/dL) Aripiprazole 31/822 3.8 Placebo 22/605 3.6 Borderline to High (≥ 100 mg/dL and < 126 mg/dL to ≥ 126 mg/dL) Aripiprazole 31/176 17.6 Placebo 13/142 9.2 At 24 weeks, the mean change in fasting glucose in aripiprazole-treated patients was not significantly different than in placebo-treated patients [+2.2 mg/dL (n=42) and +9.6 mg/dL (n=28), respectively].

Pediatric Patients and Adolescents In an analysis of two placebo-controlled trials in adolescents with schizophrenia (13 to 17 years) and pediatric patients with another disorder (10 to 17 years), the mean change in fasting glucose in aripiprazole-treated patients (+4.8 mg/dL; with a median exposure of 43 days; N=259) was not significantly different than in placebo-treated patients (+1.7 mg/dL; with a median exposure of 42 days; N=123).

In an analysis of two placebo-controlled trials in pediatric and adolescent patients with irritability associated with autistic disorder (6 to 17 years) with median exposure of 56 days, the mean change in fasting glucose in aripiprazole-treated patients (–0.2 mg/dL; N=83) was not significantly different than in placebo-treated patients (–0.6 mg/dL; N=33).

In an analysis of two placebo-controlled trials in pediatric and adolescent patients with Tourette’s disorder (6 to 18 years) with median exposure of 57 days, the mean change in fasting glucose in aripiprazole-treated patients (0.79 mg/dL; N=90) was not significantly different than in placebo-treated patients (–1.66 mg/dL; N=58).

Table 8 shows the proportion of patients with changes in fasting glucose levels from the pooled adolescent schizophrenia and other pediatric patients (median exposure of 42 to 43 days), from two placebo-controlled trials in pediatric patients (6 to 17 years) with irritability associated with autistic disorder (median exposure of 56 days), and from the two placebo-controlled trials in pediatric patients (6 to 18 year) with Tourette’s Disorder (median exposure 57 days).

Table 8: Changes in Fasting Glucose from Placebo-Controlled Trials in Pediatric and Adolescent Patients Category Change (at least once) from Baseline Indication Treatment Arm n/N % Fasting Glucose Normal to High (< 100 mg/dL to ≥ 126 mg/dL) Pooled Schizophrenia and Another Disorder Aripiprazole 2/236 0.8 Placebo 2/110 1.8 Irritability Associated with Autistic Disorder Aripiprazole 0/73 0 Placebo 0/32 0 Tourette’s Disorder Aripiprazole 3/88 3.4 Placebo 1/58 1.7 Fasting Glucose Borderline to High (≥ 100 mg/dL and < 126 mg/dL to ≥ 126 mg/dL) Pooled Schizophrenia and Another Disorder Aripiprazole 1/22 4.5 Placebo 0/12 0 Irritability Associated with Autistic Disorder Aripiprazole 0/9 0 Placebo 0/1 0 Tourette’s Disorder Aripiprazole 0/11 0 Placebo 0/4 0 At 12 weeks in the pooled adolescent schizophrenia and other pediatric disorder trials, the mean change in fasting glucose in aripiprazole-treated patients was not significantly different than in placebo-treated patients [+2.4 mg/dL (n=81) and +0.1 mg/dL (n=15), respectively].

Dyslipidemia Undesirable alterations in lipids have been observed in patients treated with atypical antipsychotics.

There were no significant differences between aripiprazole- and placebo-treated patients in the proportion with changes from normal to clinically significant levels for fasting/nonfasting total cholesterol, fasting triglycerides, fasting LDLs, and fasting/nonfasting HDLs.

Analyses of patients with at least 12 or 24 weeks of exposure were limited by small numbers of patients.

Adults Table 9 shows the proportion of adult patients, primarily from pooled schizophrenia and other monotherapy placebo-controlled trials, with changes in total cholesterol (pooled from 17 trials; median exposure 21 to 25 days), fasting triglycerides (pooled from eight trials; median exposure 42 days), fasting LDL cholesterol (pooled from eight trials; median exposure 39 to 45 days, except for placebo-treated patients with baseline normal fasting LDL measurements, who had median treatment exposure of 24 days) and HDL cholesterol (pooled from nine trials; median exposure 40 to 42 days).

