diltiazem HCl 120 MG 24HR Extended Release Oral Capsule

WARNINGS

1.

Cardiac Conduction: Diltiazem prolongs AV node refractory periods without significantly prolonging sinus node recovery time, except in patients with sick sinus syndrome.

This effect may rarely result in abnormally slow heart rates (particularly in patients with sick sinus syndrome) or second or third-degree AV block (13 of 3,290 patients or 0.40%).

Concomitant use of diltiazem with beta-blockers or digitalis may result in additive effects on cardiac conduction.

A patient with Prinzmetal’s angina developed periods of asystole (2 to 5 seconds) after a single dose of 60 mg of diltiazem.

(See ADVERSE REACTIONS section.) 2.

Congestive Heart Failure: Although diltiazem has a negative inotropic effect in isolated animal tissue preparations, hemodynamic studies in humans with normal ventricular function have not shown a reduction in cardiac index nor consistent negative effects on contractility (dp/dt).

An acute study of oral diltiazem in patients with impaired ventricular function (ejection fraction 24% ± 6%) showed improvement in indices of ventricular function without significant decrease in contractile function (dp/dt).

Worsening of congestive heart failure has been reported in patients with preexisting impairment of ventricular function.

Experience with the use of diltiazem in combination with beta-blockers in patients with impaired ventricular function is limited.

Caution should be exercised when using this combination.

3.

Hypotension: Decreases in blood pressure associated with diltiazem therapy may occasionally result in symptomatic hypotension.

4.

Acute Hepatic Injury: Mild elevations of transaminases with and without concomitant elevation in alkaline phosphatase and bilirubin have been observed in clinical studies.

Such elevations were usually transient and frequently resolved even with continued diltiazem treatment.

In rare instances, significant elevations in enzymes such as alkaline phosphatase, LDH, SGOT, SGPT, and other phenomena consistent with acute hepatic injury have been noted.

These reactions tended to occur early after therapy initiation (1 to 8 weeks) and have been reversible upon discontinuation of drug therapy.

The relationship to diltiazem is uncertain in some cases, but probable in some.

(See PRECAUTIONS .)

DRUG INTERACTIONS

Drug Interactions Due to the potential for additive effects, caution and careful titration are warranted in patients receiving diltiazem concomitantly with other agents known to affect cardiac contractility and/or conduction.

(See WARNINGS .) Pharmacologic studies indicate that there may be additive effects in prolonging AV conduction when using beta-blockers or digitalis concomitantly with diltiazem.

(See WARNINGS .) As with all drugs, care should be exercised when treating patients with multiple medications.

Diltiazem undergoes biotransformation by cytochrome P-450 mixed function oxidase.

Coadministration of diltiazem with other agents which follow the same route of biotransformation may result in the competitive inhibition of metabolism.

Especially in patients with renal and/or hepatic impairment, dosages of similarly metabolized drugs, particularly those of low therapeutic ratio, may require adjustment when starting or stopping concomitantly administered diltiazem to maintain optimum therapeutic blood levels.

Beta-Blockers:Controlled and uncontrolled domestic studies suggest that concomitant use of diltiazem and beta-blockers is usually well tolerated, but available data are not sufficient to predict the effects of concomitant treatment in patients with left ventricular dysfunction or cardiac conduction abnormalities.

Administration of diltiazem concomitantly with propranolol in five normal volunteers resulted in increased propranolol levels in all subjects and bioavailability of propranolol was increased approximately 50%.

In vitro, propranolol appears to be displaced from its binding sites by diltiazem.

If combination therapy is initiated or withdrawn in conjunction with propranolol, an adjustment in the propranolol dose may be warranted.

(See WARNINGS .) Cimetidine:A study in six healthy volunteers has shown a significant increase in peak diltiazem plasma levels (58%) and area-under-the-curve (53%) after a 1-week course of cimetidine at 1200 mg per day and a single dose of diltiazem 60 mg.

Ranitidine produced smaller, nonsignificant increases.

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

Patients currently receiving diltiazem therapy should be carefully monitored for a change in pharmacological effect when initiating and discontinuing therapy with cimetidine.

An adjustment in the diltiazem dose may be warranted.

Digitalis:Administration of diltiazem with digoxin in 24 healthy male subjects increased plasma digoxin concentrations approximately 20%.

Another investigator found no increase in digoxin levels in 12 patients with coronary artery disease.

Since there have been conflicting results regarding the effect of digoxin levels, it is recommended that digoxin levels be monitored when initiating, adjusting, and discontinuing diltiazem therapy to avoid possible over- or under-digitalization.

(See WARNINGS .) Anesthetics:The depression of cardiac contractility, conductivity, and automaticity as well as the vascular dilation associated with anesthetics may be potentiated by calcium channel blockers.

When used concomitantly, anesthetics and calcium blockers should be titrated carefully.

Cyclosporine:A pharmacokinetic interaction between diltiazem and cyclosporine has been observed during studies involving renal and cardiac transplant patients.

In renal and cardiac transplant recipients, a reduction of cyclosporine dose ranging from 15% to 48% was necessary to maintain cyclosporine trough concentrations similar to those seen prior to the addition of diltiazem.

If these agents are to be administered concurrently, cyclosporine concentrations should be monitored, especially when diltiazem therapy is initiated, adjusted or discontinued.

The effect of cyclosporine on diltiazem plasma concentrations has not been evaluated.

Carbamazepine : Concomitant administration of diltiazem with carbamazepine has been reported to result in elevated serum levels of carbamazepine (40% to 72% increase), resulting in toxicity in some cases.

Patients receiving these drugs concurrently should be monitored for a potential drug interaction.

OVERDOSAGE

The oral LD 50 ’s in mice and rats range from 415 to 740 mg/kg and from 560 to 810 mg/kg, respectively.

The intravenous LD 50 ’s in these species were 60 and 38 mg/kg, respectively.

The oral LD 50 in dogs is considered to be in excess of 50 mg/kg, while lethality was seen in monkeys at 360 mg/kg.

The toxic dose in man is not known.

Due to extensive metabolism, blood levels after a standard dose of diltiazem can vary over tenfold, limiting the usefulness of blood levels in overdose cases.

There have been 29 reports of diltiazem overdose in doses ranging from less than 1 g to 10.8 g.

Sixteen of these reports involved multiple drug ingestions.

Twenty-two reports indicated patients had recovered from diltiazem overdose ranging from less than 1 g to 10.8 g.

There were seven reports with a fatal outcome; although the amount of diltiazem ingested was unknown, multiple drug ingestions were confirmed in six of the seven reports.

Events observed following diltiazem overdose included bradycardia, hypotension, heart block, and cardiac failure.

Most reports of overdose described some supportive medical measure and/or drug treatment.

Bradycardia frequently responded favorably to atropine as did heart block, although cardiac pacing was also frequently utilized to treat heart block.

Fluids and vasopressors were used to maintain blood pressure, and in cases of cardiac failure, inotropic agents were administered.

In addition, some patients received treatment with ventilatory support, gastric lavage, activated charcoal, and/or intravenous calcium.

Evidence of the effectiveness of intravenous calcium administration to reverse the pharmacological effects of diltiazem overdose was conflicting.

In the event of overdose or exaggerated response, appropriate supportive measures should be employed in addition to gastrointestinal decontamination.

Diltiazem does not appear to be removed by peritoneal or hemodialysis.

Limited data suggest that plasmapheresis or charcoal hemoperfusion may hasten diltiazem elimination following overdose.

Based on the known pharmacological effects of diltiazem and/or reported clinical experiences, the following measures may be considered: Bradycardia: Administer atropine (0.60 to 1.0 mg).

If there is no response to vagal blockade, administer isoproterenol cautiously.

High-Degree AV Block: Treat as for bradycardia above.

Fixed high-degree AV block should be treated with cardiac pacing.

Cardiac Failure: Administer inotropic agents (isoproterenol, dopamine, or dobutamine) and diuretics.

Hypotension: Vasopressors (eg, dopamine or levarterenol bitartrate).

Actual treatment and dosage should depend on the severity of the clinical situation and the judgment and experience of the treating physician.

DESCRIPTION

Diltiazem hydrochloride is a calcium ion influx inhibitor (slow channel blocker or calcium antagonist).

Chemically, diltiazem hydrochloride is 1,5-benzothiazepin-4(5 H)one,3-(acetyloxy)-5-[2-(dimethylamino)ethyl]-2,3-dihydro-2-(4-methoxyphenyl)-, monohydrochloride,(+)-cis-.

The chemical structure is: Diltiazem hydrochloride is a white to off-white crystalline powder with a bitter taste.

It is soluble in water, methanol, and chloroform.

It has a molecular weight of 450.98.

Diltiazem hydrochloride is formulated as a once-a-day extended release capsule containing either 120 mg, 180 mg, 240 mg, or 300 mg diltiazem hydrochloride.

Each diltiazem extended-release capsule, for oral administration, contains the following inactive ingredients: 120 mg — ammonio methacrylate copolymer NF, type A, ammonio methacrylate copolymer NF, type B, ammonium hydroxide, black iron oxide, gelatin, hydroxypropyl cellulose, pharmaceutical glaze, propylene glycol, silicon dioxide, simethicone, sodium lauryl sulfate, sugar spheres, talc, titanium dioxide, triethyl citrate.

180 mg — ammonio methacrylate copolymer NF, type A, ammonio methacrylate copolymer NF, type B, ammonium hydroxide, D&C yellow #10, FD&C blue #1, FD&C green #3, gelatin, hydroxypropyl cellulose, pharmaceutical glaze, propylene glycol, silicon dioxide, simethicone, sodium lauryl sulfate, sugar spheres, talc, titanium dioxide, triethyl citrate.

240 mg — ammonio methacrylate copolymer NF, type A, ammonio methacrylate copolymer NF, type B, ammonium hydroxide, D&C yellow #10, FD&C green #3, gelatin, hydroxypropyl cellulose, pharmaceutical glaze, propylene glycol, silicon dioxide, simethicone, sodium lauryl sulfate, sugar spheres, talc, titanium dioxide, triethyl citrate.

300 mg— ammonio methacrylate copolymer NF, type A, ammonio methacrylate copolymer NF, type B, ammonium hydroxide, black iron oxide, D&C yellow #10, FD&C green #3, gelatin, hydroxypropyl cellulose, pharmaceutical glaze, propylene glycol, silicon dioxide, simethicone, sodium lauryl sulfate, sugar spheres, talc, titanium dioxide, triethyl citrate.

This drug product conforms to USP Drug release test #11.

chemical structure

HOW SUPPLIED

Diltiazem Hydrochloride Extended-release Capsules, USP are supplied as follows: 120 mg — Each #2 capsule with light gray opaque cap and body printed with and 2588 on both cap and body in white ink contains 120 mg of diltiazem hydrochloride, USP.

Capsules are supplied in: Overbagged with 10 Capsules per bag, NDC 55154-6836-0 Store at 25°C (77°F); excursions permitted to 15° to 30°C (59°-86°F).

Avoid excessive humidity.

Manufactured by: Actavis Elizabeth LLC 200 Elmora Avenue Elizabeth, NJ 07207 USA Distributed By: McKesson Packaging Concord, NC 28027 Repackaged By: Cardinal Health Zanesville, OH 43701 L50113330317 Revised — January 2008 IS-283-M13-01-A-R3 Imprint Symbol

INDICATIONS AND USAGE

Diltiazem hydrochloride extended-release capsules are indicated for the treatment of hypertension.

They may be used alone or in combination with other antihypertensive medications.

Diltiazem hydrochloride extended-release capsules are indicated for the management of chronic stable angina and angina due to coronary artery spasm.

PEDIATRIC USE

Pediatric Use Safety and effectiveness in pediatric patients have not been established.

PREGNANCY

Pregnancy Category C.

Reproduction studies have been conducted in mice, rats, and rabbits.

Administration of doses ranging from five to ten times greater (on a mg/kg basis) than the daily recommended therapeutic dose has resulted in embryo and fetal lethality.

These doses, in some studies, have been reported to cause skeletal abnormalities.

In the perinatal/postnatal studies, there was an increased incidence of stillbirths at doses of 20 times the human dose or greater.

There are no well-controlled studies in pregnant women; therefore, use diltiazem in pregnant women only if the potential benefit justifies the potential risk to the fetus.

NUSRING MOTHERS

Nursing Mothers Diltiazem is excreted in human milk.

One report suggests that concentrations in breast milk may approximate serum levels.

If use of diltiazem is deemed essential, an alternative method of infant feeding should be instituted.

DOSAGE AND ADMINISTRATION

Patients controlled on diltiazem alone or in combination with other medications may be switched to diltiazem hydrochloride extended-release capsules at the nearest equivalent total daily dose.

Higher doses of diltiazem hydrochloride extended-release capsules may be needed in some patients.

Patients should be closely monitored.

Subsequent titration to higher or lower doses may be necessary and should be initiated as clinically warranted.

There is limited general clinical experience with doses above 360 mg, but doses to 540 mg have been studied in clinical trials.

The incidence of side effects increases as the dose increases with first-degree AV block, dizziness, and sinus bradycardia bearing the strongest relationship to dose.

Hypertension: Dosage needs to be adjusted by titration to individual patient needs.

When used as monotherapy, reasonable starting doses are 180 to 240 mg once daily, although some patients may respond to lower doses.

Maximum antihypertensive effect is usually observed by 14 days of chronic therapy; therefore, dosage adjustments should be scheduled accordingly.

The usual dosage range studied in clinical trials was 240 to 360 mg once daily.

Individual patients may respond to higher doses of up to 480 mg once daily.

Angina: Dosages for the treatment of angina should be adjusted to each patient’s needs, starting with a dose of 120 or 180 mg once daily.

Individual patients may respond to higher doses of up to 480 mg once daily.

When necessary, titration may be carried out over a 7- to 14-day period.

CONCOMITANT USE WITH OTHER CARDIOVASCULAR AGENTS: 1.

Sublingual NTG – May be taken as required to abort acute anginal attacks during diltiazem hydrochloride extended-release capsules therapy.

2.

Prophylactic Nitrate Therapy – Diltiazem hydrochloride extended-release capsules may be safely coadministered with short-and long-acting nitrates.

3.

Beta-Blockers: (See WARNINGS and PRECAUTIONS ).

4.

Antihypertensives – Diltiazem hydrochloride extended-release capsules have an additive antihypertensive effect when used with other antihypertensive agents.

Therefore, the dosage of diltiazem hydrochloride extended-release capsules or the concomitant antihypertensives may need to be adjusted when adding one to the other.

Diclofenac Potassium 50 MG Oral Tablet

WARNINGS

Cardiovascular Effects Cardiovascular Thrombotic Events Clinical trials of several COX-2 selective and nonselective NSAIDs of up to three years duration have shown an increased risk of serious cardiovascular (CV) thrombotic events, myocardial infarction, and stroke, which can be fatal.

All NSAIDs, both COX-2 selective and nonselective, may have a similar risk.

Patients with known CV disease or risk factors for CV disease may be at greater risk.

To minimize the potential risk for an adverse CV event in patients treated with an NSAID, the lowest effective dose should be used for the shortest duration possible.

Physicians and patients should remain alert for the development of such events, even in the absence of previous CV symptoms.

Patients should be informed about the signs and/or symptoms of serious CV events and the steps to take if they occur.

There is no consistent evidence that concurrent use of aspirin mitigates the increased risk of serious CV thrombotic events associated with NSAID use.

The concurrent use of aspirin and an NSAID does increase the risk of serious GI events (see , GI Effects ).

Two large, controlled, clinical trials of a COX-2 selective NSAID for the treatment of pain in the first 10 to 14 days following CABG surgery found an increased incidence of myocardial infarction and stroke (see CONTRAINDICATIONS ).

Hypertension NSAIDs can lead to onset of new hypertension or worsening of preexisting hypertension, either of which may contribute to the increased incidence of CV events.

Patients taking thiazides or loop diuretics may have impaired response to these therapies when taking NSAIDs.

NSAIDs, including diclofenac potassium tablets, should be used with caution in patients with hypertension.

Blood pressure (BP) should be monitored closely during the initiation of NSAID treatment and throughout the course of therapy.

Congestive Heart Failure and Edema Fluid retention and edema have been observed in some patients taking NSAIDs.

Diclofenac potassium tablets should be used with caution in patients with fluid retention or heart failure.

Gastrointestinal (GI) Effects: Risk of GI Ulceration, Bleeding, and Perforation NSAIDs, including diclofenac potassium tablets, can cause serious gastrointestinal (GI) adverse events including inflammation, bleeding, ulceration, and perforation of the stomach, small intestine, or large intestine, which can be fatal.

These serious adverse events can occur at any time, with or without warning symptoms, in patients treated with NSAIDs.

Only one in five patients, who develop a serious upper GI adverse event on NSAID therapy, is symptomatic.

Upper GI ulcers, gross bleeding or perforation caused by NSAIDs occur in approximately 1% of patients treated for 3 to 6 months, and in about 2% to 4% of patients treated for one year.

These trends continue with longer duration of use, increasing the likelihood of developing a serious GI event at some time during the course of therapy.

However, even short-term therapy is not without risk.

NSAIDs should be prescribed with extreme caution in those with a prior history of ulcer disease or gastrointestinal bleeding.

Patients with a prior history of peptic ulcer disease and/or gastrointestinal bleeding who use NSAIDs have a greater than 10 fold increased risk for developing a GI bleed compared to patients with neither of these risk factors.

Other factors that increase the risk for GI bleeding in patients treated with NSAIDs include concomitant use of oral corticosteroids or anticoagulants, longer duration of NSAID therapy, smoking, use of alcohol, older age, and poor general health status.

Most spontaneous reports of fatal GI events are in elderly or debilitated patients and therefore special care should be taken in treating this population.

To minimize the potential risk for an adverse GI event in patients treated with an NSAID, the lowest effective dose should be used for the shortest possible duration.

Patients and physicians should remain alert for signs and symptoms of GI ulceration and bleeding during NSAID therapy and promptly initiate additional evaluation and treatment if a serious GI adverse event is suspected.

This should include discontinuation of the NSAID until a serious GI adverse event is ruled out.

For high risk patients, alternate therapies that do not involve NSAIDs should be considered.

Renal Effects Caution should be used when initiating treatment with diclofenac potassium tablets in patients with considerable dehydration.

Long-term administration of NSAIDs has resulted in renal papillary necrosis and other renal injury.

Renal toxicity has also been seen in patients in whom renal prostaglandins have a compensatory role in the maintenance of renal perfusion.