Table 9: Changes in Blood Lipid Parameters from Placebo-Controlled Monotherapy Trials in Adults Treatment Arm n/N % Total Cholesterol Normal to High (< 200 mg/dL to ≥ 240 mg/dL) Aripiprazole 34/1357 2.5 Placebo 27/973 2.8 Fasting Triglycerides Normal to High (< 150 mg/dL to ≥ 200 mg/dL) Aripiprazole 40/539 7.4 Placebo 30/431 7.0 Fasting LDL Cholesterol Normal to High (< 100 mg/dL to ≥ 160 mg/dL) Aripiprazole 2/332 0.6 Placebo 2/268 0.7 HDL Cholesterol Normal to Low (≥ 40 mg/dL to < 40 mg/dL) Aripiprazole 121/1066 11.4 Placebo 99/794 12.5 In monotherapy trials in adults, the proportion of patients at 12 weeks and 24 weeks with changes from Normal to High in total cholesterol (fasting/nonfasting), fasting triglycerides, and fasting LDL cholesterol were similar between aripiprazole- and placebo-treated patients: at 12 weeks, Total Cholesterol (fasting/nonfasting), 1/71 (1.4%) vs.

3/74 (4.1%); Fasting Triglycerides, 8/62 (12.9%) vs.

5/37 (13.5%); Fasting LDL Cholesterol, 0/34 (0%) vs.

1/25 (4.0%), respectively; and at 24 weeks, Total Cholesterol (fasting/nonfasting), 1/42 (2.4%) vs.

3/37 (8.1%); Fasting Triglycerides, 5/34 (14.7%) vs.

5/20 (25%); Fasting LDL Cholesterol, 0/22 (0%) vs.

1/18 (5.6%), respectively.

Pediatric Patients and Adolescents Table 11 shows the proportion of adolescents with schizophrenia (13 to 17 years) and pediatric patients with another disorder (10 to 17 years) with changes in total cholesterol and HDL cholesterol (pooled from two placebo-controlled trials; median exposure 42 to 43 days) and fasting triglycerides (pooled from two placebo-controlled trials; median exposure 42 to 44 days).

Table 11: Changes in Blood Lipid Parameters from Placebo-Controlled Monotherapy Trials in Pediatric and Adolescent Patients in Schizophrenia and Another Disorder Treatment Arm n/N % Total Cholesterol Normal to High (< 170 mg/dL to ≥ 200 mg/dL) Aripiprazole 3/220 1.4 Placebo 0/116 0 Fasting Triglycerides Normal to High (< 150 mg/dL to ≥ 200 mg/dL) Aripiprazole 7/187 3.7 Placebo 4/85 4.7 HDL Cholesterol Normal to Low (≥ 40 mg/dL to < 40 mg/dL) Aripiprazole 27/236 11.4 Placebo 22/109 20.2 In monotherapy trials of adolescents with schizophrenia and pediatric patients with another disorder, the proportion of patients at 12 weeks and 24 weeks with changes from Normal to High in total cholesterol (fasting/nonfasting), fasting triglycerides, and fasting LDL cholesterol were similar between aripiprazole- and placebo-treated patients: at 12 weeks, Total Cholesterol (fasting/nonfasting), 0/57 (0%) vs.

0/15 (0%); Fasting Triglycerides, 2/72 (2.8%) vs.

1/14 (7.1%), respectively; and at 24 weeks, Total Cholesterol (fasting/nonfasting), 0/36 (0%) vs.

0/12 (0%); Fasting Triglycerides, 1/47 (2.1%) vs.

1/10 (10.0%), respectively.

Table 12 shows the proportion of patients with changes in total cholesterol (fasting/nonfasting) and fasting triglycerides (median exposure 56 days) and HDL cholesterol (median exposure 55 to 56 days) from two placebo-controlled trials in pediatric patients (6 to 17 years) with irritability associated with autistic disorder.

Table 12: Changes in Blood Lipid Parameters from Placebo-Controlled Trials in Pediatric Patients with Autistic Disorder Treatment Arm n/N % Total Cholesterol Normal to High (< 170 mg/dL to ≥ 200 mg/dL) Aripiprazole 1/95 1.1 Placebo 0/34 0 Fasting Triglycerides Normal to High (< 150 mg/dL to ≥ 200 mg/dL) Aripiprazole 0/75 0 Placebo 0/30 0 HDL Cholesterol Normal to Low (≥ 40 mg/dL to < 40 mg/dL) Aripiprazole 9/107 8.4 Placebo 5/49 10.2 Table 13 shows the proportion of patients with changes in total cholesterol (fasting/nonfasting) and fasting triglycerides (median exposure 57 days) and HDL cholesterol (median exposure 57 days) from two placebo-controlled trials in pediatric patients (6 to 18 years) with Tourette’s Disorder.

Table 13: Changes in Blood Lipid Parameters from Placebo-Controlled Trials in Pediatric Patients with Tourette’s Disorder Treatment Arm n/N % Total Cholesterol Normal to High (<170 mg/dL to ≥200 mg/dL) Aripiprazole 1/85 1.2 Placebo 0/46 0 Fasting Triglycerides Normal to High (<150 mg/dL to ≥200 mg/dL) Aripiprazole 5/94 5.3 Placebo 2/55 3.6 HDL Cholesterol Normal to Low (≥40 mg/dL to <40 mg/dL) Aripiprazole 4/108 3.7 Placebo 2/67 3.0 Weight Gain Weight gain has been observed with atypical antipsychotic use.

Clinical monitoring of weight is recommended.

Adults In an analysis of 13 placebo-controlled monotherapy trials, primarily from pooled schizophrenia and patients with another disorder, with a median exposure of 21 to 25 days, the mean change in body weight in aripiprazole-treated patients was +0.3 kg (N=1673) compared to –0.1 kg (N=1100) in placebo-controlled patients.

At 24 weeks, the mean change from baseline in body weight in aripiprazole-treated patients was –1.5 kg (n=73) compared to –0.2 kg (n=46) in placebo-treated patients.

In the trials adding aripiprazole to antidepressants, patients first received 8 weeks of antidepressant treatment followed by 6 weeks of adjunctive aripiprazole or placebo in addition to their ongoing antidepressant treatment.

The mean change in body weight in patients receiving adjunctive aripiprazole was +1.7 kg (N=347) compared to +0.4 kg (N=330) in patients receiving adjunctive placebo.

Table 14 shows the percentage of adult patients with weight gain ≥7% of body weight by indication.

Table 14: Percentage of Patients from Placebo-Controlled Trials in Adult Patients with Weight Gain ≥ 7% of Body Weight Indication Treatment Arm N Patients n (%) Weight gain ≥ 7% of body weight Schizophrenia * Aripiprazole 852 69 (8.1) Placebo 379 12 (3.2) Another Disorder † Aripiprazole 719 16 (2.2) Placebo 598 16 (2.7) Another Disorder ‡ Aripiprazole 347 18 (5.2) Placebo 330 2 (0.6) * 4 to 6 weeks duration.

† 3 weeks duration.

‡ 6 weeks duration.

Pediatric Patients and Adolescents In an analysis of two placebo-controlled trials in adolescents with schizophrenia (13 to 17 years) and pediatric patients with another disorder (10 to 17 years) with median exposure of 42 to 43 days, the mean change in body weight in aripiprazole-treated patients was +1.6 kg (N=381) compared to +0.3 kg (N=187) in placebo-treated patients.

At 24 weeks, the mean change from baseline in body weight in aripiprazole-treated patients was +5.8 kg (n=62) compared to +1.4 kg (n=13) in placebo-treated patients.

In two short-term, placebo-controlled trials in patients (6 to 17 years) with irritability associated with autistic disorder with median exposure of 56 days, the mean change in body weight in aripiprazole-treated patients was +1.6 kg (n=209) compared to +0.4 kg (n=98) in placebo-treated patients.

In two short-term, placebo-controlled trials in patients (6 to 18 years) with Tourette’s Disorder with median exposure of 57 days, the mean change in body weight in aripiprazole-treated patients was +1.5 kg (n=105) compared to +0.4 kg (n=66) in placebo-treated patients.

Table 15 shows the percentage of pediatric and adolescent patients with weight gain ≥ 7% of body weight by indication.