In these patients, administration of a non-steroidal anti-inflammatory drug may cause a dose-dependent reduction in prostaglandin formation and, secondarily, in renal blood flow, which may precipitate overt renal decompensation.

Patients at greatest risk of this reaction are those with impaired renal function, heart failure, liver dysfunction, those taking diuretics and ACE inhibitors, and the elderly.

Discontinuation of NSAID therapy is usually followed by recovery to the pretreatment state.

Advanced Renal Disease No information is available from controlled clinical studies regarding the use of diclofenac in patients with advanced renal disease.

Therefore, treatment with diclofenac potassium tablets is not recommended in these patients with advanced renal disease.

If diclofenac potassium tablet therapy must be initiated, close monitoring of the patient’s renal function is advisable.

Hepatic Effects Elevations of one or more liver tests may occur during therapy with diclofenac potassium.

These laboratory abnormalities may progress, may remain unchanged, or may be transient with continued therapy.

Borderline elevations (i.e., less than 3 times the ULN [ULN = the upper limit of the normal range]) or greater elevations of transaminases occurred in about 15% of diclofenac-treated patients.

Of the markers of hepatic function, ALT (SGPT) is recommended for the monitoring of liver injury.

In clinical trials, meaningful elevations (i.e., more than 3 times the ULN) of AST (GOT) (ALT was not measured in all studies) occurred in about 2% of approximately 5,700 patients at some time during diclofenac treatment.

In a large, open-label, controlled trial of 3,700 patients treated for 2 to 6 months, patients were monitored first at 8 weeks and 1,200 patients were monitored again at 24 weeks.

Meaningful elevations of ALT and/or AST occurred in about 4% of patients and included marked elevations (i.e., more than 8 times the ULN) in about 1% of the 3,700 patients.

In that open-label study, a higher incidence of borderline (less than 3 times the ULN), moderate (3 to 8 times the ULN), and marked (> 8 times the ULN) elevations of ALT or AST was observed in patients receiving diclofenac when compared to other NSAIDs.

Elevations in transaminases were seen more frequently in patients with osteoarthritis than in those with rheumatoid arthritis.

Almost all meaningful elevations in transaminases were detected before patients became symptomatic.

Abnormal tests occurred during the first 2 months of therapy with diclofenac in 42 of the 51 patients in all trials who developed marked transaminase elevations.

In postmarketing reports, cases of drug-induced hepatotoxicity have been reported in the first month, and in some cases, the first 2 months of therapy, but can occur at any time during treatment with diclofenac.

Postmarketing surveillance has reported cases of severe hepatic reactions, including liver necrosis, jaundice, fulminant hepatitis with and without jaundice, and liver failure.

Some of these reported cases resulted in fatalities or liver transplantation.

Physicians should measure transaminases periodically in patients receiving long-term therapy with diclofenac, because severe hepatotoxicity may develop without a prodrome of distinguishing symptoms.

The optimum times for making the first and subsequent transaminase measurements are not known.

Based on clinical trial data and postmarketing experiences, transaminases should be monitored within 4 to 8 weeks after initiating treatment with diclofenac.

However, severe hepatic reactions can occur at any time during treatment with diclofenac.

If abnormal liver tests persist or worsen, if clinical signs and/or symptoms consistent with liver disease develop, or if systemic manifestations occur (e.g., eosinophilia, rash, abdominal pain, diarrhea, dark urine, etc.), diclofenac potassium should be discontinued immediately.

To minimize the possibility that hepatic injury will become severe between transaminase measurements, physicians should inform patients of the warning signs and symptoms of hepatotoxicity (e.g., nausea, fatigue, lethargy, diarrhea, pruritus, jaundice, right upper quadrant tenderness, and “flu-like” symptoms), and the appropriate action patients should take if these signs and symptoms appear.

To minimize the potential risk for an adverse liver related event in patients treated with diclofenac potassium, the lowest effective dose should be used for the shortest duration possible.

Caution should be exercised in prescribing diclofenac potassium with concomitant drugs that are known to be potentially hepatotoxic (e.g., antibiotics, anti-epileptics).

Anaphylactic Reactions As with other NSAIDs, anaphylactic reactions may occur both in patients with the aspirin triad and in patients without known sensitivity to NSAIDs or known prior exposure to diclofenac potassium tablets.

Diclofenac potassium tablets should not be given to patients with the aspirin triad.

This symptom complex typically occurs in asthmatic patients who experience rhinitis with or without nasal polyps, or who exhibit severe, potentially fatal bronchospasm after taking aspirin or other NSAIDs (see CONTRAINDICATIONS and PRECAUTIONS , Preexisting Asthma ).

Anaphylaxis-type reactions have been reported with NSAID products, including with diclofenac products, such as diclofenac potassium tablets.

Emergency help should be sought in cases where an anaphylactic reaction occurs.

Skin Reactions NSAIDs, including diclofenac potassium tablets, can cause serious skin adverse events such as exfoliative dermatitis, Stevens-Johnson syndrome (SJS), and toxic epidermal necrolysis (TEN), which can be fatal.

These serious events may occur without warning.

Patients should be informed about the signs and symptoms of serious skin manifestations and use of the drug should be discontinued at the first appearance of skin rash or any other sign of hypersensitivity.

Pregnancy In late pregnancy, as with other NSAIDs, diclofenac potassium tablets should be avoided because they may cause premature closure of the ductus arteriosus.

DRUG INTERACTIONS

Drug Interactions Aspirin: When diclofenac is administered with aspirin, its protein binding is reduced.

The clinical significance of this interaction is not known; however, as with other NSAIDs, concomitant administration of diclofenac and aspirin is not generally recommended because of the potential of increased adverse effects.

Methotrexate: NSAIDs have been reported to competitively inhibit methotrexate accumulation in rabbit kidney slices.

This may indicate that they could enhance the toxicity of methotrexate.

Caution should be used when NSAIDs are administered concomitantly with methotrexate.

Cyclosporine: Diclofenac potassium tablets, like other NSAIDs, may affect renal prostaglandins and increase the toxicity of certain drugs.

Therefore, concomitant therapy with diclofenac potassium tablets may increase cyclosporine’s nephrotoxicity.

Caution should be used when diclofenac potassium tablets are administered concomitantly with cyclosporine.

ACE Inhibitors: Reports suggest that NSAIDs may diminish the antihypertensive effect of ACE inhibitors.

This interaction should be given consideration in patients taking NSAIDs concomitantly with ACE inhibitors.

Furosemide: Clinical studies, as well as postmarketing observations, have shown that diclofenac can reduce the natriuretic effect of furosemide and thiazides in some patients.

This response has been attributed to inhibition of renal prostaglandin synthesis.

During concomitant therapy with NSAIDs, the patient should be observed closely for signs of renal failure (see WARNINGS , Renal Effects ), as well as to assure diuretic efficacy.

Lithium: NSAIDs have produced an elevation of plasma lithium levels and a reduction in renal lithium clearance.

The mean minimum lithium concentration increased 15% and the renal clearance was decreased by approximately 20%.

These effects have been attributed to inhibition of renal prostaglandin synthesis by the NSAID.

Thus, when NSAIDs and lithium are administered concurrently, subjects should be observed carefully for signs of lithium toxicity.

Warfarin: The effects of warfarin and NSAIDs on GI bleeding are synergistic, such that users of both drugs together have a risk of serious GI bleeding higher than users of either drug alone.

CYP2C9 Inhibitors or Inducers: Diclofenac is metabolized by cytochrome P450 enzymes, predominantly by CYP2C9.

Coadministration of diclofenac with CYP2C9 inhibitors (e.g., voriconazole) may enhance the exposure and toxicity of diclofenac whereas coadministration with CYP2C9 inducers (e.g., rifampin) may lead to compromised efficacy of diclofenac.

Use caution when dosing diclofenac with CYP2C9 inhibitors or inducers, a dosage adjustment may be warranted (see CLINICAL PHARMACOLOGY , Pharmacokinetics , Drug Interactions ).

OVERDOSAGE

Symptoms following acute NSAID overdoses are usually limited to lethargy, drowsiness, nausea, vomiting, and epigastric pain, which are generally reversible with supportive care.

Gastrointestinal bleeding can occur.

Hypertension, acute renal failure, respiratory depression and coma may occur, but are rare.

Anaphylactoid reactions have been reported with therapeutic ingestion of NSAIDs, and may occur following an overdose.

Patients should be managed by symptomatic and supportive care following an NSAID overdose.

There are no specific antidotes.

Emesis and/or activated charcoal (60 to 100 g in adults, 1 to 2 g/kg in children) and/or osmotic cathartic may be indicated in patients seen within 4 hours of ingestion with symptoms or following a large overdose (5 to 10 times the usual dose).

Forced diuresis, alkalinization of urine, hemodialysis, or hemoperfusion may not be useful due to high protein binding.

DESCRIPTION

Diclofenac potassium tablets USP are a benzeneacetic acid derivative.

Diclofenac potassium tablets USP, 50 mg are available as orange, film-coated tablets for oral administration.

The chemical name is 2-[(2,6-dichlorophenyl)amino] benzeneacetic acid, monopotassium salt.

The structural formula is: C 14 H 10 Cl 2 KNO 2 M.W.

334.25 Diclofenac potassium is a faintly yellowish white to light beige, virtually odorless, slightly hygroscopic crystalline powder.

It is freely soluble in methanol, soluble in ethanol and water, and practically insoluble in chloroform and in dilute acid.

The n-octanol/water partition coefficient is 13.4 at pH 7.4 and 1545 at pH 5.2.

It has a single dissociation constant (pKa) of 4.0 ± 0.2 at 25°C in water.

Each tablet, for oral administration, contains 50 mg of diclofenac potassium.

In addition, each tablet contains the following inactive ingredients: colloidal silicon dioxide, corn starch, FD&C Blue No.

2, FD&C Red No.

40, FD&C Yellow No.

6, hypromellose, lactose monohydrate, magnesium stearate, microcrystalline cellulose, polyethylene glycol 4000, povidone, sodium starch glycolate, titanium dioxide, and tricalcium phosphate.

diclofenac potassium structural formula

HOW SUPPLIED

Diclofenac potassium tablets USP, 50 mg are available as orange, round, unscored, biconvex, film-coated tablets debossed with the numbers “93” and “948” on one face of the tablet and plain on the other, in bottles of 100 and 500.

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

Protect from moisture.

Dispense in a tight, light-resistant container as defined in the USP, with a child-resistant closure (as required).

Manufactured In Israel By: TEVA PHARMACEUTICAL IND.

LTD.

Jerusalem, 91010, Israel Manufactured For: TEVA PHARMACEUTICALS USA Sellersville, PA 18960 Rev.

J 3/2011

GERIATRIC USE

Geriatric Use As with any NSAIDs, caution should be exercised in treating the elderly (65 years and older).

INDICATIONS AND USAGE

Carefully consider the potential benefits and risks of diclofenac potassium tablets and other treatment options before deciding to use diclofenac potassium tablets.

Use the lowest effective dose for the shortest duration consistent with individual patient treatment goals (see WARNINGS ).

Diclofenac potassium tablets are indicated: For treatment of primary dysmenorrhea For relief of mild to moderate pain For relief of the signs and symptoms of osteoarthritis For relief of the signs and symptoms of rheumatoid arthritis

PEDIATRIC USE

Pediatric Use Safety and effectiveness in pediatric patients have not been established.

PREGNANCY

Pregnancy Teratogenic Effects Pregnancy category C Reproductive studies conducted in rats and rabbits have not demonstrated evidence of developmental abnormalities.

However, animal reproduction studies are not always predictive of human response.

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

Nonteratogenic Effects Because of the known effects of non-steroidal anti-inflammatory drugs on the fetal cardiovascular system (closure of ductus arteriosus), use during pregnancy (particularly late pregnancy) should be avoided.

NUSRING MOTHERS

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

Because many drugs are excreted in human milk and because of the potential for serious adverse reactions in nursing infants from diclofenac, 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.

BOXED WARNING

CARDIOVASCULAR RISK NSAIDs 1 may cause an increased risk of serious cardiovascular thrombotic events, myocardial infarction, and stroke, which can be fatal.

This risk may increase with duration of use.

Patients with cardiovascular disease or risk factors for cardiovascular disease may be at greater risk (see WARNINGS ).

Diclofenac potassium tablets are contraindicated for the treatment of perioperative pain in the setting of coronary artery bypass graft (CABG) surgery (see WARNINGS ).

GASTROINTESTINAL RISK NSAIDs cause an increased risk of serious gastrointestinal adverse events including inflammation, bleeding, ulceration, and perforation of the stomach or intestines, which can be fatal.

These events can occur at any time during use and without warning symptoms.

Elderly patients are at greater risk for serious gastrointestinal events (see WARNINGS ).

1 Throughout this package insert, the term NSAID refers to a non-aspirin non-steroidal anti-inflammatory drug.

INFORMATION FOR PATIENTS

Information for Patients Patients should be informed of the following information before initiating therapy with an NSAID and periodically during the course of ongoing therapy.

Patients should also be encouraged to read the NSAID Medication Guide that accompanies each prescription dispensed.

Diclofenac potassium tablets, like other NSAIDs, may cause serious CV side effects, such as MI or stroke, which may result in hospitalization and even death.

Although serious CV events can occur without warning symptoms, patients should be alert for the signs and symptoms of chest pain, shortness of breath, weakness, slurring of speech, and should ask for medical advice when observing any indicative sign or symptoms.

Patients should be apprised of the importance of this follow-up (see WARNINGS , Cardiovascular Effects ).

Diclofenac potassium tablets, like other NSAIDs, can cause GI discomfort and, rarely, more serious GI side effects, such as ulcers and bleeding, which may result in hospitalization and even death.

Although serious GI tract ulcerations and bleeding can occur without warning symptoms, patients should be alert for the signs and symptoms of ulcerations and bleeding, and should ask for medical advice when observing any indicative sign or symptoms including epigastric pain, dyspepsia, melena, and hematemesis.

Patients should be apprised of the importance of this follow-up (see WARNINGS , Gastrointestinal (GI) Effects: Risk of GI Ulceration, Bleeding, and Perforation ).

Diclofenac potassium tablets, like other NSAIDs, can cause serious skin side effects such as exfoliative dermatitis, SJS, and TEN, which may result in hospitalizations and even death.

Although serious skin reactions may occur without warning, patients should be alert for the signs and symptoms of skin rash and blisters, fever, or other signs of hypersensitivity such as itching, and should ask for medical advice when observing any indicative signs or symptoms.

Patients should be advised to stop the drug immediately if they develop any type of rash and contact their physicians as soon as possible.

Patients should promptly report signs or symptoms of unexplained weight gain or edema to their physicians.

Patients should be informed of the warning signs and symptoms of hepatotoxicity (e.g., nausea, fatigue, lethargy, pruritus, jaundice, right upper quadrant tenderness, and “flu-like” symptoms).

If these occur, patients should be instructed to stop therapy and seek immediate medical therapy (see WARNINGS , Hepatic Effects ).

Patients should be informed of the signs of an anaphylactic reaction (e.g., difficulty breathing, swelling of the face or throat).

If these occur, patients should be instructed to seek immediate emergency help (see WARNINGS , Anaphylactic Reactions ).

In late pregnancy, as with other NSAIDs, diclofenac potassium tablets should be avoided because they will cause premature closure of the ductus arteriosus.

DOSAGE AND ADMINISTRATION

Carefully consider the potential benefits and risks of diclofenac potassium tablets USP and other treatment options before deciding to use diclofenac potassium tablets USP.

Use the lowest effective dose for the shortest duration consistent with individual patient treatment goals (see WARNINGS ).

After observing the response to initial therapy with diclofenac potassium tablets USP, the dose and frequency should be adjusted to suit an individual patient’s needs.

For treatment of pain or primary dysmenorrhea the recommended dosage is 50 mg t.i.d.

With experience, physicians may find that in some patients an initial dose of 100 mg of diclofenac potassium tablets USP, followed by 50 mg doses, will provide better relief.

For the relief of osteoarthritis the recommended dosage is 100 to 150 mg/day in divided doses, 50 mg b.i.d.

or t.i.d.

For the relief of rheumatoid arthritis the recommended dosage is 150 to 200 mg/day in divided doses, 50 mg t.i.d.

or q.i.d.

Different formulations of diclofenac [diclofenac sodium enteric-coated tablets; diclofenac sodium extended-release tablets; diclofenac potassium tablets USP] are not necessarily bioequivalent even if the milligram strength is the same.

ofloxacin 0.3 % Ophthalmic Solution

WARNINGS

NOT FOR INJECTION.

Ofloxacin Ophthalmic Solution should not be injected subconjunctivally, nor should it be introduced directly into the anterior chamber of the eye.

Serious and occasionally fatal hypersensitivity (anaphylactic) reactions, some following the first dose, have been reported in patients receiving systemic quinolones, including ofloxacin.

Some reactions were accompanied by cardiovascular collapse, loss of consciousness, angioedema (including laryngeal, pharyngeal or facial edema), airway obstruction, dyspnea, urticaria, and itching.

A rare occurrence of Stevens-Johnson syndrome, which progressed to toxic epidermal necrolysis, has been reported in a patient who was receiving topical ophthalmic ofloxacin.

If an allergic reaction to ofloxacin occurs, discontinue the drug.

Serious acute hypersensitivity reactions may require immediate emergency treatment.

Oxygen and airway management, including intubation should be administered as clinically indicated.

DESCRIPTION

Ofloxacin Ophthalmic Solution USP, 0.3% is a sterile ophthalmic solution.

It is a fluorinated carboxyquinolone anti-infective for topical ophthalmic use.

C 18 H 20 FN 3 O 4 Mol Wt 361.37 Chemical Name: (±)-9-Fluoro-2, 3-dihydro-3-methyl-10-(4-methyl-1-piperazinyl)-7-oxo-7H-pyrido[1,2,3-de]-1,4 benzoxazine-6-carboxylic acid.

Contains: Active: ofloxacin 0.3% (3 mg/mL).

Preservative: benzalkonium chloride 0.005%.

Inactives: sodium chloride, hydrochloric acid and/or sodium hydroxide (to adjust pH) and purified water.

Ofloxacin Ophthalmic Solution is unbuffered and formulated with a pH of 6.4 (range – 6.0 to 6.8).

It has an osmolality of 300 mOsm/kg.

Ofloxacin is a fluorinated 4-quinolone which differs from other fluorinated 4-quinolones in that there is a six member (pyridobenzoxazine) ring from positions 1 to 8 of the basic ring structure.