Table 15: Percentage of Patients from Placebo-Controlled Monotherapy Trials in Pediatric and Adolescent Patients with Weight Gain ≥ 7% of Body Weight Indication Treatment Arm N Patients n (%) Weight gain ≥7% of body weight Pooled Schizophrenia and Another Disorder * Aripiprazole 381 20 (5.2) Placebo 187 3 (1.6) Irritability Associated with Autistic Disorder † Aripiprazole 209 55 (26.3) Placebo 98 7 (7.1) Tourette’s Disorder ‡ Aripiprazole 105 21 (20.0) Placebo 66 5 (7.6) * 4 to 6 weeks duration.

† 8 weeks duration.

‡ 8 to 10 weeks duration.

In an open-label trial that enrolled patients from the two placebo-controlled trials of adolescents with schizophrenia (13 to 17 years) and pediatric patients with another disorder (10 to 17 years), 73.2% of patients (238/325) completed 26 weeks of therapy with aripiprazole.

After 26 weeks, 32.8% of patients gained ≥7% of their body weight, not adjusted for normal growth.

To adjust for normal growth, z-scores were derived (measured in standard deviations [SD]), which normalize for the natural growth of pediatric patients and adolescents by comparisons to age- and gender-matched population standards.

A z-score change <0.5 SD is considered not clinically significant.

After 26 weeks, the mean change in z-score was 0.09 SD.

In an open-label trial that enrolled patients from two short-term, placebo-controlled trials, patients (6 to 17 years) with irritability associated with autistic disorder, as well as de novo patients, 60.3% (199/330) completed one year of therapy with aripiprazole.

The mean change in weight z-score was 0.26 SDs for patients receiving >9 months of treatment.

When treating pediatric patients for any indication, weight gain should be monitored and assessed against that expected for normal growth.

5.7 Pathological Gambling and Other Compulsive Behaviors Post-marketing case reports suggest that patients can experience intense urges, particularly for gambling, and the inability to control these urges while taking aripiprazole.

Other compulsive urges, reported less frequently, include: sexual urges, shopping, eating or binge eating, and other impulsive or compulsive behaviors.

Because patients may not recognize these behaviors as abnormal, it is important for prescribers to ask patients or their caregivers specifically about the development of new or intense gambling urges, compulsive sexual urges, compulsive shopping, binge or compulsive eating, or other urges while being treated with aripiprazole.

It should be noted that impulse-control symptoms can be associated with the underlying disorder.

In some cases, although not all, urges were reported to have stopped when the dose was reduced or the medication was discontinued.

Compulsive behaviors may result in harm to the patient and others if not recognized.

Consider dose reduction or stopping the medication if a patient develops such urges.

5.8 Orthostatic Hypotension Aripiprazole may cause orthostatic hypotension, perhaps due to its α 1 -adrenergic receptor antagonism.

The incidence of orthostatic hypotension-associated events from short-term, placebo-controlled trials of adult patients on oral aripiprazole (n=2467) included (aripiprazole incidence, placebo incidence) orthostatic hypotension (1%, 0.3%), postural dizziness (0.5%, 0.3%), and syncope (0.5%, 0.4%); of pediatric patients 6 to 18 years of age (n=732) on oral aripiprazole included orthostatic hypotension (0.5%, 0%), postural dizziness (0.4%, 0%), and syncope (0.2%, 0%) [see Adverse Reactions (6.1) ].

The incidence of a significant orthostatic change in blood pressure (defined as a decrease in systolic blood pressure ≥20 mmHg accompanied by an increase in heart rate ≥25 bpm when comparing standing to supine values) for aripiprazole was not meaningfully different from placebo (aripiprazole incidence, placebo incidence): in adult oral aripiprazole-treated patients (4%, 2%), or in pediatric oral aripiprazole-treated patients aged 6 to 18 years (0.4%, 1%).

Aripiprazole should be used with caution in patients with known cardiovascular disease (history of myocardial infarction or ischemic heart disease, heart failure or conduction abnormalities), cerebrovascular disease, or conditions which would predispose patients to hypotension (dehydration, hypovolemia, and treatment with antihypertensive medications) [see Drug Interactions (7.1) ] .

5.9 Falls Antipsychotics, including aripiprazole, may cause somnolence, postural hypotension, motor and sensory instability, which may lead to falls and, consequently, fractures or other injuries.

For patients with diseases, conditions, or medications that could exacerbate these effects, complete fall risk assessments when initiating antipsychotic treatment and recurrently for patients on long-term antipsychotic therapy.