MM1

HOW SUPPLIED

Ofloxacin Ophthalmic Solution USP, 0.3% is supplied sterile in plastic DROP-TAINER* of the following sizes: 5 mL NDC 61314-015-05 10 mL NDC 61314-015-10 STORAGE: Store at 15°-25°C (59°-77°F).

Rx Only *DROP-TAINER is a registered trademark of Alcon Manufacturing, Ltd.

INDICATIONS AND USAGE

INDICATIONS & USAGE Ofloxacin Ophthalmic Solution is indicated for the treatment of infections caused by susceptible strains of the following bacteria in the conditions listed below: CONJUNCTIVITIS: Gram-positive bacteria: Staphylococcus aureus Staphylococcus epidermidis Streptococcus pneumoniae Pseudomonas aeruginosa Gram-negative bacteria: Enterobacter cloacae Haemophilus influenza Proteus mirabilis Pseudomonas aeruginosa CORNEAL ULCERS: Gram-positive bacteria: Staphylococcus aureus Staphylococcus epidermidis Streptococcus pneumonia Gram-negative bacteria: Pseudomonas aeruginosa Serratia marcescens* Anaerobic species: Propionibacterium acnes *Efficacy for this organism was studied in fewer than 10 infections.

DOSAGE AND ADMINISTRATION

DOSAGE & ADMINISTRATION The recommended dosage regimen for the treatment of bacterial conjunctivitis is: Days 1 and 2 Instill one to two drops every two to four hours in the affected eye(s).

Days 3 through 7 Instill one to two drops four times daily.

The recommended dosage regimen for the treatment of bacterial corneal ulcer is: Days 1 and 2 Instill one to two drops into the affected eye every 30 minutes, while awake.

Awaken at approximately four and six hours after retiring and instill one to two drops.

Days 3 through 7 to 9 Instill one to two drops hourly, while awake.

Days 7 to 9 through treatment completion Instill one to two drops, four times daily.

Bumetanide 0.5 MG Oral Tablet

WARNINGS

Volume and Electrolyte Depletion The dose of bumetanide should be adjusted to the patient’s need.

Excessive doses or too frequent administration can lead to profound water loss, electrolyte depletion, dehydration, reduction in blood volume and circulatory collapse with the possibility of vascular thrombosis and embolism, particularly in elderly patients.

Hypokalemia Hypokalemia can occur as a consequence of bumetanide administration.

Prevention of hypokalemia requires particular attention in the following conditions: patients receiving digitalis and diuretics for congestive heart failure, hepatic cirrhosis and ascites, states of aldosterone excess with normal renal function, potassium-losing nephropathy, certain diarrheal states, or other states where hypokalemia is thought to represent particular added risks to the patient, i.e., history of ventricular arrhythmias.

In patients with hepatic cirrhosis and ascites, sudden alterations of electrolyte balance may precipitate hepatic encephalopathy and coma.

Treatment in such patients is best initiated in the hospital with small doses and careful monitoring of the patient’s clinical status and electrolyte balance.

Supplemental potassium and/or spironolactone may prevent hypokalemia and metabolic alkalosis in these patients.

Ototoxicity In cats, dogs and guinea pigs, bumetanide has been shown to produce ototoxicity.

In these test animals bumetanide was 5 to 6 times more potent than furosemide and, since the diuretic potency of bumetanide is about 40 to 60 times furosemide, it is anticipated that blood levels necessary to produce ototoxicity will rarely be achieved.

The potential exists, however, and must be considered a risk of intravenous therapy, especially at high doses, repeated frequently in the face of renal excretory function impairment.

Potentiation of aminoglycoside ototoxicity has not been tested for bumetanide.

Like other members of this class of diuretics, bumetanide probably shares this risk.

Allergy to Sulfonamides Patients allergic to sulfonamides may show hypersensitivity to bumetanide.

Thrombocytopenia Since there have been rare spontaneous reports of thrombocytopenia from postmarketing experience, patients should be observed regularly for possible occurrence of thrombocytopenia.

DRUG INTERACTIONS

Drug Interactions Drugs With Ototoxic Potential (see WARNINGS ) Especially in the presence of impaired renal function, the use of parenterally administered bumetanide in patients to whom aminoglycoside antibiotics are also being given should be avoided, except in life-threatening conditions.

Drugs With Nephrotoxic Potential There has been no experience with the concurrent use of bumetanide with drugs known to have a nephrotoxic potential.

Therefore, the simultaneous administration of these drugs should be avoided.

Lithium Lithium should generally not be given with diuretics (such as bumetanide) because they reduce its renal clearance and add a high risk of lithium toxicity.

Probenecid Pretreatment with probenecid reduces both the natriuresis and hyperreninemia produced by bumetanide.

This antagonistic effect of probenecid on bumetanide natriuresis is not due to a direct action on sodium excretion but is probably secondary to its inhibitory effect on renal tubular secretion of bumetanide.

Thus, probenecid should not be administered concurrently with bumetanide.

Indomethacin Indomethacin blunts the increases in urine volume and sodium excretion seen during bumetanide treatment and inhibits the bumetanide-induced increase in plasma renin activity.

Concurrent therapy with bumetanide is thus not recommended.

Antihypertensives Bumetanide may potentiate the effect of various antihypertensive drugs, necessitating a reduction in the dosage of these drugs.

Digoxin Interaction studies in humans have shown no effect on digoxin blood levels.

Anticoagulants Interaction studies in humans have shown bumetanide to have no effect on warfarin metabolism or on plasma prothrombin activity.

OVERDOSAGE

Overdosage can lead to acute profound water loss, volume and electrolyte depletion, dehydration, reduction of blood volume and circulatory collapse with a possibility of vascular thrombosis and embolism.

Electrolyte depletion may be manifested by weakness, dizziness, mental confusion, anorexia, lethargy, vomiting and cramps.

Treatment consists of replacement of fluid and electrolyte losses by careful monitoring of the urine and electrolyte output and serum electrolyte levels.

DESCRIPTION

Bumetanide is a loop diuretic, available as scored tablets, 0.5 mg (light green), 1 mg (yellow) and 2 mg (peach) for oral administration; each tablet also contains: anhydrous lactose, magnesium stearate, microcrystalline cellulose, corn starch and talc, with the following colorants: 0.5 mg (D&C Yellow No.

10 Aluminum Lake and FD&C Blue No.

1 Aluminum Lake); 1 mg (D&C Yellow No.

10 Aluminum Lake); 2 mg (Ferric oxide red).

Chemically, bumetanide is 3-(butylamino)-4-phenoxy-5-sulfamoyl-benzoic acid.

It is a practically white powder.

It is slightly soluble in water and soluble in alkaline solutions.

It has the following structural formula: Image of Structural Formula

HOW SUPPLIED

Bumetanide Tablets USP are available as light green, flat, round, bevel-edged tablets, debossed “0.5” on one side and bisected on the other side with company logo on the upper half and “4232” on the lower, containing 0.5 mg bumetanide, USP packaged in bottles of 100 tablets.

Bumetanide Tablets USP are available as yellow, flat, round, bevel-edged tablets, debossed “1” on one side and bisected on the other side with company logo on the upper half and “4233” on the lower, containing 1 mg bumetanide, USP packaged in bottles of 100 and 1000 tablets.

Bumetanide Tablets USP are available as peach, flat, round, bevel-edged tablets, debossed “2” on one side and bisected on the other side with company logo on the upper half and “4234” on the lower, containing 2 mg bumetanide, USP packaged in bottles of 100 and 1000 tablets.

PHARMACIST: Dispense in a tight, light-resistant container as defined in the USP, with a child-resistant closure (as required).

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

Manufactured In India By: EMCURE PHARMACEUTICALS LTD.

Hinjwadi, Pune, India Manufactured For: TEVA PHARMACEUTICALS USA Sellersville, PA 18960 Rev.

A 3/2010

GERIATRIC USE

Geriatric Use Clinical studies of bumetanide did not include sufficient numbers of subjects aged 65 and over to determine whether they responded differently from younger subjects.

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

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

This drug is known to be substantially excreted by the kidney, and the risk of toxic reactions to this drug may be greater in patients with impaired renal function.

Because elderly patients are more likely to have decreased renal function, care should be taken in dose selection, and it may be useful to monitor renal function.

INDICATIONS AND USAGE

Bumetanide tablets are indicated for the treatment of edema associated with congestive heart failure, hepatic and renal disease, including the nephrotic syndrome.

Almost equal diuretic response occurs after oral and parenteral administration of bumetanide.

Therefore, if impaired gastrointestinal absorption is suspected or oral administration is not practical, bumetanide should be given by the intramuscular or intravenous route.

Successful treatment with bumetanide following instances of allergic reactions to furosemide suggests a lack of cross-sensitivity.

PEDIATRIC USE

Pediatric Use Safety and effectiveness in pediatric patients below the age of 18 have not been established.

In vitro studies using pooled sera from critically ill neonates have shown bumetanide to be a potent displacer of bilirubin (see CLINICAL PHARMACOLOGY , Pediatric Pharmacology ).

The administration of bumetanide could present a particular concern if given to critically ill or jaundiced neonates at risk for kernicterus.

NUSRING MOTHERS

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

As a general rule, nursing should not be undertaken while the patient is on bumetanide since it may be excreted in human milk.

BOXED WARNING

WARNING Bumetanide is a potent diuretic which, if given in excessive amounts, can lead to a profound diuresis with water and electrolyte depletion.

Therefore, careful medical supervision is required, and dose and dosage schedule have to be adjusted to the individual patient’s needs.

(See DOSAGE AND ADMINISTRATION.

)

DOSAGE AND ADMINISTRATION

Dosage should be individualized with careful monitoring of patient response.

Oral Administration The usual total daily dosage of bumetanide is 0.5 to 2 mg and in most patients is given as a single dose.

If the diuretic response to an initial dose of bumetanide is not adequate, in view of its rapid onset and short duration of action, a second or third dose may be given at 4- to 5-hour intervals up to a maximum daily dose of 10 mg.

An intermittent dose schedule, whereby bumetanide is given on alternate days or for 3 to 4 days with rest periods of 1 to 2 days in between, is recommended as the safest and most effective method for the continued control of edema.

In patients with hepatic failure, the dosage should be kept to a minimum, and if necessary, dosage increased very carefully.

Because cross-sensitivity with furosemide has rarely been observed, bumetanide can be substituted at approximately a 1:40 ratio of bumetanide to furosemide in patients allergic to furosemide.

Parenteral Administration Bumetanide Injection may be administered parenterally (IV or IM) to patients in whom gastrointestinal absorption may be impaired or in whom oral administration is not practical.

Parenteral treatment should be terminated and oral treatment instituted as soon as possible.

Flavoxate Hydrochloride 100 MG Oral Tablet

WARNINGS

Flavoxate hydrochloride should be given cautiously in patients with suspected glaucoma.

OVERDOSAGE

The oral LD 50 for flavoxate hydrochloride in rats is 4273 mg/kg.

The oral LD 50 for flavoxate hydrochloride in mice is 1837 mg/kg.

It is not known whether flavoxate hydrochloride is dialyzable.

DESCRIPTION

Flavoxate hydrochloride tablets contain flavoxate hydrochloride, a synthetic urinary tract spasmolytic.

Chemically, flavoxate hydrochloride is 2-piperidinoethyl 3-methyl-4-oxo-2-phenyl-4 H -1-benzopyran-8-carboxylate hydrochloride.

The empirical formula of flavoxate hydrochloride is C 24 H 25 NO 4 •HCl.

The molecular weight is 427.94.

The structural formula appears below.

Each tablet for oral administration contains 100 mg flavoxate hydrochloride.

In addition, each tablet contains the following inactive ingredients: colloidal silicon dioxide, corn starch, dibasic calcium phosphate dihydrate, hypromellose, polydextrose, magnesium stearate, titanium dioxide and triacetin.

Chemical Structure – Flavoxate

HOW SUPPLIED

Flavoxate hydrochloride 100 mg tablets are available as white, round biconvex, film-coated tablets, debossed “ Є 58 ” on one side and plain on the other side, and are available in bottles of 100 (NDC 60429-290-01) .

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

Dispense contents in a tight, light-resistant container.

Manufactured by: Epic Pharma, LLC Laurelton, NY 11413 Marketed/Packaged by: GSMS Inc.

Camarillo, CA 93012 Made in USA Issued 01/11 MF058IS01/11 OE1140

INDICATIONS AND USAGE

Flavoxate hydrochloride tablets are indicated for symptomatic relief of dysuria, urgency, nocturia, suprapubic pain, frequency and incontinence as may occur in cystitis, prostatitis, urethritis, urethrocystitis/urethrotrigonitis.

Flavoxate hydrochloride tablets are not indicated for definitive treatment, but are compatible with drugs used for the treatment of urinary tract infections.

PEDIATRIC USE

Pediatric Use Safety and effectiveness in children below the age of 12 years have not been established.

PREGNANCY

Pregnancy Teratogenic Effects-Pregnancy Category B.

Reproduction studies have been performed in rats and rabbits at doses up to 34 times the human dose and revealed no evidence of impaired fertility or harm to the fetus due to flavoxate hydrochloride.

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

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

NUSRING MOTHERS

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

Because many drugs are excreted in human milk, caution should be exercised when flavoxate hydrochloride is administered to a nursing woman.

INFORMATION FOR PATIENTS

Information for Patients Patients should be informed that if drowsiness and blurred vision occur, they should not operate a motor vehicle or machinery or participate in activities where alertness is required.

DOSAGE AND ADMINISTRATION

Adults and children over 12 years of age One or two 100 mg tablets 3 or 4 times a day.

With improvement of symptoms, the dose may be reduced.

This drug cannot be recommended for infants and children under 12 years of age because safety and efficacy have not been demonstrated in this age group.

Clonidine Hydrochloride 0.2 MG Oral Tablet

WARNINGS

Withdrawal: Patients should be instructed not to discontinue therapy without consulting their physician.

Sudden cessation of clonidine treatment has, in some cases, resulted in symptoms such as nervousness, agitation, headache, and tremor accompanied or followed by a rapid rise in blood pressure and elevated catecholamine concentrations in the plasma.

The likelihood of such reactions to discontinuation of clonidine therapy appears to be greater after administration of higher doses or continuation of concomitant beta-blocker treatment and special caution is therefore advised in these situations.

Rare instances of hypertensive encephalopathy, cerebrovascular accidents and death have been reported after clonidine withdrawal.

When discontinuing therapy with clonidine hydrochloride tablets, the physician should reduce the dose gradually over 2 to 4 days to avoid withdrawal symptomatology.

An excessive rise in blood pressure following discontinuation of clonidine hydrochloride tablets therapy can be reversed by administration of oral clonidine hydrochloride or by intravenous phentolamine.

If therapy is to be discontinued in patients receiving a beta-blocker and clonidine concurrently, the beta-blocker should be withdrawn several days before the gradual discontinuation of clonidine hydrochloride tablets.

Because children commonly have gastrointestinal illnesses that lead to vomiting, they may be particularly susceptible to hypertensive episodes resulting from abrupt inability to take medication.

OVERDOSAGE

Hypertension may develop early and may be followed by hypotension, bradycardia, respiratory depression, hypothermia, drowsiness, decreased or absent reflexes, weakness, irritability and miosis.

The frequency of CNS depression may be higher in children than adults.

Large overdoses may result in reversible cardiac conduction defects or dysrhythmias, apnea, coma and seizures.

Signs and symptoms of overdose generally occur within 30 minutes to two hours after exposure.

As little as 0.1 mg of clonidine has produced signs of toxicity in children.

There is no specific antidote for clonidine overdosage.

Clonidine overdosage may result in the rapid development of CNS depression; therefore, induction of vomiting with ipecac syrup is not recommended.

Gastric lavage may be indicated following recent and/or large ingestions.

Administration of activated charcoal and/or a cathartic may be beneficial.

Supportive care may include atropine sulfate for bradycardia, intravenous fluids and/or vasopressor agents for hypotension and vasodilators for hypertension.

Naloxone may be a useful adjunct for the management of clonidine-induced respiratory depression, hypotension and/or coma; blood pressure should be monitored since the administration of naloxone has occasionally resulted in paradoxical hypertension.

Tolazoline administration has yielded inconsistent results and is not recommended as first-line therapy.

Dialysis is not likely to significantly enhance the elimination of clonidine.

The largest overdose reported to date involved a 28-year old male who ingested 100 mg of clonidine hydrochloride powder.

This patient developed hypertension followed by hypotension, bradycardia, apnea, hallucinations, semicoma, and premature ventricular contractions.

The patient fully recovered after intensive treatment.

Plasma clonidine levels were 60 ng/mL after 1 hour, 190 ng/mL after 1.5 hours, 370 ng/mL after 2 hours, and 120 ng/mL after 5.5 and 6.5 hours.

In mice and rats, the oral LD 50 of clonidine is 206 and 465 mg/kg, respectively.

DESCRIPTION

Clonidine hydrochloride, USP is a centrally acting alpha-agonist hypotensive agent available as tablets for oral administration in three dosage strengths: 0.1 mg, 0.2 mg, and 0.3 mg.

The 0.1 mg tablet is equivalent to 0.087 mg of the free base.

The following inactive ingredients are contained in these products: corn starch, D&C yellow #10 Aluminum Lake, FD&C yellow #6 Aluminum Lake (Sunset Yellow Lake), lactose monohydrate, magnesium stearate, and sodium starch glycolate.

Clonidine hydrochloride, USP is an imidazoline derivative and exists as a mesomeric compound.

The chemical name is 2-(2,6-dichlorophenylamino)-2-imidazoline hydrochloride.

The following is the structural formula: Clonidine hydrochloride, USP is an odorless, bitter, white, crystalline substance soluble in water and alcohol.

836600d0-figure-01

HOW SUPPLIED

Clonidine hydrochloride tablets, USP are supplied as follows: 0.2 mg — Each orange, round tablet imprinted with on one side and 128 and bisect on the other side contains 0.2 mg of clonidine hydrochloride USP.

Bottle of 30 – 68788-9213-3 Bottle of 60 – 68788-9213-6 Bottle of 90 – 68788-9213-9 Dispense in a tight, light-resistant container as defined in the USP.

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

Manufactured by: Actavis Elizabeth LLC Elizabeth, NJ 07207 USA Distributed by: Actavis Pharma, Inc.