5.10 Leukopenia, Neutropenia, and Agranulocytosis In clinical trials and/or post-marketing experience, events of leukopenia and neutropenia have been reported temporally related to antipsychotic agents, including aripiprazole.

Agranulocytosis has also been reported.

Possible risk factors for leukopenia/neutropenia include pre-existing low white blood cell count (WBC)/absolute neutrophil count (ANC) and history of drug-induced leukopenia/neutropenia.

In patients with a history of a clinically significant low WBC/ANC or drug-induced leukopenia/neutropenia, perform a complete blood count (CBC) frequently during the first few months of therapy.

In such patients, consider discontinuation of aripiprazole at the first sign of a clinically significant decline in WBC in the absence of other causative factors.

Monitor patients with clinically significant neutropenia for fever or other symptoms or signs of infection and treat promptly if such symptoms or signs occur.

Discontinue aripiprazole in patients with severe neutropenia (absolute neutrophil count <1000/mm 3 ) and follow their WBC counts until recovery.

5.11 Seizures/Convulsions In short-term, placebo-controlled trials, patients with a history of seizures excluded seizures/convulsions occurred in 0.1% (3/2467) of undiagnosed adult patients treated with oral aripiprazole and in 0.1% (1/732) of pediatric patients (6 to 18 years).

As with other antipsychotic drugs, aripiprazole should be used cautiously in patients with a history of seizures or with conditions that lower the seizure threshold.

Conditions that lower the seizure threshold may be more prevalent in a population of 65 years or older.

5.12 Potential for Cognitive and Motor Impairment Aripiprazole, like other antipsychotics, may have the potential to impair judgment, thinking, or motor skills.

For example, in short-term, placebo-controlled trials, somnolence (including sedation) was reported as follows (aripiprazole incidence, placebo incidence): in adult patients (n=2467) treated with oral aripiprazole (11%, 6%), and in pediatric patients ages 6 to 17 (n=611) (24%, 6%).

Somnolence (including sedation) led to discontinuation in 0.3% (8/2467) of adult patients and 3% (20/732) of pediatric patients (6 to 18 years) on oral aripiprazole in short-term, placebo-controlled trials.

Despite the relatively modest increased incidence of these events compared to placebo, patients should be cautioned about operating hazardous machinery, including automobiles, until they are reasonably certain that therapy with aripiprazole does not affect them adversely.

5.13 Body Temperature Regulation Disruption of the body’s ability to reduce core body temperature has been attributed to antipsychotic agents.

Appropriate care is advised when prescribing aripiprazole for patients who will be experiencing conditions which may contribute to an elevation in core body temperature, (e.g., exercising strenuously, exposure to extreme heat, receiving concomitant medication with anticholinergic activity, or being subject to dehydration) [see Adverse Reactions (6.2) ] .

5.14 Suicide The possibility of a suicide attempt is inherent in psychotic illnesses, and close supervision of high-risk patients should accompany drug therapy.

Prescriptions for aripiprazole should be written for the smallest quantity consistent with good patient management in order to reduce the risk of overdose [see Adverse Reactions (6.1 , 6.2 )] .

5.15 Dysphagia Esophageal dysmotility and aspiration have been associated with antipsychotic drug use, including aripiprazole.

Aspiration pneumonia is a common cause of morbidity and mortality in elderly patients, in particular those with advanced Alzheimer’s dementia.

Aripiprazole and other antipsychotic drugs should be used cautiously in patients at risk for aspiration pneumonia [see Warnings and Precautions (5.1) and Adverse Reactions (6.2) ] .

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION Advise the patient to read the FDA-approved patient labeling (Medication Guide).

Discuss the following issues with patients prescribed aripiprazole: Clinical Worsening of Depression and Suicide Risk Patients, their families, and their caregivers should be encouraged to be alert to the emergence of anxiety, agitation, panic attacks, insomnia, irritability, hostility, aggressiveness, impulsivity, akathisia (psychomotor restlessness), hypomania, mania, other unusual changes in behavior, worsening of depression, and suicidal ideation, especially early during antidepressant treatment and when the dose is adjusted up or down.

Families and caregivers of patients should be advised to look for the emergence of such symptoms on a day-to-day basis, since changes may be abrupt.