Parsippany, NJ 07054 USA 40-9183 Revised — October 2015 Repackaged by Preferred Pharmaceuticals, Inc.

0.2mg image

INDICATIONS AND USAGE

Clonidine hydrochloride tablets, USP are indicated in the treatment of hypertension.

Clonidine hydrochloride tablets may be employed alone or concomitantly with other antihypertensive agents.

DOSAGE AND ADMINISTRATION

Adults: The dose of clonidine hydrochloride tablets, USP must be adjusted according to the patient’s individual blood pressure response.

The following is a general guide to its administration.

Initial Dose: 0.1 mg tablet twice daily (morning and bedtime).

Elderly patients may benefit from a lower initial dose.

Maintenance Dose: Further increments of 0.1 mg per day may be made at weekly intervals if necessary until the desired response is achieved.

Taking the larger portion of the oral daily dose at bedtime may minimize transient adjustment effects of dry mouth and drowsiness.

The therapeutic doses most commonly employed have ranged from 0.2 mg to 0.6 mg per day given in divided doses.

Studies have indicated that 2.4 mg is the maximum effective daily dose, but doses as high as this have rarely been employed.

Renal Impairment: Patients with renal impairment may benefit from a lower initial dose.

Patients should be carefully monitored.

Since only a minimal amount of clonidine is removed during routine hemodialysis, there is no need to give supplemental clonidine following dialysis.

For questions regarding this product call Actavis at 1-800-432-8534.

Progesterone 100 MG Oral Capsule [Prometrium]

WARNINGS

See BOXED WARNING .

1.

Cardiovascular disorders An increased risk of pulmonary embolism, deep vein thrombosis (DVT), stroke, and myocardial infarction has been reported with estrogen plus progestin therapy.

Should any of these occur or be suspected, estrogen with progestin therapy should be discontinued immediately.

Risk factors for arterial vascular disease (for example, hypertension, diabetes mellitus, tobacco use, hypercholesterolemia, and obesity) and/or venous thromboembolism (for example, personal history or family history of venous thromboembolism [VTE], obesity, and systemic lupus erythematosus) should be managed appropriately.

a.

Stroke In the Women’s Health Initiative (WHI) estrogen plus progestin substudy, a statistically significant increased risk of stroke was reported in women 50 to 79 years of age receiving daily CE (0.625 mg) plus MPA (2.5 mg) compared to women in the same age group receiving placebo (33 versus 25 per 10,000 women-years).

The increase in risk was demonstrated after the first year and persisted.

(See CLINICAL STUDIES .) Should a stroke occur or be suspected, estrogen plus progestin therapy should be discontinued immediately.

b.

Coronary Heart Disease In the WHI estrogen plus progestin substudy, there was a statistically non-significant increased risk of coronary heart disease (CHD) events (defined as nonfatal myocardial infarction [MI], silent MI, or CHD death) reported in women receiving daily CE (0.625 mg) plus MPA (2.5 mg) compared to women receiving placebo (41 versus 34 per 10,000 women-years).

An increase in relative risk was demonstrated in year 1 and a trend toward decreasing relative risk was reported in years 2 through 5.

(See CLINICAL STUDIES .) In postmenopausal women with documented heart disease (n = 2,763, average age 66.7 years), in a controlled clinical trial of secondary prevention of cardiovascular disease (Heart and Estrogen/Progestin Replacement Study [HERS]), treatment with daily CE (0.625 mg) plus MPA (2.5 mg) demonstrated no cardiovascular benefit.

During an average follow-up of 4.1 years, treatment with CE plus MPA did not reduce the overall rate of CHD events in postmenopausal women with established coronary heart disease.

There were more CHD events in the CE plus MPA-treated group than in the placebo group in year 1, but not during the subsequent years.

Two thousand, three hundred and twenty-one (2,321) women from the original HERS trial agreed to participate in an open-label extension of HERS, HERS II.

Average follow-up in HERS II was an additional 2.7 years, for a total of 6.8 years overall.

Rates of CHD events were comparable among women in the CE plus MPA group and the placebo group in HERS, HERS II, and overall.

c.

Venous Thromboembolism In the WHI estrogen plus progestin substudy, a statistically significant 2-fold greater rate of VTE (DVT and pulmonary embolism [PE]) was reported in women receiving daily CE (0.625 mg) plus MPA (2.5 mg) compared to women receiving placebo (35 versus 17 per 10,000 women-years).

Statistically significant increases in risk for both DVT (26 versus 13 per 10,000 women-years) and PE (18 versus 8 per 10,000 women-years) were also demonstrated.

The increase in VTE risk was demonstrated during the first year and persisted.

(See CLINICAL STUDIES .) Should a VTE occur or be suspected, estrogen plus progestin therapy should be discontinued immediately.

If feasible, estrogens with progestins should be discontinued at least 4 to 6 weeks before surgery of the type associated with an increased risk of thromboembolism, or during periods of prolonged immobilization.

2.

Malignant neoplasms a.

Breast Cancer The most important randomized clinical trial providing information about breast cancer in estrogen plus progestin users is the Women’s Health Initiative (WHI) substudy of daily CE (0.625 mg) plus MPA (2.5 mg).

After a mean follow-up of 5.6 years, the estrogen plus progestin substudy reported an increased risk of invasive breast cancer in women who took daily CE plus MPA.

In this substudy, prior use of estrogen-alone or estrogen plus progestin therapy was reported by 26 percent of the women.

The relative risk of invasive breast cancer was 1.24 (95 percent nCI, 1.01-1.54), and the absolute risk was 41 versus 33 cases per 10,000 women-years, for CE plus MPA compared with placebo.

Among women who reported prior use of hormone therapy, the relative risk of invasive breast cancer was 1.86, and the absolute risk was 46 versus 25 cases per 10,000 women-years, for estrogen plus progestin compared with placebo.

Among women who reported no prior use of hormone therapy, the relative risk of invasive breast cancer was 1.09, and the absolute risk was 40 versus 36 cases per 10,000 women-years for CE plus MPA compared with placebo.

In the same substudy, invasive breast cancers were larger, were more likely to be node positive, and were diagnosed at a more advanced stage in the CE (0.625 mg) plus MPA (2.5 mg) group compared with the placebo group.

Metastatic disease was rare, with no apparent difference between the two groups.

Other prognostic factors such as histologic subtype, grade and hormone receptor status did not differ between the groups.

(See CLINICAL STUDIES .) Consistent with the WHI clinical trials, observational studies have also reported an increased risk of breast cancer for estrogen plus progestin therapy, and a smaller increased risk for estrogen-alone therapy, after several years of use.

The risk increased with duration of use, and appeared to return to baseline over about 5 years after stopping treatment (only the observational studies have substantial data on risk after stopping).

Observational studies also suggest that the risk of breast cancer was greater, and became apparent earlier, with estrogen plus progestin therapy as compared to estrogen-alone therapy.

However, these studies have not generally found significant variation in the risk of breast cancer among different estrogen plus progestin combinations, doses, or routes of administration.

The use of estrogen plus progestin has been reported to result in an increase in abnormal mammograms requiring further evaluation.

All women should receive yearly breast examinations by a healthcare provider and perform monthly breast self-examinations.

In addition, mammography examinations should be scheduled based on patient age, risk factors, and prior mammogram results.

b.

Endometrial Cancer An increased risk of endometrial cancer has been reported with the use of unopposed estrogen therapy in a woman with a uterus.

The reported endometrial cancer risk among unopposed estrogen users is about 2 to 12 times greater than in non-users, and appears dependent on duration of treatment and on estrogen dose.

Most studies show no significant increased risk associated with the use of estrogens for less than 1 year.

The greatest risk appears associated with prolonged use, with increased risks of 15- to 24-fold for 5 to 10 years or more and this risk has been shown to persist for at least 8 to 15 years after estrogen therapy is discontinued.

Clinical surveillance of all women using estrogen plus progestin therapy is important.

Adequate diagnostic measures, including directed or random endometrial sampling when indicated, should be undertaken to rule out malignancy in all cases of undiagnosed persistent or recurring abnormal genital bleeding.

There is no evidence that the use of natural estrogens results in a different endometrial risk profile than synthetic estrogens of equivalent estrogen dose.

Adding a progestin to estrogen therapy in postmenopausal women has been shown to reduce the risk of endometrial hyperplasia, which may be a precursor to endometrial cancer.

c.

Ovarian Cancer The WHI estrogen plus progestin substudy reported a statistically non-significant increased risk of ovarian cancer.

After an average follow-up of 5.6 years, the relative risk for ovarian cancer for CE plus MPA versus placebo was 1.58 (95 percent nCI, 0.77 – 3.24).

The absolute risk for CE plus MPA versus placebo was 4 versus 3 cases per 10,000 women-years.

In some epidemiologic studies, the use of estrogen plus progestin and estrogen-only products, in particular for 5 or more years, has been associated with an increased risk of ovarian cancer.

However, the duration of exposure associated with increased risk is not consistent across all epidemiologic studies and some report no association.

3.

Probable dementia In the estrogen plus progestin Women’s Health Initiative Memory Study (WHIMS), an ancillary study of WHI, a population of 4,532 postmenopausal women 65 to 79 years of age was randomized to daily CE (0.625 mg) plus MPA (2.5 mg) or placebo.

In the WHIMS estrogen plus progestin ancillary study, after an average follow-up of 4 years, 40 women in the CE plus MPA group and 21 women in the placebo group were diagnosed with probable dementia.

The relative risk of probable dementia for estrogen plus progestin versus placebo was 2.05 (95 percent CI, 1.21-3.48).

The absolute risk of probable dementia for CE plus MPA versus placebo was 45 versus 22 cases per 10,000 women-years.

It is unknown whether these findings apply to younger postmenopausal women.

(See CLINICAL STUDIES and PRECAUTIONS, Geriatric Use .) 4.

Vision abnormalities Retinal vascular thrombosis has been reported in patients receiving estrogen.

Discontinue estrogen plus progestin therapy pending examination if there is sudden partial or complete loss of vision, or if there is a sudden onset of proptosis, diplopia or migraine.

If examination reveals papilledema or retinal vascular lesions, estrogen plus progestin therapy should be permanently discontinued.

DRUG INTERACTIONS

G.

Drug Interactions The metabolism of progesterone by human liver microsomes was inhibited by ketoconazole (IC 50 < 0.1 μM).

Ketoconazole is a known inhibitor of cytochrome P450 3A4, hence these data suggest that ketoconazole or other known inhibitors of this enzyme may increase the bioavailability of progesterone.

The clinical relevance of the in vitro findings is unknown.

Coadministration of conjugated estrogens and PROMETRIUM Capsules to 29 postmenopausal women over a 12-day period resulted in an increase in total estrone concentrations (C max 3.68 ng/mL to 4.93 ng/mL) and total equilin concentrations (C max 2.27 ng/mL to 3.22 ng/mL) and a decrease in circulating 17β estradiol concentrations (C max 0.037 ng/mL to 0.030 ng/mL).

The half-life of the conjugated estrogens was similar with coadministration of PROMETRIUM Capsules.

Table 2 summarizes the pharmacokinetic parameters.

TABLE 2.

Mean (± S.D.) Pharmacokinetic Parameters for Estradiol, Estrone, and Equilin Following Coadministration of Conjugated Estrogens 0.625 mg and PROMETRIUM Capsules 200 mg for 12 Days to Postmenopausal Women a Total estrogens is the sum of conjugated and unconjugated estrogen.

Conjugated Estrogens Conjugated Estrogens plus PROMETRIUM Capsules Drug C max (ng/mL) T max (hr) AUC (0-24h) (ng × h/mL) C max (ng/mL) T max (hr) AUC (0-24h) (ng × h/mL) Estradiol 0.037 ± 0.048 12.7 ± 9.1 0.676 ± 0.737 0.030 ± 0.032 17.32 ± 1.21 0.561 ± 0.572 Estrone Total a 3.68 ± 1.55 10.6 ± 6.8 61.3 ± 26.36 4.93 ± 2.07 7.5 ± 3.8 85.9 ± 41.2 Equilin Total a 2.27 ± 0.95 6.0 ± 4.0 28.8 ± 13.0 3.22 ± 1.13 5.3 ± 2.6 38.1 ± 20.2

OVERDOSAGE

No studies on overdosage have been conducted in humans.

In the case of overdosage, PROMETRIUM Capsules should be discontinued and the patient should be treated symptomatically.

DESCRIPTION

PROMETRIUM (progesterone, USP) Capsules contain micronized progesterone for oral administration.

Progesterone has a molecular weight of 314.47 and a molecular formula of C 21 H 30 O 2 .

Progesterone (pregn-4-ene-3, 20-dione) is a white or creamy white, odorless, crystalline powder practically insoluble in water, soluble in alcohol, acetone and dioxane and sparingly soluble in vegetable oils, stable in air, melting between 126° and 131°C.

The structural formula is: Progesterone is synthesized from a starting material from a plant source and is chemically identical to progesterone of human ovarian origin.

PROMETRIUM Capsules are available in multiple strengths to afford dosage flexibility for optimum management.

PROMETRIUM Capsules contain 100 mg or 200 mg micronized progesterone.

The inactive ingredients for PROMETRIUM Capsules 100 mg include: peanut oil NF, gelatin NF, glycerin USP, lecithin NF, titanium dioxide USP, FD&C Red No.

40, and D&C Yellow No.

10.

The inactive ingredients for PROMETRIUM Capsules 200 mg include: peanut oil NF, gelatin NF, glycerin USP, lecithin NF, titanium dioxide USP, D&C Yellow No.

10, and FD&C Yellow No.

6.

Structural Formula

CLINICAL STUDIES

Effects on the endometrium In a randomized, double-blind clinical trial, 358 postmenopausal women, each with an intact uterus, received treatment for up to 36 months.

The treatment groups were: PROMETRIUM Capsules at the dose of 200 mg per day for 12 days per 28-day cycle in combination with conjugated estrogens 0.625 mg per day (n=120); conjugated estrogens 0.625 mg per day only (n=119); or placebo (n=119).

The subjects in all three treatment groups were primarily Caucasian women (87 percent or more of each group).

The results for the incidence of endometrial hyperplasia in women receiving up to 3 years of treatment are shown in Table 3 .

A comparison of the PROMETRIUM Capsules plus conjugated estrogens treatment group to the conjugated estrogens only group showed a significantly lower rate of hyperplasia (6 percent combination product versus 64 percent estrogen alone) in the PROMETRIUM Capsules plus conjugated estrogens treatment group throughout 36 months of treatment.

TABLE 3.

Incidence of Endometrial Hyperplasia in Women Receiving 3 Years of Treatment a Most advanced result to least advanced result: Adenocarcinoma > atypical hyperplasia > complex hyperplasia > simple hyperplasia Endometrial Diagnosis Treatment Group Conjugated Estrogens 0.625 mg + PROMETRIUM Capsules 200 mg (cyclical) Conjugated Estrogens 0.625 mg (alone) Placebo Number of patients % of patients Number of patients % of patients Number of patients % of patients n=117 n=115 n=116 HYPERPLASIA a 7 6 74 64 3 3 Adenocarcinoma 0 0 0 0 1 1 Atypical hyperplasia 1 1 14 12 0 0 Complex hyperplasia 0 0 27 23 1 1 Simple hyperplasia 6 5 33 29 1 1 The times to diagnosis of endometrial hyperplasia over 36 months of treatment are shown in Figure 1 .

This figure illustrates graphically that the proportion of patients with hyperplasia was significantly greater for the conjugated estrogens group (64 percent) compared to the conjugated estrogens plus PROMETRIUM Capsules group (6 percent).

Figure 1.

Time to Hyperplasia in Women Receiving up to 36 Months of Treatment The discontinuation rates due to hyperplasia over the 36 months of treatment are as shown in Table 4 .

For any degree of hyperplasia, the discontinuation rate for patients who received conjugated estrogens plus PROMETRIUM Capsules was similar to that of the placebo only group, while the discontinuation rate for patients who received conjugated estrogens alone was significantly higher.

Women who permanently discontinued treatment due to hyperplasia were similar in demographics to the overall study population.

TABLE 4.

Discontinuation Rate Due to Hyperplasia Over 36 Months of Treatment Most Advanced Biopsy Result Through 36 Months of Treatment Treatment Group Conjugated Estrogens + PROMETRIUM Capsules (cyclical) Conjugated Estrogens (alone) Placebo n=120 n=119 n=119 Number of patients % of patients Number of patients % of patients Number of patients % of patients Adenocarcinoma 0 0 0 0 1 1 Atypical hyperplasia 1 1 10 8 0 0 Complex hyperplasia 0 0 21 18 1 1 Simple hyperplasia 1 1 13 11 0 0 Figure 1 Effects on secondary amenorrhea In a single-center, randomized, double-blind clinical study that included premenopausal women with secondary amenorrhea for at least 90 days, administration of 10 days of PROMETRIUM Capsules therapy resulted in 80 percent of women experiencing withdrawal bleeding within 7 days of the last dose of PROMETRIUM Capsules, 300 mg per day (n=20), compared to 10 percent of women experiencing withdrawal bleeding in the placebo group (n=21).

In a multicenter, parallel-group, open label, postmarketing dosing study that included premenopausal women with secondary amenorrhea for at least 90 days, administration of 10 days of PROMETRIUM Capsules during two 28-day treatment cycles, 300 mg per day (n=107) or 400 mg per day (n=99), resulted in 73.8 percent and 76.8 percent of women, respectively, experiencing withdrawal bleeding.

The rate of secretory transformation was evaluated in a multicenter, randomized, double-blind clinical study in estrogen-primed postmenopausal women.

PROMETRIUM Capsules administered orally for 10 days at 400 mg per day (n=22) induced complete secretory changes in the endometrium in 45 percent of women compared to 0 percent in the placebo group (n=23).

A second multicenter, parallel-group, open label postmarketing dosing study in premenopausal women with secondary amenorrhea for at least 90 days also evaluated the rate of secretory transformation.

All subjects received daily oral conjugated estrogens over 3 consecutive 28-day treatment cycles and PROMETRIUM Capsules, 300 mg per day (n=107) or 400 mg per day (n=99) for 10 days of each treatment cycle.

The rate of complete secretory transformation was 21.5 percent and 28.3 percent, respectively.

Women’s Health Initiative Studies The Women’s Health Initiative (WHI) enrolled approximately 27,000 predominantly healthy postmenopausal women in two substudies to assess the risks and benefits of daily oral conjugated estrogens (CE) [0.625 mg]-alone or in combination with medroxyprogesterone acetate (MPA) [2.5 mg] compared to placebo in the prevention of certain chronic diseases.

The primary endpoint was the incidence of coronary heart disease [(CHD) defined as nonfatal myocardial infarction (MI), silent MI and CHD death], with invasive breast cancer as the primary adverse outcome.

A “global index” included the earliest occurrence of CHD, invasive breast cancer, stroke, pulmonary embolism (PE), endometrial cancer (only in the CE plus MPA substudy), colorectal cancer, hip fracture, or death due to other cause.

These sub studies did not evaluate the effects of CE–alone or CE plus MPA on menopausal symptoms.

WHI Estrogen Plus Progestin Substudy The WHI estrogen plus progestin substudy was stopped early.

According to the predefined stopping rule, after an average follow-up of 5.6 years of treatment, the increased risk of breast cancer and cardiovascular events exceeded the specified benefits included in the “global index.” The absolute excess risk of events in the “global index” was 19 per 10,000 women-years.

For those outcomes included in the WHI “global index” that reached statistical significance after 5.6 years of follow-up, the absolute excess risks per 10,000 women-years in the group treated with CE plus MPA were 7 more CHD events, 8 more strokes, 10 more PEs, and 8 more invasive breast cancers, while the absolute risk reductions per 10,000 women-years were 6 fewer colorectal cancers and 5 fewer hip fractures.

Results of the estrogen plus progestin substudy, which included 16,608 women (average 63 years of age, range 50 to 79; 83.9 percent White, 6.8 percent Black, 5.4 percent Hispanic, 3.9 percent Other) are presented in Table 5 .

These results reflect centrally adjudicated data after an average follow-up of 5.6 years.

TABLE 5.

Relative and Absolute Risk Seen in the Estrogen Plus Progestin Substudy of WHI at an Average of 5.6 Years a, b a Adapted from numerous WHI publications.

WHI publications can be viewed at www.nhlbi.nih.gov/whi.

b Results are based on centrally adjudicated data.

c Nominal confidence intervals unadjusted for multiple looks and multiple comparisons.

d Not included in Global Index.

e Includes metastatic and non-metastatic breast cancer with the exception of in situ breast cancer.

f All deaths, except from breast or colorectal cancer, definite or probable CHD, PE or cerebrovascular disease.

g A subset of the events was combined in a “global index” defined as the earliest occurrence of CHD events, invasive breast cancer, stroke, pulmonary embolism, endometrial cancer, colorectal cancer, hip fracture, or death due to other causes.

Event Relative Risk CE/MPA versus Placebo (95% nCI c ) CE/MPA n = 8,506 Placebo n = 8,102 Absolute Risk per 10,000 Women-Years CHD events 1.23 (0.99-1.53) 41 34 Non-fatal MI 1.28 (1.00-1.63) 31 25 CHD death 1.10 (0.70-1.75) 8 8 All stroke 1.31 (1.03-1.88) 33 25 Ischemic Stroke 1.44 (1.09-1.90) 26 18 Deep vein thrombosis d 1.95 (1.43-2.67) 26 13 Pulmonary embolism 2.13 (1.45-3.11) 18 8 Invasive breast cancer e 1.24 (1.01-1.54) 41 33 Colorectal cancer 0.61 (0.42-0.87) 10 16 Endometrial cancer d 0.81 (0.48-1.36) 6 7 Cervical cancer d 1.44 (0.47-4.42) 2 1 Hip fracture 0.67 (0.47-0.96) 11 16 Vertebral fractures d 0.65 (0.46-0.92) 11 17 Lower arm/wrist fractures d 0.71 (0.59-0.85) 44 62 Total fractures d 0.76 (0.69-0.83) 152 199 Overall mortality f 1.00 (0.83-1.19) 52 52 Global Index g 1.13 (1.02-1.25) 184 165 Timing of the initiation of estrogen plus progestin therapy relative to the start of menopause may affect the overall risk benefit profile.

The WHI estrogen plus progestin substudy stratified for age showed in women 50 to 59 years of age a non-significant trend toward reducing risk of overall mortality [hazard ratio (HR) 0.69 (95 percent CI, 0.44-1.07)].

Women’s Health Initiative Memory Study The estrogen plus progestin Women’s Health Initiative Memory Study (WHIMS), an ancillary study of WHI, enrolled 4,532 predominantly healthy postmenopausal women 65 years of age and older (47 percent were 65 to 69 years of age; 35 percent were 70 to 74 years of age; and 18 percent were 75 years of age and older) to evaluate the effects of daily CE (0.625 mg) plus MPA (2.5 mg) on the incidence of probable dementia (primary outcome) compared to placebo.

After an average follow-up of 4 years, the relative risk of probable dementia for CE plus MPA versus placebo was 2.05 (95 percent CI, 1.21 – 3.48).

The absolute risk of probable dementia for CE plus MPA versus placebo was 45 versus 22 per 10,000 women-years.

Probable dementia as defined in this study included Alzheimer’s disease (AD), vascular dementia (VaD) and mixed type (having features of both AD and VaD).

The most common classification of probable dementia in the treatment group and the placebo group was AD.

Since the ancillary study was conducted in women 65 to 79 years of age, it is unknown whether these findings apply to younger postmenopausal women.

(See WARNINGS, Probable dementia and PRECAUTIONS, Geriatric Use .)

HOW SUPPLIED

PROMETRIUM (progesterone, USP) Capsules 100 mg are round, peach-colored capsules branded with black imprint “SV.” NDC 0032-1708-01 (Bottle of 100) PROMETRIUM (progesterone, USP) Capsules 200 mg are oval, pale yellow-colored capsules branded with black imprint “SV2.” NDC 0032-1711-01 (Bottle of 100) Store at 25°C (77°F); excursions permitted to 15° to 30°C (59° to 86°F) [See USP Controlled Room Temperature].

Protect from excessive moisture.

Dispense in tight, light-resistant container as defined in USP/NF, accompanied by a Patient Insert.

Keep out of reach of children.

Manufactured by: Catalent Pharma Solutions St.

Petersburg, FL 33716 Marketed by: AbbVie Inc.

North Chicago, IL 60064, USA © AbbVie Inc.

2013

GERIATRIC USE

H.

Geriatric Use There have not been sufficient numbers of geriatric women involved in clinical studies utilizing PROMETRIUM Capsules to determine whether those over 65 years of age differ from younger subjects in their response to PROMETRIUM Capsules.

The Women’s Health Initiative Study In the Women’s Health Initiative (WHI) estrogen plus progestin substudy (daily CE [0.625 mg] plus MPA [2.5 mg] versus placebo), there was a higher relative risk of nonfatal stroke and invasive breast cancer in women greater than 65 years of age.

(See CLINICAL STUDIES and WARNINGS, Cardiovascular disorders and Malignant neoplasms .) The Women’s Health Initiative Memory Study In the Women’s Health Initiative Memory Study (WHIMS) of postmenopausal women 65 to 79 years of age, there was an increased risk of developing probable dementia in the estrogen plus progestin ancillary study when compared to placebo.

(See CLINICAL STUDIES and WARNINGS, Probable dementia .)

INDICATIONS AND USAGE

PROMETRIUM Capsules are indicated for use in the prevention of endometrial hyperplasia in nonhysterectomized postmenopausal women who are receiving conjugated estrogens tablets.

They are also indicated for use in secondary amenorrhea.

PEDIATRIC USE

G.

Pediatric Use PROMETRIUM Capsules are not indicated in children.

Clinical studies have not been conducted in the pediatric population.

PREGNANCY

E.

Pregnancy PROMETRIUM Capsules should not be used during pregnancy.

(See CONTRAINDICATIONS ).

Pregnancy Category B: Reproductive studies have been performed in mice at doses up to 9 times the human oral dose, in rats at doses up to 44 times the human oral dose, in rabbits at a dose of 10 mcg/day delivered locally within the uterus by an implanted device, in guinea pigs at doses of approximately one-half the human oral dose and in rhesus monkeys at doses approximately the human dose, all based on body surface area, and have revealed little or no evidence of impaired fertility or harm to the fetus due to progesterone.

NUSRING MOTHERS

F.

Nursing Women Detectable amounts of progestin have been identified in the milk of nursing women receiving progestins.

Caution should be exercised when PROMETRIUM Capsules are administered to a nursing woman.

BOXED WARNING

WARNING: CARDIOVASCULAR DISORDERS, BREAST CANCER and PROBABLE DEMENTIA FOR ESTROGEN PLUS PROGESTIN THERAPY Cardiovascular Disorders and Probable Dementia Estrogens plus progestin therapy should not be used for the prevention of cardiovascular disease or dementia.

(See CLINICAL STUDIES and WARNINGS, Cardiovascular disorders and Probable dementia .) The Women’s Health Initiative (WHI) estrogen plus progestin substudy reported increased risks of deep vein thrombosis, pulmonary embolism, stroke and myocardial infarction in postmenopausal women (50 to 79 years of age) during 5.6 years of treatment with daily oral conjugated estrogens (CE) [0.625 mg] combined with medroxyprogesterone acetate (MPA) [2.5 mg], relative to placebo.

(See CLINICAL STUDIES and WARNINGS, Cardiovascular disorders .) The WHI Memory Study (WHIMS) estrogen plus progestin ancillary study of the WHI reported an increased risk of developing probable dementia in postmenopausal women 65 years of age or older during 4 years of treatment with daily CE (0.625 mg) combined with MPA (2.5 mg), relative to placebo.

It is unknown whether this finding applies to younger postmenopausal women.

(See CLINICAL STUDIES and WARNINGS, Probable dementia and PRECAUTIONS, Geriatric Use .) Breast Cancer The WHI estrogen plus progestin substudy also demonstrated an increased risk of invasive breast cancer.

(See CLINICAL STUDIES and WARNINGS, Malignant neoplasms, Breast Cancer .) In the absence of comparable data, these risks should be assumed to be similar for other doses of CE and MPA, and other combinations and dosage forms of estrogens and progestins.

Progestins with estrogens should be prescribed at the lowest effective doses and for the shortest duration consistent with treatment goals and risks for the individual woman.

INFORMATION FOR PATIENTS

PATIENT INFORMATION PROMETRIUM® (progesterone, USP) Capsules 100 mg Capsules 200 mg Read this PATIENT INFORMATION before you start taking PROMETRIUM Capsules and read what you get each time you refill your PROMETRIUM Capsules prescription.

There may be new information.

This information does not take the place of talking to your healthcare provider about your medical condition or your treatment.

WHAT IS THE MOST IMPORTANT INFORMATION I SHOULD KNOW ABOUT PROMETRIUM CAPSULES (A Progesterone Hormone)? Progestins with estrogens should not be used to prevent heart disease, heart attacks, strokes, or dementia.

Using progestins with estrogens may increase your chance of getting heart attacks, strokes, breast cancer, and blood clots.

Using progestins with estrogens may increase your chance of getting dementia, based on a study of women age 65 and older.

You and your healthcare provider should talk regularly about whether you still need treatment with PROMETRIUM Capsules.

THIS PRODUCT CONTAINS PEANUT OIL AND SHOULD NOT BE USED IF YOU ARE ALLERGIC TO PEANUTS.

What is PROMETRIUM Capsules? PROMETRIUM Capsules contain the female hormone called progesterone.

What is PROMETRIUM Capsules used for? Treatment of Menstrual Irregularities PROMETRIUM Capsules are used for the treatment of secondary amenorrhea (absence of menstrual periods in women who have previously had a menstrual period) due to a decrease in progesterone.

When you do not produce enough progesterone, menstrual irregularities can occur.

If your healthcare provider has determined your body does not produce enough progesterone on its own, PROMETRIUM Capsules may be prescribed to provide the progesterone you need.

Protection of the Endometrium (Lining of the Uterus) PROMETRIUM Capsules are used in combination with estrogen-containing medications in a postmenopausal woman with a uterus (womb).

Taking estrogen-alone increases the chance of developing a condition called endometrial hyperplasia that may lead to cancer of the lining of the uterus (womb).

The addition of a progestin is generally recommended for a woman with a uterus to reduce the chance of getting cancer of the uterus (womb).

Who should not take PROMETRIUM Capsules? Do not start taking PROMETRIUM Capsules if you: Are allergic to peanuts Have unusual vaginal bleeding Currently have or have had certain cancers Estrogen plus progestin treatment may increase the chance of getting certain types of cancers, including cancer of the breast or uterus.

If you have or have had cancer, talk with your healthcare provider about whether you should take PROMETRIUM Capsules.

Had a stroke or heart attack Currently have or have had blood clots Currently have or have had liver problems Are allergic to PROMETRIUM Capsules or any of its ingredients See the list of ingredients in PROMETRIUM Capsules at the end of this leaflet.

Think you may be pregnant Tell your healthcare provider: If you are breastfeeding.

The hormone in PROMETRIUM Capsules can pass into your breast milk.

About all of your medical problems.

Your healthcare provider may need to check you more carefully if you have certain conditions, such as asthma (wheezing), epilepsy (seizures), diabetes, migraine, endometriosis, lupus, problems with your heart, liver, thyroid, or kidneys, or have high calcium levels in your blood.

About all the medicines you take.

This includes prescription and nonprescription medicines, vitamins, and herbal supplements.

Some medicines may affect how PROMETRIUM Capsules work.

PROMETRIUM Capsules may also affect how your other medicines work.

How should I take PROMETRIUM Capsules? Prevention of Endometrial Hyperplasia: A postmenopausal woman with a uterus who is taking estrogens should take a single daily dose of 200 mg PROMETRIUM Capsules at bedtime for 12 continuous days per 28-day cycle.

Secondary Amenorrhea: PROMETRIUM Capsules may be given as a single daily dose of 400 mg at bedtime for 10 days.

PROMETRIUM Capsules are to be taken at bedtime as some women become very drowsy and/or dizzy after taking PROMETRIUM Capsules.

In a few cases, symptoms may include blurred vision, difficulty speaking, difficulty with walking, and feeling abnormal.

If you experience these symptoms, discuss them with your healthcare provider right away.

If you experience difficulty in swallowing PROMETRIUM Capsules, it is recommended that you take your daily dose at bedtime with a glass of water while in the standing position.

What are the possible side effects of PROMETRIUM Capsules? Side effects are grouped by how serious they are and how often they happen when you are treated: Serious, but less common side effects include: Risk to the Fetus: Cases of cleft palate, cleft lip, hypospadias, ventricular septal defect, patent ductus arteriosus, and other congenital heart defects.

Abnormal Blood Clotting: Stroke, heart attack, pulmonary embolus, visual loss or blindness.

Some of the warning signs of serious side effects include: Changes in vision or speech Sudden new severe headaches Severe pains in your chest or legs with or without shortness of breath, weakness and fatigue Dizziness and faintness Vomiting Call your healthcare provider right away if you get any of these warning signs, or any other unusual symptoms that concern you.

Less serious, but common side effects include: Headaches Breast pain Irregular vaginal bleeding or spotting Stomach or abdominal cramps, bloating Nausea and vomiting Hair loss Fluid retention Vaginal yeast infection These are not all the possible side effects of PROMETRIUM Capsules.

For more information, ask your healthcare provider or pharmacist for advice about side effects.

You may report side effects to AbbVie Inc.

at 1-800-633-9110 or to FDA at 1-800-FDA-1088.

What can I do to lower my chances of getting a serious side effect with PROMETRIUM Capsules? Talk with your healthcare provider regularly about whether you should continue taking PROMETRIUM Capsules.

See your healthcare provider right away if you get unusual vaginal bleeding while taking PROMETRIUM Capsules.

Have a pelvic exam, breast exam, and mammogram (breast X-ray) every year unless your healthcare provider tells you something else.

If members of your family have had breast cancer or if you have ever had breast lumps or an abnormal mammogram, you may need to have breast exams more often.

If you have high blood pressure, high cholesterol (fat in the blood), diabetes, are overweight, or if you use tobacco, you may have higher chances for getting heart disease.

Ask your healthcare provider for ways to lower your chances for getting heart disease.

General information about safe and effective use of PROMETRIUM Capsules Medicines are sometimes prescribed for conditions that are not mentioned in patient information leaflets.

Do not take PROMETRIUM Capsules for conditions for which it was not prescribed.

Your healthcare provider has prescribed this drug for you and you alone.

Do not give PROMETRIUM Capsules to other people, even if they have the same symptoms you have.

It may harm them.

PROMETRIUM Capsules should be taken as a single daily dose at bedtime.

Some women may experience extreme dizziness and/or drowsiness during initial therapy.

In a few cases, symptoms may include blurred vision, difficulty speaking, difficulty with walking, and feeling abnormal.

If you experience these symptoms, discuss them with your healthcare provider right away.

Use caution when driving a motor vehicle or operating machinery as dizziness or drowsiness may occur.

Keep PROMETRIUM Capsules out of the reach of children.

This leaflet provides a summary of the most important information about PROMETRIUM Capsules.

If you would like more information, talk with your healthcare provider or pharmacist.

You can ask for information about PROMETRIUM Capsules that is written for health professionals.

You can get more information by calling the toll free number 1-800-633-9110.

What are the ingredients in PROMETRIUM Capsules? Active ingredient: 100 mg or 200 mg micronized progesterone The inactive ingredients for PROMETRIUM Capsules 100 mg include: peanut oil NF, gelatin NF, glycerin USP, lecithin NF, titanium dioxide USP, FD&C Red No.

40, and D&C Yellow No.

10.

The inactive ingredients for PROMETRIUM Capsules 200 mg include: peanut oil NF, gelatin NF, glycerin USP, lecithin NF, titanium dioxide USP, D&C Yellow No.

10, and FD&C Yellow No.

6.

HOW SUPPLIED PROMETRIUM Capsules 100 mg are round, peach-colored capsules branded with black imprint “SV.” PROMETRIUM Capsules 200 mg are oval, pale yellow-colored capsules branded with black imprint “SV2.” Store at 25°C (77°F); excursions permitted to 15° to 30°C (59° to 86°F) [See USP Controlled Room Temperature].

Protect from excessive moisture.

Manufactured by: Catalent Pharma Solutions St.

Petersburg, FL 33716 Marketed by: AbbVie Inc.

North Chicago, IL 60064, USA © AbbVie Inc.

2013 500032 Rev 09/13 September, 2013

DOSAGE AND ADMINISTRATION

Prevention of Endometrial Hyperplasia PROMETRIUM Capsules should be given as a single daily dose at bedtime, 200 mg orally for 12 days sequentially per 28-day cycle, to a postmenopausal woman with a uterus who is receiving daily conjugated estrogens tablets.