Such symptoms should be reported to the patient’s prescriber or health professional, especially if they are severe, abrupt in onset, or were not part of the patient’s presenting symptoms.

Symptoms such as these may be associated with an increased risk for suicidal thinking and behavior and indicate a need for very close monitoring and possibly changes in the medication [see Warnings and Precautions (5.3) ] .

Prescribers or other health professionals should inform patients, their families, and their caregivers about the benefits and risks associated with treatment with aripiprazole and should counsel them in its appropriate use.

A patient Medication Guide including information about “Antidepressant Medicines, Depression and other Serious Mental Illness, and Suicidal Thoughts or Actions” is available for aripiprazole.

The prescriber or health professional should instruct patients, their families, and their caregivers to read the Medication Guide and should assist them in understanding its contents.

Patients should be given the opportunity to discuss the contents of the Medication Guide and to obtain answers to any questions they may have.

It should be noted that aripiprazole is not approved as a single agent for treatment of depression and has not been evaluated in pediatric major depressive disorder.

Pathological Gambling and Other Compulsive Behaviors Advise patients and their caregivers of the possibility that they may experience compulsive urges to shop, intense urges to gamble, compulsive sexual urges, binge eating and/or other compulsive urges and the inability to control these urges while taking aripiprazole.

In some cases, but not all, the urges were reported to have stopped when the dose was reduced or stopped [see Warnings and Precautions (5.7) ] .

Interference with Cognitive and Motor Performance Because aripiprazole may have the potential to impair judgment, thinking, or motor skills, patients should be cautioned about operating hazardous machinery, including automobiles, until they are reasonably certain that aripiprazole therapy does not affect them adversely [see Warnings and Precautions (5.12) ] .

Concomitant Medication Patients should be advised to inform their physicians if they are taking, or plan to take, any prescription or over-the-counter drugs, since there is a potential for interactions [see Drug Interactions (7) ] .

Heat Exposure and Dehydration Patients should be advised regarding appropriate care in avoiding overheating and dehydration [see Warnings and Precautions (5.13) ] .

Pregnancy Advise patients to notify their healthcare provider if they become pregnant or intend to become pregnant during treatment with aripiprazole.

Advise patients that aripiprazole may cause extrapyramidal and/or withdrawal symptoms (agitation, hypertonia, hypotonia, tremor, somnolence, respiratory distress, and feeding disorder) in a neonate.

Advise patients that there is a pregnancy registry that monitors pregnancy outcomes in women exposed to aripiprazole during pregnancy [see Use in Specific Populations (8.1) ].

Manufactured by: Amneal Pharmaceuticals Pvt.

Ltd.

Ahmedabad, 382220 INDIA Distributed by: Amneal Pharmaceuticals LLC Bridgewater, NJ 08807 Rev.

05-2023-12 Dispense with Medication Guide available at: documents.amneal.com/mg/aripiprazole.pdf

DOSAGE AND ADMINISTRATION

2 Initial Dose Recommended Dose Maximum Dose Schizophrenia – adults (2.1) 10 to 15 mg/day 10 to 15 mg/day 30 mg/day Schizophrenia – adolescents (2.1) 2 mg/day 10 mg/day 30 mg/day Irritability associated with autistic disorder – pediatric patients (2.4) 2 mg/day 5 to 10 mg/day 15 mg/day Tourette’s disorder – (2.5) Patients <50 kg 2 mg/day 5 mg/day 10 mg/day Patients ≥50 kg 2 mg/day 10 mg/day 20 mg/day Oral formulations: Administer once daily without regard to meals.

( 2 ) Known CYP2D6 poor metabolizers: Half of the usual dose.

( 2.7 ) 2.1 Schizophrenia Adults The recommended starting and target dose for aripiprazole tablets is 10 mg/day or 15 mg/day administered on a once-a-day schedule without regard to meals.

Aripiprazole tablets have been systematically evaluated and shown to be effective in a dose range of 10 mg/day to 30 mg/day, when administered as the tablet formulation; however, doses higher than 10 mg/day or 15 mg/day were not more effective than 10 mg/day or 15 mg/day.

Dosage increases should generally not be made before 2 weeks, the time needed to achieve steady-state [see Clinical Studies (14.1) ] .