Treatment of Secondary Amenorrhea PROMETRIUM Capsules may be given as a single daily dose of 400 mg at bedtime for 10 days.

Some women may experience difficulty swallowing PROMETRIUM Capsules.

For these women, PROMETRIUM Capsules should be taken with a glass of water while in the standing position.

Astepro 0.206 MG/ACTUAT Metered Dose Nasal Spray

DRUG INTERACTIONS

7 7.1 Central Nervous System Depressants Concurrent use of ASTEPRO Nasal Spray with alcohol or other central nervous system depressants should be avoided because reductions in alertness and impairment of central nervous system performance may occur [ see Warnings and Precautions (5.1) ].

7.2 Erythromycin and Ketoconazole Interaction studies investigating the cardiac effects, as measured by the corrected QT interval (QTc), of concomitantly administered oral azelastine hydrochloride and erythromycin or ketoconazole were conducted.

Oral erythromycin (500 mg three times daily for 7 days) had no effect on azelastine pharmacokinetics or QTc based on analyses of serial electrocardiograms.

Ketoconazole (200 mg twice daily for 7 days) interfered with the measurement of azelastine plasma concentrations on the analytic HPLC; however, no effects on QTc were observed [ see Clinical Pharmacology (12.2) and (12.3) ].

7.3 Cimetidine Cimetidine (400 mg twice daily) increased the mean C max and AUC of orally administered azelastine hydrochloride (4 mg twice daily) by approximately 65% [ see Clinical Pharmacology (12.3) ].

OVERDOSAGE

10 There have been no reported overdosages with ASTEPRO.

Acute overdosage by adults with this dosage form is unlikely to result in clinically significant adverse events, other than increased somnolence, since one 30-mL bottle of ASTEPRO 0.1% contains up to 30 mg of azelastine hydrochloride and one 30-mL bottle of ASTEPRO 0.15% contains up to 45 mg of azelastine hydrochloride.

Clinical trials in adults with single doses of the oral formulation of azelastine hydrochloride (up to 16 mg) have not resulted in increased incidence of serious adverse events.

General supportive measures should be employed if overdosage occurs.

There is no known antidote to ASTEPRO.

Oral ingestion of antihistamines has the potential to cause serious adverse effects in children.

Accordingly, ASTEPRO should be kept out of the reach of children.

DESCRIPTION

11 ASTEPRO (azelastine hydrochloride) 0.1% nasal spray is an antihistamine (H 1 receptor antagonist) formulated as a metered-spray solution for intranasal administration.

ASTEPRO (azelastine hydrochloride) 0.15% nasal spray is an antihistamine (H 1 receptor antagonist) formulated as a metered-spray solution for intranasal administration.

Azelastine hydrochloride occurs as a white, almost odorless, crystalline powder with a bitter taste.

It has a molecular weight of 418.37.

It is sparingly soluble in water, methanol, and propylene glycol and slightly soluble in ethanol, octanol, and glycerine.

It has a melting point of about 225°C and the pH of a saturated solution is between 5.0 and 5.4.

Its chemical name is (±)-1-(2H)-phthalazinone,4-[(4-chlorophenyl) methyl]-2-(hexahydro-1-methyl-1H-azepin-4-yl)-, monohydrochloride.

Its molecular formula is C 22 H 24 ClN 3 O•HCl with the following chemical structure: ASTEPRO 0.1% contains 0.1% azelastine hydrochloride in an isotonic aqueous solution containing sorbitol, sucralose, hypromellose, sodium citrate, edetate disodium, benzalkonium chloride (125 mcg/mL), and purified water (pH 6.4).

After priming [ see Dosage and Administration (2.3) ], each metered spray delivers a 0.137 mL mean volume containing 137 mcg of azelastine hydrochloride (equivalent to 125 mcg of azelastine base).

The 30-mL (net weight 30 gm of solution) bottle provides 200 metered sprays.

ASTEPRO 0.15% contains 0.15% azelastine hydrochloride in an isotonic aqueous solution containing sorbitol, sucralose, hypromellose, sodium citrate, edetate disodium, benzalkonium chloride (125 mcg/mL), and purified water (pH 6.4).

After priming [ see Dosage and Administration (2.3) ], each metered spray delivers a 0.137 mL mean volume containing 205.5 mcg of azelastine hydrochloride (equivalent to 187.6 mcg of azelastine base).

The 30-mL (net weight 30 gm of solution) bottle provides 200 metered sprays.

Azelastine Hydrochloride Structural Formula

CLINICAL STUDIES

14 14.1 Seasonal Allergic Rhinitis ASTEPRO 0.1% The efficacy and safety of ASTEPRO 0.1% was evaluated in a 2-week, randomized, multicenter, double-blind, placebo-controlled clinical trial including 834 adult and adolescent patients 12 years of age and older with symptoms of seasonal allergic rhinitis.

The population was 12 to 83 years of age (60% female, 40% male; 69% white, 16% black, 12% Hispanic, 2% Asian, 1% other).

Patients were randomized to one of six treatment groups: 1 spray per nostril of either ASTEPRO 0.1%, Astelin (azelastine hydrochloride) Nasal Spray or vehicle placebo twice daily; or 2 sprays per nostril of ASTEPRO 0.1%, Astelin or vehicle placebo twice daily.

Assessment of efficacy was based on the 12-hour reflective total nasal symptom score (rTNSS) assessed daily in the morning and evening, in addition to the instantaneous total nasal symptom score (iTNSS) and other supportive secondary efficacy variables.

TNSS is calculated as the sum of the patients’ scoring of the four individual nasal symptoms (rhinorrhea, nasal congestion, sneezing, and nasal itching) on a 0 to 3 categorical severity scale (0 = absent, 1 = mild, 2 = moderate, 3 = severe).

The rTNSS required patients to record symptom severity over the previous 12 hours.

For the primary efficacy endpoint, the mean change from baseline rTNSS, morning (AM) and evening (PM) rTNSS scores were summed for each day (maximum score of 24) and then averaged over the 2 weeks.

The iTNSS, recorded immediately prior to the next dose, were assessed as an indication of whether the effect was maintained over the dosing interval.

In this trial, ASTEPRO 0.1% two sprays twice a day demonstrated a greater decrease in rTNSS and iTNSS than placebo and the difference was statistically significant.

The trial results are presented in Table 4 (Trial 1).

The efficacy of ASTEPRO 0.1% one spray per nostril twice daily for seasonal allergic rhinitis is supported by two, 2-week, placebo-controlled clinical trials with Astelin (azelastine hydrochloride) Nasal Spray in 413 patients with seasonal allergic rhinitis.

In these trials, efficacy was assessed using the TNSS (described above).

Astelin demonstrated a greater decrease from baseline in the summed AM and PM rTNSS compared with placebo and the difference was statistically significant.

The efficacy of ASTEPRO 0.1% and ASTEPRO 0.15% in children 6 months to 5 years of age with allergic rhinitis was explored in a 4 week, randomized, open-label safety trial in 191 patients.

While the primary objective was to determine the safety of ASTEPRO in this age group, the study included an exploratory efficacy assessment of daily overall allergy symptom scores.

Efficacy in children 6 months to 5 years of age was supported by a numerical decrease in the overall allergy symptom score in both treatment groups.

There was no statistically significant difference between the two treatment groups.

ASTEPRO 0.15% The efficacy and safety of ASTEPRO 0.15% in seasonal allergic rhinitis was evaluated in five randomized, multicenter, double-blind, placebo-controlled clinical trials in 2499 adult and adolescent patients 12 years and older with symptoms of seasonal allergic rhinitis (Trials 2, 3, 4, 5, and 6).

The population of the trials was 12 to 83 years of age (64% female, 36% male, 81% white, 12% black, <2% Asian, 5% other; 23% Hispanic, 77% non-Hispanic).

Assessment of efficacy was based on the rTNSS, iTNSS as described above, and other supportive secondary efficacy variables.

The primary efficacy endpoint was the mean change from baseline in rTNSS over 2 weeks.

Two 2-week seasonal allergic rhinitis trials evaluated the efficacy of ASTEPRO Nasal Spray 0.15% dosed at 2 sprays twice daily.

The first trial (Trial 2) compared the efficacy of ASTEPRO 0.15% and Astelin (azelastine hydrochloride) Nasal Spray to vehicle placebo.

The other trial (Trial 3) compared the efficacy of ASTEPRO 0.15% and ASTEPRO 0.1% to vehicle placebo.

In these two trials, ASTEPRO 0.15% demonstrated greater decreases in rTNSS than placebo and the differences were statistically significant (Table 4).

Three 2-week seasonal allergic rhinitis trials evaluated the efficacy of ASTEPRO 0.15% dosed at 2 sprays once daily compared to the vehicle placebo.

Trial 4 demonstrated a greater decrease in rTNSS than placebo and the difference was statistically significant (Table 4).

Trial 5 and Trial 6 were conducted in patients with Texas mountain cedar allergy.

In Trial 5 and Trial 6, ASTEPRO 0.15% demonstrated a greater decrease in rTNSS than placebo and the differences were statistically significant (Trials 5 and 6; Table 4).

Instantaneous TNSS results for the once daily dosing regimen of ASTEPRO 0.15% are shown in Table 5.

In Trials 5 and 6, ASTEPRO 0.15% demonstrated a greater decrease in iTNSS than placebo and the differences were statistically significant.

Table 4.

Mean Change from Baseline in Reflective TNSS over 2 Weeks * in Adults and Children ≥ 12 years with Seasonal Allergic Rhinitis *Sum of AM and PM rTNSS for each day (Maximum score=24) and averaged over the 14 day treatment period Treatment (sprays per nostril) n Baseline LS Mean Change from Baseline Difference From Placebo LS Mean 95% CI P value Trial 1 Two sprays twice daily ASTEPRO 0.1% 146 18.0 -5.0 -2.2 -3.2,-1.2 <0.001 Astelin Nasal Spray 137 18.2 -4.2 -1.4 -2.4,-0.4 0.01 Vehicle Placebo 138 18.2 -2.8 One spray twice daily ASTEPRO 0.1% 139 18.2 -4.2 -0.7 -1.7, 0.3 0.18 Astelin Nasal Spray 137 18.1 -4.0 -0.4 -1.5, 0.6 0.41 Vehicle Placebo 137 18.0 -3.5 Trial 2 Two sprays twice daily ASTEPRO 0.15% 153 18.2 -4.3 -1.2 -2.1, -0.3 0.01 Astelin Nasal Spray 153 17.9 -3.9 -0.9 -1.8, 0.1 0.07 Vehicle Placebo 153 18.1 -3.0 Trial 3 Two sprays twice daily ASTEPRO 0.15% 177 17.7 -5.1 -3.0 -3.9, -2.1 <0.001 ASTEPRO 0.1% 169 18.2 -4.2 -2.1 -3.0, -1.2 <0.001 Vehicle Placebo 177 17.7 -2.1 Trial 4 Two sprays once daily ASTEPRO 0.15% 238 17.4 -3.4 -1.0 -1.7, -0.3 0.008 Vehicle Placebo 242 17.4 -2.4 Trial 5 Two sprays once daily ASTEPRO 0.15% 266 18.5 -3.3 -1.4 -2.1, -0.8 <0.001 Vehicle Placebo 266 18.0 -1.9 Trial 6 Two sprays once daily ASTEPRO 0.15% 251 18.5 -3.4 -1.4 -2.1, -0.7 <0.001 Vehicle Placebo 254 18.8 -2.0 Table 5.

Mean Change from Baseline AM Instantaneous TNS over 2 Weeks* in Adults and Children ≥ 12 years with Seasonal Allergic Rhinitis *AM iTNSS for each day (Maximum score=12) and averaged over the 14 day treatment period Treatment (sprays per nostril once daily) n Baseline LS Mean Change from Baseline Difference From Placebo LS Mean 95% CI P value Trial 4 Two sprays once daily ASTEPRO 0.15% 238 8.1 -1.3 -0.2 -0.6, 0.1 0.15 Vehicle Placebo 242 8.3 -1.1 Trial 5 Two sprays once daily ASTEPRO 0.15% 266 8.7 -1.4 -0.7 -1.0, -0.4 <0.001 Vehicle Placebo 266 8.3 -0.7 Trial 6 Two sprays once daily ASTEPRO 0.15% 251 8.9 -1.4 -0.6 -0.9, -0.3 <0.001 Vehicle Placebo 254 8.9 -0.8 ASTEPRO 0.15% at a dose of 1 spray twice daily was not studied.

The ASTEPRO 0.15% 1 spray twice daily dosing regimen is supported by previous findings of efficacy for Astelin (azelastine hydrochloride) Nasal Spray and a favorable comparison of ASTEPRO 0.15% to Astelin Nasal Spray and ASTEPRO 0.1% (Table 4).

The efficacy and safety of ASTEPRO 0.1% and 0.15% in children 6 to 11 years of age with seasonal allergic rhinitis was evaluated in a clinical study that enrolled pediatric patients with perennial allergic rhinitis, with or without concomitant seasonal allergic rhinitis (described below in Section 14.2 ).

14.2 Perennial Allergic Rhinitis ASTEPRO 0.1% and ASTEPRO 0.15% The efficacy and safety of ASTEPRO 0.15% in perennial allergic rhinitis was evaluated in one randomized, multicenter, double-blind, placebo-controlled clinical trial in 578 adult and adolescent patients 12 years and older with symptoms of perennial allergic rhinitis.

The population of the trial was 12 to 84 years of age (68% female, 32% male; 85% white, 11% black, 1% Asian, 3% other; 17% Hispanic, 83% non-Hispanic).

Assessment of efficacy was based on the 12-hour reflective total nasal symptom score (rTNSS) assessed daily in the morning and evening, the instantaneous total nasal symptom score (iTNSS), and other supportive secondary efficacy variables.

The primary efficacy endpoint was the mean change from baseline rTNSS over 4 weeks.

The one 4-week perennial allergic rhinitis trial evaluated the efficacy of ASTEPRO 0.15%, ASTEPRO 0.1%, and vehicle placebo dosed at 2 sprays per nostril twice daily.

In this trial, ASTEPRO 0.15% demonstrated a greater decrease in rTNSS than placebo and the difference was statistically significant (Table 6).

Table 6.

Mean Change from Baseline in Reflective TNSS over 4 Weeks* in Adults and Children ≥ 12 years with Perennial Allergic Rhinitis *Sum of AM and PM rTNSS for each day (Maximum score=24) and averaged over the 28 day treatment period Treatment (sprays per nostril twice daily) n Baseline LS Mean Change from Baseline Difference From Placebo LS Mean 95% CI P value Two sprays twice daily ASTEPRO 0.15% 192 15.8 -4.0 -0.9 -1.7, -0.1 0.03 ASTEPRO 0.1% 194 15.5 -3.8 -0.7 -1.5, 0.1 0.08 Vehicle Placebo 192 14.7 -3.1 The efficacy and safety of ASTEPRO 0.1% and ASTEPRO 0.15% in pediatric patients 6 to 11 years of age with perennial allergic rhinitis, with or without concomitant seasonal allergic rhinitis, was evaluated in a randomized, double-blind, placebo-controlled clinical trial in 486 patients.

All patients received one spray per nostril twice daily.

The study population was 58% males and 42% females; 78% white, 13% black, 3% Asian, and 6% other.

Assessment of efficacy was based on the 12-hour reflective total nasal symptom score (rTNSS) assessed daily in the morning and evening.

The primary efficacy endpoint was the mean change from baseline rTNSS over 4 weeks (Table 7).

Both active treatments demonstrated statistically significant decreases in rTNSS compared to placebo.

There was no statistically significant difference between the two active-treatment groups.

There was also no difference in treatment effect between patients with perennial allergic rhinitis only compared to those with perennial allergic rhinitis and concomitant seasonal allergic rhinitis.

Table 7.

Mean Change from Baseline in Reflective TNSS over 4 Weeks* in Children 6 to 11 years with Perennial Allergic Rhinitis *Sum of AM and PM rTNSS for each day (Maximum score=24) and averaged over the 28 day treatment period Treatment (sprays per nostril twice daily) n Baseline LS Mean Change from Baseline Difference From Placebo LS Mean 95% CI P value One spray twice daily ASTEPRO 0.15% 159 16.6 -3.5 -1.0 -1.7, -0.3 0.005 ASTEPRO 0.1% 166 16.4 -3.4 -0.9 -1.6, -0.2 0.015 Vehicle Placebo 161 16.1 -2.5 The efficacy of ASTEPRO 0.1% and ASTEPRO 0.15% in children 6 months to 5 years of age with allergic rhinitis was explored in a clinical study (described above in Section 14.1 ).

HOW SUPPLIED

16 /STORAGE AND HANDLING ASTEPRO (azelastine hydrochloride) 0.1% nasal spray is supplied as a 30-mL package (NDC 0037-0242-30) delivering 200 metered sprays in a high-density polyethylene (HDPE) bottle fitted with a metered-dose spray pump unit.

The spray pump unit consists of a nasal spray pump fitted with a blue safety clip and a blue plastic dust cover.

The net content of the bottle is 30 mL (net weight 30 gm of solution).

Each bottle contains 30 mg (1 mg/mL) of azelastine hydrochloride.

After priming [ see Dosage and Administration (2.3) ], each spray delivers a fine mist containing a mean volume of 0.137 mL solution containing 137 mcg of azelastine hydrochloride.

The correct amount of medication in each spray cannot be assured before the initial priming and after 200 sprays have been used, even though the bottle is not completely empty.

The bottle should be discarded after 200 sprays have been used.

ASTEPRO (azelastine hydrochloride) 0.15% nasal spray is supplied as a 30-mL package (NDC 0037-0243-30) delivering 200 metered sprays in a high-density polyethylene (HDPE) bottle fitted with a metered-dose spray pump unit.

The spray pump unit consists of a nasal spray pump fitted with a blue safety clip and a blue plastic dust cover.

The net content of the bottle is 30 mL (net weight 30 gm of solution).

The 30-mL bottle contains 45 mg (1.5 mg/mL) of azelastine hydrochloride.

After priming [ see Dosage and Administration (2.3) ], each spray delivers a fine mist containing a mean volume of 0.137 mL solution containing 205.5 mcg of azelastine hydrochloride.

The correct amount of medication in each spray cannot be assured before the initial priming and after 200 sprays for the 30-mL bottle have been used, even though the bottle is not completely empty.

The bottle should be discarded after 200 sprays have been used.

ASTEPRO should not be used after the expiration date “EXP” printed on the medicine label and carton.

Storage: Store at controlled room temperature 20° to 25°C (68° to 77°F).

Protect from freezing.

GERIATRIC USE

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

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

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

DOSAGE FORMS AND STRENGTHS

3 ASTEPRO is a nasal spray solution available in two dosage strengths: • Each spray of ASTEPRO 0.1% delivers a volume of 0.137 mL solution containing 137 mcg of azelastine hydrochloride.

• Each spray of ASTEPRO 0.15% delivers a volume of 0.137 mL solution containing 205.5 mcg of azelastine hydrochloride.

Nasal spray solution available in two dosage strengths: • ASTEPRO Nasal Spray 0.1%: 137 mcg of azelastine hydrochloride in each 0.137 mL spray ( 3 ).

• ASTEPRO Nasal Spray 0.15%: 205.5 mcg of azelastine hydrochloride in each 0.137 mL spray ( 3 ).

MECHANISM OF ACTION

12.1 Mechanism of Action Azelastine hydrochloride, a phthalazinone derivative, exhibits histamine H 1 -receptor antagonist activity in isolated tissues, animal models, and humans.

ASTEPRO is administered as a racemic mixture with no difference in pharmacologic activity noted between the enantiomers in in vitro studies.

The major metabolite, desmethylazelastine, also possesses H 1 -receptor antagonist activity.

INDICATIONS AND USAGE

1 ASTEPRO is an H 1 -receptor antagonist indicated for the relief of the symptoms of: • Seasonal allergic rhinitis in patients 2 years of age and older.

( 1.1 ) • Perennial allergic rhinitis in patients 6 months of age and older.

( 1.1 ) 1.1 Allergic Rhinitis ASTEPRO Nasal Spray is indicated for the relief of the symptoms of seasonal allergic rhinitis in patients 2 years of age and older and perennial allergic rhinitis in patients 6 months of age and older.

PEDIATRIC USE

8.4 Pediatric Use The safety and effectiveness of ASTEPRO have been established for seasonal allergic rhinitis in pediatric patients 2 to 17 years of age and perennial allergic rhinitis in pediatric patients 6 months of age to 17 years of age [ see Clinical Studies (14) ].

The safety and effectiveness of ASTEPRO in pediatric patients below 6 months of age have not been established.

PREGNANCY

8.1 Pregnancy Risk Summary Limited data from postmarketing experience over decades of use with ASTEPRO in pregnant women have not identified any drug associated risks of miscarriage, birth defects, or other adverse maternal or fetal outcomes.

In animal reproduction studies, there was no evidence of fetal harm at oral doses approximately 4 times the clinical daily dose.

Oral administration of azelastine hydrochloride to pregnant mice, rats, and rabbits, during the period of organogenesis, produced developmental toxicity that included structural abnormalities, decreased embryo-fetal survival, and decreased fetal body weights at doses 180 times and higher than the maximum recommended human daily intranasal dose (MRHDID) of 1.644 mg.

However, the relevance of these findings in animals to pregnant women was considered questionable based upon the high animal to human dose multiple.

The estimated background risk of major birth defects and miscarriage for the indicated populations is unknown.

All pregnancies have a background risk of birth defect, loss, or other adverse outcomes.

In the U.S.

general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2-4% and 15-20%, respectively.

Data Animal Data In an embryo-fetal development study in mice dosed during the period of organogenesis, azelastine hydrochloride caused embryo-fetal death, structural abnormalities (cleft palate; short or absent tail; fused, absent or branched ribs), delayed ossification, and decreased fetal weight at approximately 200 times the maximum recommended human daily intranasal dose (MRHDID) in adults (on a mg/m 2 basis at a maternal oral dose of 68.6 mg/kg/day), which also caused maternal toxicity as evidenced by decreased maternal body weight.

Neither fetal nor maternal effects occurred in mice at approximately 9 times the MRHDID in adults (on a mg/m 2 basis at a maternal oral dose of 3 mg/kg/day).

In an embryo-fetal development study in pregnant rats dosed during the period of organogenesis from gestation days 7 to 17, azelastine hydrochloride caused structural abnormalities (oligo-and brachydactylia), delayed ossification, and skeletal variations, in the absence of maternal toxicity, at approximately 180 times the MRHDID in adults (on a mg/m 2 basis at a maternal oral dose of 30 mg/kg/day).

Azelastine hydrochloride caused embryo-fetal death and decreased fetal weight and severe maternal toxicity at approximately 410 times the MRHDID (on a mg/m 2 basis at a maternal oral dose of 68.6 mg/kg/day).

Neither fetal nor maternal effects occurred at approximately 10 times the MRHDID (on a mg/m 2 basis at a maternal oral dose of 2 mg/kg/day).

In an embryo-fetal development study in pregnant rabbits dosed during the period of organogenesis from gestation days 6 to 18, azelastine hydrochloride caused abortion, delayed ossification and decreased fetal weight and severe maternal toxicity at approximately 360 times the MRHDID in adults (on a mg/m 2 basis at a maternal oral dose of 30 mg/kg/day).

Neither fetal nor maternal effects occurred at approximately 4 times the MRHDID (on a mg/m 2 basis at a maternal oral dose of 0.3 mg/kg/day).

In a prenatal and postnatal development study in pregnant rats dosed from late in the gestation period and through the lactation period from gestation day 17 through lactation day 21, azelastine hydrochloride produced no adverse developmental effects on pups at maternal doses up to approximately 180 times the MRHDID (on mg/m 2 basis at a maternal dose of 30 mg/kg/day).

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS • Somnolence: Avoid engaging in hazardous occupations requiring complete mental alertness such as driving or operating machinery when taking ASTEPRO ( 5.1 ) • Avoid concurrent use of alcohol or other central nervous system (CNS) depressants with ASTEPRO because further decreased alertness and impairment of CNS performance may occur.

( 5.1 ) 5.1 Activities Requiring Mental Alertness In clinical trials, the occurrence of somnolence has been reported in some patients taking ASTEPRO [ see Adverse Reactions (6.1) ].

Patients should be cautioned against engaging in hazardous occupations requiring complete mental alertness and motor coordination such as operating machinery or driving a motor vehicle after administration of ASTEPRO.

Concurrent use of ASTEPRO with alcohol or other central nervous system depressants should be avoided because additional reductions in alertness and additional impairment of central nervous system performance may occur [ see Drug Interactions (7.1) ].

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION See FDA-approved patient labeling ( Patient Information and Instructions for Use ).

Activities Requiring Mental Alertness Somnolence has been reported in some patients taking ASTEPRO.

Caution patients against engaging in hazardous occupations requiring complete mental alertness and motor coordination such as driving or operating machinery after administration of ASTEPRO [ see Warnings and Precautions (5.1) ].

Concurrent Use of Alcohol and other Central Nervous System Depressants Avoid concurrent use of ASTEPRO with alcohol or other central nervous system depressants because additional reductions in alertness and additional impairment of central nervous system performance may occur [ see Warnings and Precautions (5.1) ].

Common Adverse Reactions Inform patients that the treatment with ASTEPRO may lead to adverse reactions, most common of which include pyrexia, dysgeusia, nasal discomfort, epistaxis, headache, sneezing, fatigue, somnolence, upper respiratory infection, cough, rhinalgia, vomiting, otitis media, contact dermatitis, and oropharyngeal pain.

[ see Adverse Reactions (6.1) ].

Priming Instruct patients to prime the pump before initial use and when ASTEPRO has not been used for 3 or more days [ see Dosage and Administration (2.3) ].

Keep Spray Out of Eyes Instruct patients to avoid spraying ASTEPRO into their eyes.

Keep Out of Children’s Reach Instruct patients to keep ASTEPRO out of the reach of children.

If a child accidentally ingests ASTEPRO, seek medical help or call a poison control center immediately.

Manufactured for: Meda Pharmaceuticals Inc.

Somerset, New Jersey 08873-4120 ©2018 Meda Pharmaceuticals Inc.

ASTEPRO and MEDA PHARMACEUTICALS are registered trademarks of Meda Pharmaceuticals Inc.

or a related entity.

U.S.

Patents 8,071,073; 8,518,919

DOSAGE AND ADMINISTRATION

2 • For intranasal use only ( 2.3 ).

• Seasonal allergic rhinitis : ▪ 2 to 5 years : ASTEPRO 0.1%: 1 spray per nostril twice daily ( 2.1 ) ▪ 6 to 11 years : ASTEPRO 0.1% or ASTEPRO 0.15%: 1 spray per nostril twice daily ( 2.1 ) ▪ Adults and adolescents 12 years of age and older : o ASTEPRO 0.1% or ASTEPRO 0.15%: 1 or 2 sprays per nostril twice daily ( 2.1 ), or o ASTEPRO 0.15%: 2 sprays per nostril once daily ( 2.1 ) • Perennial allergic rhinitis : ▪ 6 months to 5 years : ASTEPRO 0.1%: 1 spray per nostril twice daily ( 2.2 ) ▪ 6 to 11 years : ASTEPRO 0.1% or ASTEPRO 0.15%: 1 spray per nostril twice daily ( 2.2 ) ▪ Adults and adolescents 12 years of age and older : ASTEPRO 0.15%: 2 sprays per nostril twice daily ( 2.2 ) • Prime ASTEPRO before initial use and when it has not been used for 3 or more days.

( 2.3 ) 2.1 Seasonal Allergic Rhinitis Children 2 to 5 years of age: ASTEPRO 0.1%, 1 spray per nostril twice daily.

Children 6 to 11 years of age: ASTEPRO 0.1% or ASTEPRO 0.15%, 1 spray per nostril twice daily.

Adults and adolescents 12 years of age and older: ASTEPRO 0.1% or ASTEPRO 0.15%, 1 or 2 sprays per nostril twice daily.

ASTEPRO 0.15% may also be administered as 2 sprays per nostril once daily.

2.2 Perennial Allergic Rhinitis Children 6 months to 5 years of age: ASTEPRO 0.1%, 1 spray per nostril twice daily.

Children 6 to 11 years of age: ASTEPRO 0.1% or ASTEPRO 0.15%, 1 spray per nostril twice daily.

Adults and adolescents 12 years of age and older: ASTEPRO 0.15%, 2 sprays per nostril twice daily.

2.3 Important Administration Instructions Administer ASTEPRO by the intranasal route only.

Priming : Prime ASTEPRO before initial use by releasing 6 sprays or until a fine mist appears.

When ASTEPRO has not been used for 3 or more days, reprime with 2 sprays or until a fine mist appears.

Avoid spraying ASTEPRO into the eyes.

homatropine methylbromide 1.5 MG / hydrocodone bitartrate 5 MG per 5 ML Syrup

WARNINGS

May be habit forming.

Hydrocodone can produce drug dependence of the morphine type and, therefore, has the potential for being abused.

Psychic dependence, physical dependence and tolerance may develop upon repeated administration of hydrocodone bitartrate and homatropine methylbromide and it should be prescribed and administered with the same degree of caution appropriate to the use of other narcotic drugs (see DRUG ABUSE AND DEPENDENCE ).

Respiratory Depression Hydrocodone bitartrate and homatropine methylbromide produces dose-related respiratory depression by directly acting on brain stem respiratory centers.

If respiratory depression occurs, it may be antagonized by the use of naloxone hydrochloride and other supportive measures when indicated.

Head Injury and Increased Intracranial Pressure The respiratory depression properties of narcotics and their capacity to elevate cerebrospinal fluid pressure may be markedly exaggerated in the presence of head injury, other intracranial lesions or a pre-existing increase in intracranial pressure.

Furthermore, narcotics produce adverse reactions which may obscure the clinical course of patients with head injuries.

Acute Abdominal Conditions The administration of hydrocodone bitartrate and homatropine methylbromide or other narcotics may obscure the diagnosis or clinical course of patients with acute abdominal conditions.

Pediatric Use In young pediatric patients, as well as adults, the respiratory center is sensitive to the depressant action of narcotic cough suppressants in a dose-dependent manner.

Benefit to risk ratio should be carefully considered especially in the pediatric population with respiratory embarrassment (e.g., croup).

DRUG INTERACTIONS

Drug Interactions Patients receiving narcotics, antihistamines, antipsychotics, antianxiety agents or other CNS depressants (including alcohol) concomitantly with hydrocodone bitartrate and homatropine methylbromide may exhibit an additive CNS depression.

When combined therapy is contemplated, the dose of one or both agents should be reduced.

The use of MAO inhibitors or tricyclic antidepressants with hydrocodone preparations may increase the effect of either the antidepressant or hydrocodone.

OVERDOSAGE

Signs and Symptoms Serious overdosage with hydrocodone is characterized by respiratory depression (a decrease in respiratory rate and/or tidal volume, Cheyne-Stokes respiration, cyanosis), extreme somnolence progressing to stupor or coma, skeletal muscle flaccidity, cold and clammy skin, and sometimes bradycardia and hypotension.

In severe overdosage, apnea, circulatory collapse, cardiac arrest and death may occur.

The ingestion of very large amounts of hydrocodone bitartrate and homatropine methylbromide may, in addition, result in acute homatropine intoxication.

Treatment Primary attention should be given to the reestablishment of adequate respiratory exchange through provision of a patent airway and the institution of assisted or controlled ventilation.

The narcotic antagonist naloxone hydrochloride is a specific antidote for respiratory depression which may result from overdosage or unusual sensitivity to narcotics including hydrocodone.

Therefore, an appropriate dose of naloxone hydrochloride should be administered, preferably by the intravenous route, simultaneously with efforts at respiratory resuscitation.

For further information, see full prescribing information for naloxone hydrochloride.

An antagonist should not be administered in the absence of clinically significant respiratory depression.

Oxygen, intravenous fluids, vasopressors and other supportive measures should be employed as indicated.

Gastric emptying may be useful in removing unabsorbed drug.

DESCRIPTION

This product contains hydrocodone (dihydrocodeinone) bitartrate, a semisynthetic centrally-acting narcotic antitussive.

Homatropine methylbromide is included in a subtherapeutic amount to discourage deliberate overdosage.

Each teaspoonful (5 mL) for oral administration contains: Hydrocodone Bitartrate, USP 5 mg WARNING: May be habit forming.

Homatropine Methylbromide, USP 1.5 mg Hydrocodone Bitartrate and Homatropine Methylbromide Syrup also contains: Caramel color, cherry flavor, citric acid, FD&C Red #40, methylparaben, propylparaben, purified water, sorbitol solution and sucrose syrup.

Citric acid and/or sodium citrate may be added to adjust pH.

The hydrocodone component is a 4,5α- epoxy-3-methoxy-17-methylmorphinan- 6-one tartrate (1:1) hydrate (2:5), a fine white crystal or crystalline powder, which is derived from the opium alkaloid, thebaine, has a molecular weight of (494.50) and may be represented by the following structural formula: Homatropine methylbromide is 8- Azoniabicyclo [3.2.1]octane,3-[(hydroxyphenylacetyl) oxy]-8,8-dimethyl-,bromide, endo-; a white crystal or fine white crystalline powder, with a molecular weight of (370.29).

Structural Formula for hydrocodone bitartrate Structural Formula for homatropine methylbromide

HOW SUPPLIED

Hydrocodone Bitartrate and Homatropine Methylbromide Syrup is available as a clear red colored, cherry flavored syrup in: Bottles of 16 fl.

oz.

(one pint) ……

NDC 54868-5168-0 Store at 20°-25°C (68°-77°F) [See USP Controlled Room Temperature].

Dispense in a tight, light-resistant container, as defined in the USP.

Oral prescription where permitted by state law.

Manufactured By: Hi-Tech Pharmacal Co., Inc.

Amityville, NY 11701 Rev.

043:00 3/09 MG #21271 Additional barcode label applied by: Physicians Total Care, Inc.

Tulsa, Oklahoma 74146

INDICATIONS AND USAGE

Hydrocodone bitartrate and homatropine methylbromide syrup is indicated for the symptomatic relief of cough.

PEDIATRIC USE

Pediatric Use Safety and effectiveness of hydrocodone bitartrate and homatropine methylbromide in pediatric patients under six have not been established.

PREGNANCY

Pregnancy Teratogenic Effects Pregnancy Category C: Animal reproduction studies have not been conducted with hydrocodone bitartrate and homatropine methylbromide.

It is also not known whether hydrocodone bitartrate and homatropine methylbromide can cause fetal harm when administered to a pregnant woman or can affect reproduction capacity.

Hydrocodone bitartrate and homatropine methylbromide should be given to a pregnant woman only if clearly needed.

Nonteratogenic effects Babies born to mothers who have been taking opioids regularly prior to delivery will be physically dependent.

The withdrawal signs include irritability and excessive crying, tremors, hyperactive reflexes, increased respiratory rate, increased stools, sneezing, yawning, vomiting and fever.

The intensity of the syndrome does not always correlate with the duration of maternal opioid use or dose.

NUSRING MOTHERS

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

Because many drugs are excreted in human milk and because of the potential for serious adverse reactions in nursing infants from hydrocodone bitartrate and homatropine methylbromide, 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.

INFORMATION FOR PATIENTS

Information for patients Hydrocodone may impair the mental and/or physical abilities required for the performance of potentially hazardous tasks such as driving a car or operating machinery.

The patient using hydrocodone bitartrate and homatropine methylbromide should be cautioned accordingly.

DOSAGE AND ADMINISTRATION

Adults One (1) teaspoonful (5 mL) of the syrup every 4 to 6 hours as needed; do not exceed six (6) teaspoonfuls in 24 hours.

Children 6 to 12 Years of Age One-half (1/2) teaspoonful (2.5 mL) of the syrup every 4 to 6 hours as needed; do not exceed three (3) teaspoonfuls in 24 hours.

formoterol fumarate 0.005 MG / mometasone furoate 0.1 MG per ACTUAT Metered Dose Inhaler, 60 ACTUAT

DRUG INTERACTIONS

7 In clinical trials, concurrent administration of DULERA and other drugs, such as short-acting beta 2 -agonist and intranasal corticosteroids have not resulted in an increased frequency of adverse drug reactions.

No formal drug interaction studies have been performed with DULERA.

The drug interactions of the combination are expected to reflect those of the individual components.

Strong cytochrome P450 3A4 inhibitors (e.g., ritonavir): Use with caution.

May cause increased systemic corticosteroid effects.

( 7.1 ) Adrenergic agents: Use with caution.

Additional adrenergic drugs may potentiate sympathetic effects.

( 7.2 ) Xanthine derivatives and diuretics: Use with caution.