Maintenance Treatment: Maintenance of efficacy in schizophrenia was demonstrated in a trial involving patients with schizophrenia who had been symptomatically stable on other antipsychotic medications for periods of 3 months or longer.

These patients were discontinued from those medications and randomized to either aripiprazole tablets 15 mg/day or placebo, and observed for relapse [see Clinical Studies (14.1) ] .

Patients should be periodically reassessed to determine the continued need for maintenance treatment.

Adolescents The recommended target dose of aripiprazole tablets is 10 mg/day.

Aripiprazole was studied in adolescent patients 13 to 17 years of age with schizophrenia at daily doses of 10 mg and 30 mg.

The starting daily dose of the tablet formulation in these patients was 2 mg, which was titrated to 5 mg after 2 days and to the target dose of 10 mg after 2 additional days.

Subsequent dose increases should be administered in 5 mg increments.

The 30 mg/day dose was not shown to be more efficacious than the 10 mg/day dose.

Aripiprazole tablets can be administered without regard to meals [see Clinical Studies (14.1) ] .

Patients should be periodically reassessed to determine the need for maintenance treatment.

Switching from Other Antipsychotics There are no systematically collected data to specifically address switching patients with schizophrenia from other antipsychotics to aripiprazole tablets or concerning concomitant administration with other antipsychotics.

While immediate discontinuation of the previous antipsychotic treatment may be acceptable for some patients with schizophrenia, more gradual discontinuation may be most appropriate for others.

In all cases, the period of overlapping antipsychotic administration should be minimized.

2.4 Irritability Associated with Autistic Disorder Pediatric Patients (6 to 17 years) The recommended dosage range for the treatment of pediatric patients with irritability associated with autistic disorder is 5 mg/day to 15 mg/day.

Dosing should be initiated at 2 mg/day.

The dose should be increased to 5 mg/day, with subsequent increases to 10 mg/day or 15 mg/day if needed.

Dose adjustments of up to 5 mg/day should occur gradually, at intervals of no less than one week [see Clinical Studies (14.4) ] .

Patients should be periodically reassessed to determine the continued need for maintenance treatment.

2.5 Tourette’s Disorder Pediatric Patients (6 to 18 years) The recommended dosage range for Tourette’s Disorder is 5 mg/day to 20 mg/day.

For patients weighing less than 50 kg, dosing should be initiated at 2 mg/day with a target dose of 5 mg/day after 2 days.

The dose can be increased to 10 mg/day in patients who do not achieve optimal control of tics.

Dosage adjustments should occur gradually at intervals of no less than one week.

For patients weighing 50 kg or more, dosing should be initiated at 2 mg/day for 2 days, and then increased to 5 mg/day for 5 days, with a target dose of 10 mg/day on Day 8.

The dose can be increased up to 20 mg/day for patients who do not achieve optimal control of tics.

Dosage adjustments should occur gradually in increments of 5 mg/day at intervals of no less than one week [see Clinical Studies (14.5) ] .

Patients should be periodically reassessed to determine the continued need for maintenance treatment.

2.7 Dosage Adjustments for Cytochrome P450 Considerations Dosage adjustments are recommended in patients who are known CYP2D6 poor metabolizers and in patients taking concomitant CYP3A4 inhibitors or CYP2D6 inhibitors or strong CYP3A4 inducers (see Table 2).

When the co-administered drug is withdrawn from the combination therapy, aripiprazole tablets dosage should then be adjusted to its original level.

When the co-administered CYP3A4 inducer is withdrawn, aripiprazole tablets dosage should be reduced to the original level over 1 to 2 weeks.

Patients who may be receiving a combination of strong, moderate, and weak inhibitors of CYP3A4 and CYP2D6 (e.g., a strong CYP3A4 inhibitor and a moderate CYP2D6 inhibitor or a moderate CYP3A4 inhibitor with a moderate CYP2D6 inhibitor), the dosing may be reduced to one-quarter (25%) of the usual dose initially and then adjusted to achieve a favorable clinical response.

Table 2: Dose Adjustments for Aripiprazole Tablets in Patients who are known CYP2D6 Poor Metabolizers and Patients Taking Concomitant CYP2D6 Inhibitors, 3A4 Inhibitors, and/or CYP3A4 Inducers Factors Dosage Adjustments for Aripiprazole Tablets Known CYP2D6 Poor Metabolizers Administer half of usual dose Known CYP2D6 Poor Metabolizers taking concomitant strong CYP3A4 inhibitors (e.g., itraconazole, clarithromycin) Administer a quarter of usual dose Strong CYP2D6 (e.g., quinidine, fluoxetine, paroxetine) or CYP3A4 inhibitors (e.g., itraconazole, clarithromycin) Administer half of usual dose Strong CYP2D6 and CYP3A4 inhibitors Administer a quarter of usual dose Strong CYP3A4 inducers (e.g., carbamazepine, rifampin) Double usual dose over 1 to 2 weeks

Sudafed PE Children’s Cold & Cough 5 MG / 2.5 MG per 5 ML Oral Solution

Generic Name: DEXTROMETHORPHAN HYDROBROMIDE AND PHENYLEPHRINE HYDROCHLORIDE
Brand Name: Childrens SUDAFED PE Cold plus Cough
  • Substance Name(s):
  • DEXTROMETHORPHAN HYDROBROMIDE
  • PHENYLEPHRINE HYDROCHLORIDE

WARNINGS

Warnings Do not use in a child who is taking a prescription monoamine oxidase inhibitor (MAOI) (certain drugs for depression, psychiatric or emotional conditions, or Parkinson’s disease), or for 2 weeks after stopping the MAOI drug.

If you do not know if your child’s prescription drug contains an MAOI, ask a doctor or pharmacist before giving this product.

Ask a doctor before use if the child has heart disease high blood pressure thyroid disease diabetes persistent or chronic cough such as occurs with asthma cough that occurs with too much phlegm (mucus) a sodium-restricted diet When using this product do not exceed recommended dose Stop use and ask a doctor if nervousness, dizziness, or sleeplessness occur symptoms do not improve within 7 days or occur with a fever cough gets worse or lasts for more than 7 days cough tends to come back or occurs with fever, rash or headache that lasts These could be signs of a serious condition.

Keep out of reach of children.

In case of overdose, get medical help or contact a Poison Control Center right away.

(1-800-222-1222)

INDICATIONS AND USAGE

Uses temporarily relieves these symptoms due to the common cold, hay fever, or other upper respiratory allergies: cough nasal congestion sinus congestion and pressure

INACTIVE INGREDIENTS

Inactive ingredients anhydrous citric acid, carboxymethylcellulose sodium, edetate disodium, FD&C blue no.

1, FD&C red no.

40, flavors, glycerin, purified water, sodium benzoate, sodium citrate, sorbitol solution, sucralose

PURPOSE

Active ingredients (in each 5 mL) Purposes Dextromethorphan HBr 5 mg Cough suppressant Phenylephrine HCl 2.5 mg Nasal decongestant

KEEP OUT OF REACH OF CHILDREN

Keep out of reach of children.

In case of overdose, get medical help or contact a Poison Control Center right away.

(1-800-222-1222)

ASK DOCTOR

Ask a doctor before use if the child has heart disease high blood pressure thyroid disease diabetes persistent or chronic cough such as occurs with asthma cough that occurs with too much phlegm (mucus) a sodium-restricted diet

DOSAGE AND ADMINISTRATION

Directions find right dose on chart below mL = milliliters repeat dose every 4 hours do not give more than 6 times in 24 hours Age (yr) Dose (mL) under 4 years do not use 4 to 5 years 5 mL 6 to 11 years 10 mL Attention: use only enclosed dosing cup specifically designed for use with this product.

Do not use any other dosing device.

DO NOT USE

Do not use in a child who is taking a prescription monoamine oxidase inhibitor (MAOI) (certain drugs for depression, psychiatric or emotional conditions, or Parkinson’s disease), or for 2 weeks after stopping the MAOI drug.

If you do not know if your child’s prescription drug contains an MAOI, ask a doctor or pharmacist before giving this product.

STOP USE

Stop use and ask a doctor if nervousness, dizziness, or sleeplessness occur symptoms do not improve within 7 days or occur with a fever cough gets worse or lasts for more than 7 days cough tends to come back or occurs with fever, rash or headache that lasts These could be signs of a serious condition.

ACTIVE INGREDIENTS

Active ingredients (in each 5 mL) Purposes Dextromethorphan HBr 5 mg Cough suppressant Phenylephrine HCl 2.5 mg Nasal decongestant