May potentiate ECG changes and/or hypokalemia.

( 7.3 , 7.4 ) MAO inhibitors, tricyclic antidepressants, macrolides, and drugs that prolong QTc interval: Use with extreme caution.

May potentiate effect on the cardiovascular system.

( 7.5 ) Beta-blockers: Use with caution and only when medically necessary.

May decrease effectiveness and produce severe bronchospasm.

( 7.6 ) Halogenated hydrocarbons: There is an elevated risk of arrhythmias in patients receiving concomitant anesthesia with halogenated hydrocarbons.

( 7.7 ) 7.1 Inhibitors of Cytochrome P450 3A4 The main route of metabolism of corticosteroids, including mometasone furoate, a component of DULERA, is via cytochrome P450 (CYP) isoenzyme 3A4 (CYP3A4).

After oral administration of ketoconazole, a strong inhibitor of CYP3A4, the mean plasma concentration of orally inhaled mometasone furoate increased.

Concomitant administration of CYP3A4 inhibitors may inhibit the metabolism of, and increase the systemic exposure to, mometasone furoate and potentially increase the risk for systemic corticosteroid side effects.

Caution should be exercised when considering the coadministration of DULERA with long-term ketoconazole and other known strong CYP3A4 inhibitors (e.g., ritonavir, cobicistat-containing products, atazanavir, clarithromycin, indinavir, itraconazole, nefazodone, nelfinavir, saquinavir, telithromycin) [see Warnings and Precautions (5.8) and Clinical Pharmacology (12.3) ] .

Consider the benefit of coadministration versus the potential risk of systemic corticosteroid effects, in which case patients should be monitored for systemic corticosteroid side effects.

7.2 Adrenergic Agents If additional adrenergic drugs are to be administered by any route, they should be used with caution because the pharmacologically predictable sympathetic effects of formoterol, a component of DULERA, may be potentiated.

7.3 Xanthine Derivatives Concomitant treatment with xanthine derivatives may potentiate any hypokalemic effect of formoterol, a component of DULERA.

7.4 Diuretics Concomitant treatment with diuretics may potentiate the possible hypokalemic effect of adrenergic agonists.

The ECG changes and/or hypokalemia that may result from the administration of non-potassium-sparing diuretics (such as loop or thiazide diuretics) can be acutely worsened by beta-agonists, especially when the recommended dose of the beta-agonist is exceeded.

Although the clinical significance of these effects is not known, caution is advised in the coadministration of DULERA with non-potassium-sparing diuretics.

7.5 Monoamine Oxidase Inhibitors, Tricyclic Antidepressants, and Drugs Known to Prolong the QTc Interval DULERA should be administered with caution to patients being treated with monoamine oxidase inhibitors, tricyclic antidepressants, macrolides, or drugs known to prolong the QTc interval or within 2 weeks of discontinuation of such agents, because the action of formoterol, a component of DULERA, on the cardiovascular system may be potentiated by these agents.

Drugs that are known to prolong the QTc interval have an increased risk of ventricular arrhythmias.

7.6 Beta-Adrenergic Receptor Antagonists Beta-adrenergic receptor antagonists (beta-blockers) and formoterol may inhibit the effect of each other when administered concurrently.

Beta-blockers not only block the therapeutic effects of beta 2 -agonists, such as formoterol, a component of DULERA, but may produce severe bronchospasm in patients with asthma.

Therefore, patients with asthma should not normally be treated with beta-blockers.

However, under certain circumstances, e.g., as prophylaxis after myocardial infarction, there may be no acceptable alternatives to the use of beta-blockers in patients with asthma.

In this setting, cardioselective beta-blockers could be considered, although they should be administered with caution.

7.7 Halogenated Hydrocarbons There is an elevated risk of arrhythmias in patients receiving concomitant anesthesia with halogenated hydrocarbons.

OVERDOSAGE

10 10.1 Signs and Symptoms DULERA: DULERA contains both mometasone furoate and formoterol fumarate; therefore, the risks associated with overdosage for the individual components described below apply to DULERA.

Mometasone Furoate: Chronic overdosage may result in signs/symptoms of hypercorticism [see Warnings and Precautions (5.7) ] .

Single oral doses up to 8000 mcg of mometasone furoate have been studied on adult subjects with no adverse reactions reported.

Formoterol Fumarate: The expected signs and symptoms with overdosage of formoterol are those of excessive beta-adrenergic stimulation and/or occurrence or exaggeration of any of the following signs and symptoms: angina, hypertension or hypotension, tachycardia, with rates up to 200 beats/min., arrhythmias, nervousness, headache, tremor, seizures, muscle cramps, dry mouth, palpitation, nausea, dizziness, fatigue, malaise, hypokalemia, hyperglycemia, and insomnia.

Metabolic acidosis may also occur.

Cardiac arrest and even death may be associated with an overdose of formoterol.

The minimum acute lethal inhalation dose of formoterol fumarate in rats is 156 mg/kg (approximately 63,000 times the MRHD on a mcg/m 2 basis).

The median lethal oral doses in Chinese hamsters, rats, and mice provide even higher multiples of the MRHD.

10.2 Treatment DULERA: Treatment of overdosage consists of discontinuation of DULERA together with institution of appropriate symptomatic and/or supportive therapy.

The judicious use of a cardioselective beta-receptor blocker may be considered, bearing in mind that such medication can produce bronchospasm.

There is insufficient evidence to determine if dialysis is beneficial for overdosage of DULERA.

Cardiac monitoring is recommended in cases of overdosage.

DESCRIPTION

11 DULERA 50 mcg/5 mcg, DULERA 100 mcg/5 mcg, and DULERA 200 mcg/5 mcg are combinations of mometasone furoate and formoterol fumarate dihydrate for oral inhalation only.

One active component of DULERA is mometasone furoate, a corticosteroid having the chemical name 9,21-dichloro-11(Beta),17-dihydroxy-16 (alpha)-methylpregna-1,4-diene-3,20-dione 17-(2-furoate) with the following chemical structure: Mometasone furoate is a white powder with an empirical formula of C 27 H 30 Cl 2 O 6 , and molecular weight 521.44.

It is practically insoluble in water; slightly soluble in methanol, ethanol, and isopropanol; soluble in acetone.

One active component of DULERA is formoterol fumarate dihydrate, a racemate.

Formoterol fumarate dihydrate is a selective beta 2 -adrenergic bronchodilator having the chemical name of (±)-2-hydroxy-5-[(1RS)-1-hydroxy-2-[[(1RS)-2-(4-methoxyphenyl)-1-methylethyl]-amino]ethyl]formanilide fumarate dihydrate with the following chemical structure: Formoterol fumarate dihydrate has a molecular weight of 840.9, and its empirical formula is (C 19 H 24 N 2 O 4 ) 2 •C 4 H 4 O 4 •2H 2 O.

Formoterol fumarate dihydrate is a white to yellowish powder, which is freely soluble in glacial acetic acid, soluble in methanol, sparingly soluble in ethanol and isopropanol, slightly soluble in water, and practically insoluble in acetone, ethyl acetate, and diethyl ether.

DULERA 50 mcg/5 mcg, 100 mcg/5 mcg, and 200 mcg/5 mcg are each formulated as a hydrofluoroalkane (HFA-227; 1, 1, 1, 2, 3, 3, 3-heptafluoropropane) propelled pressurized metered dose inhaler containing sufficient amount of drug for 60 or 120 inhalations [see How Supplied/Storage and Handling (16) ] .

After priming, each actuation of the inhaler delivers 60, 115, or 225 mcg of mometasone furoate and 5.5 mcg of formoterol fumarate dihydrate in 69.6 mg of suspension from the valve and delivers 50, 100, or 200 mcg of mometasone furoate and 5 mcg of formoterol fumarate dihydrate from the actuator.

The actual amount of drug delivered to the lung may depend on patient factors, such as the coordination between actuation of the device and inspiration through the delivery system.

DULERA also contains anhydrous alcohol as a cosolvent and oleic acid as a surfactant.

DULERA should be primed before using for the first time by releasing 4 test sprays into the air, away from the face, shaking well before each spray.

In cases where the inhaler has not been used for more than 5 days, prime the inhaler again by releasing 4 test sprays into the air, away from the face, shaking well before each spray.

Image of Mometasone Furoate Chemical Structure Image of Formoterol Fumarate Dihydrate Chemical Structure

CLINICAL STUDIES

14 14.1 Asthma Adult and Adolescent Patients Aged 12 Years of Age and Older The safety and efficacy of DULERA were demonstrated in two randomized, double-blind, parallel group, multicenter clinical trials of 12 to 26 weeks in duration involving 1509 patients 12 years of age and older with persistent asthma uncontrolled on medium or high dose inhaled corticosteroids (baseline FEV 1 means of 66% to 73% of predicted normal).

These studies included a 2 to 3-week run-in period with mometasone furoate to establish a certain level of asthma control.

One clinical trial compared DULERA to placebo and the individual components, mometasone furoate and formoterol (Trial 1) and one clinical trial compared two different strengths of DULERA to mometasone furoate alone (Trial 2) .

Trial 1: Clinical Trial with DULERA 100 mcg/5 mcg This 26-week, placebo-controlled trial (NCT00383240) evaluated 781 patients 12 years of age and older comparing DULERA 100 mcg/5 mcg (n=191 patients), mometasone furoate 100 mcg (n=192 patients), formoterol fumarate 5 mcg (n=202 patients) and placebo (n=196 patients); each administered as 2 inhalations twice daily by metered dose inhalation aerosols.

All other maintenance therapies were discontinued.

This study included a 2 to 3-week run-in period with mometasone furoate 100 mcg, 2 inhalations twice daily.

This trial included patients ranging from 12 to 76 years of age, 41% male and 59% female, and 72% Caucasian and 28% non-Caucasian.

Patients had persistent asthma and were not well controlled on medium dose of inhaled corticosteroids prior to randomization.

All treatment groups were balanced with regard to baseline characteristics.

Mean FEV 1 and mean percent predicted FEV 1 were similar among all treatment groups (2.33 L, 73%).

Eight (4%) patients receiving DULERA 100 mcg/5 mcg, 13 (7%) patients receiving mometasone furoate 100 mcg, 47 (23%) patients receiving formoterol fumarate 5 mcg and 46 (23%) patients receiving placebo discontinued the study early due to treatment failure.

FEV 1 AUC (0-12 hr) was assessed as a co-primary efficacy endpoint to evaluate the contribution of the formoterol component to DULERA.

Patients receiving DULERA 100 mcg/5 mcg had significantly higher increases from baseline at Week 12 in mean FEV 1 AUC (0-12 hr) compared to mometasone furoate 100 mcg (the primary treatment comparison) and vs.

placebo (both p<0.001) ( Figure 1 ).

These differences were maintained through Week 26.

Figure 1 shows the change from baseline post-dose serial FEV 1 evaluations in Trial 1.

Figure 1 Trial 1 – DULERA 100 mcg/5 mcg – FEV 1 Serial Evaluations for Observed Cases at Week 12 Change from Baseline by Treatment Clinically judged deteriorations in asthma or reductions in lung function were assessed as another primary endpoint to evaluate the contribution of mometasone furoate 100 mcg to DULERA 100 mcg/5 mcg (primary treatment comparison DULERA vs.

formoterol).

Deteriorations in asthma were defined as any of the following: a 20% decrease in FEV 1 ; a 30% decrease in PEF on two or more consecutive days; emergency treatment, hospitalization, or treatment with systemic corticosteroids or other asthma medications not allowed per protocol.

Fewer patients who received DULERA 100 mcg/5 mcg reported an event compared to patients who received formoterol 5 mcg (p<0.001).

Table 3: Trial 1 – Clinically Judged Deterioration in Asthma or Reduction in Lung Function Includes only the first event day for each patient.

Patients could have experienced more than one event criterion.

DULERA 100 mcg/ 5 mcg Two inhalations, twice daily.

(n=191) Mometasone Furoate 100 mcg (n=192) Formoterol 5 mcg (n=202) Placebo (n=196) Clinically judged deterioration in asthma or reduction in lung function 58 (30%) 65 (34%) 109 (54%) 109 (56%) Decrease in FEV 1 Decrease in absolute FEV 1 below the treatment period stability limit (defined as 80% of the average of the two predose FEV 1 measurements taken 30 minutes and immediately prior to the first dose of randomized trial medication).

18 (9%) 19 (10%) 31 (15%) 41 (21%) Decrease in PEF Decrease in AM or PM peak expiratory flow (PEF) on 2 or more consecutive days below the treatment period stability limit (defined as 70% of the AM or PM PEF obtained over the last 7 days of the run-in period).

37 (19%) 41 (21%) 62 (31%) 61 (31%) Emergency treatment 0 1 (<1%) 4 (2%) 1 (<1%) Hospitalization 1 (<1%) 0 0 0 Treatment with excluded asthma medication Thirty patients received glucocorticosteroids; 1 patient received formoterol via dry powder inhaler in the Formoterol 5 mcg group.

2 (1%) 4 (2%) 17 (8%) 8 (4%) The change in mean trough FEV 1 from baseline to Week 12 was assessed as another endpoint to evaluate the contribution of mometasone furoate 100 mcg to DULERA 100 mcg/5 mcg.

A significantly greater increase in mean trough FEV 1 was observed for DULERA 100 mcg/5 mcg compared to formoterol 5 mcg (the primary treatment comparison) as well as to placebo ( Table 4 ).

Table 4: Trial 1 – Change in Trough FEV 1 from Baseline to Week 12 Treatment Arm N Baseline (L) Change From Baseline at Week 12 (L) Treatment Difference from Placebo (L) P-Value vs.

Placebo P-Value vs.

Formoterol LS means and p-values are from Week 12 estimates of a longitudinal analysis model.

DULERA 100 mcg/5 mcg 167 2.33 0.13 0.18 <0.001 <0.001 Mometasone furoate 100 mcg 175 2.36 0.07 0.12 <0.001 0.058 Formoterol fumarate 5 mcg 141 2.29 0.00 0.05 0.170 Placebo 145 2.30 -0.05 The effect of DULERA 100 mcg/5 mcg, two inhalations twice daily on selected secondary efficacy endpoints, including proportion of nights with nocturnal awakenings (-60% vs.

-15%), change in total rescue medication use (-0.6 vs.

+1.1 puffs/day), change in morning peak flow (+18.1 vs.

-28.4 L/min) and evening peak flow (+10.8 vs.

-32.1 L/min) further supports the efficacy of DULERA 100 mcg/5 mcg compared to placebo.

The subjective impact of asthma on patients’ health-related quality of life was evaluated by the Asthma Quality of Life Questionnaire (AQLQ(S)) (based on a 7-point scale where 1 = maximum impairment and 7 = no impairment).

A change from baseline ≥0.5 points is considered a clinically meaningful improvement.

The mean difference in AQLQ between patients receiving DULERA 100 mcg/5 mcg and placebo was 0.5 [95% CI 0.32, 0.68].

Image of Figure 1 Trial 2: Clinical Trial With DULERA 200 mcg/5 mcg This 12-week double-blind trial (NCT00381485) evaluated 728 patients 12 years of age and older comparing DULERA 200 mcg/5 mcg (n=255 patients) with DULERA 100 mcg/5 mcg (n=233 patients) and mometasone furoate 200 mcg (n=240 patients), each administered as 2 inhalations twice daily by metered dose inhalation aerosols.

All other maintenance therapies were discontinued.

This trial included a 2 to 3-week run-in period with mometasone furoate 200 mcg, 2 inhalations twice daily.

Patients had persistent asthma and were uncontrolled on high dose inhaled corticosteroids prior to study entry.

All treatment groups were balanced with regard to baseline characteristics.

This trial included patients ranging from 12 to 84 years of age, 44% male and 56% female, and 89% Caucasian and 11% non-Caucasian.

Mean FEV 1 and mean percent predicted FEV 1 values were similar among all treatment groups (2.05 L, 66%).

Eleven (5%) patients receiving DULERA 100 mcg/5 mcg, 8 (3%) patients receiving DULERA 200 mcg/5 mcg and 13 (5%) patients receiving mometasone furoate 200 mcg discontinued the trial early due to treatment failure.

The primary efficacy endpoint was the mean change in FEV 1 AUC (0-12 hr) from baseline to Week 12.

Patients receiving DULERA 100 mcg/5 mcg and DULERA 200 mcg/5 mcg had significantly greater increases from baseline at Day 1 in mean FEV 1 AUC (0-12 hr) compared to mometasone furoate 200 mcg.

The difference was maintained over 12 weeks of therapy.

Mean change in trough FEV 1 from baseline to Week 12 was also assessed to evaluate the relative contribution of mometasone furoate to DULERA 100 mcg/5 mcg and DULERA 200 mcg/5 mcg ( Table 5 ).

A greater numerical increase in the mean trough FEV 1 was observed for DULERA 200 mcg/5 mcg compared to DULERA 100 mcg/5 mcg and mometasone furoate 200 mcg.

Table 5: Trial 2 – Change in Trough FEV 1 from Baseline to Week 12 Treatment Arm N Baseline (L) Change from Baseline at Week 12 (L) DULERA 100 mcg/5 mcg 232 2.10 0.14 DULERA 200 mcg/5 mcg 255 2.05 0.19 Mometasone furoate 200 mcg 239 2.07 0.10 Clinically judged deterioration in asthma or reduction in lung function was assessed as an additional endpoint.

Fewer patients who received DULERA 200 mcg/5 mcg or DULERA 100/5 mcg compared to mometasone furoate 200 mcg alone reported an event, defined as in Trial 1 by any of the following: a 20% decrease in FEV 1 ; a 30% decrease in PEF on two or more consecutive days; emergency treatment, hospitalization, or treatment with systemic corticosteroids or other asthma medications not allowed per protocol.

Table 6: Trial 2 – Clinically Judged Deterioration in Asthma or Reduction in Lung Function Includes only the first event day for each patient.

Patients could have experienced more than one event criterion.

DULERA 100 mcg/ 5 mcg Two inhalations, twice daily.

(n=233) DULERA 200 mcg/ 5 mcg (n=255) Mometasone Furoate 200 mcg (n=240) Clinically judged deterioration in asthma or reduction in lung function 29 (12%) 31 (12%) 44 (18%) Decrease in FEV 1 Decrease in absolute FEV 1 below the treatment period stability limit (defined as 80% of the average of the two predose FEV 1 measurements taken 30 minutes and immediately prior to the first dose of randomized trial medication).

23 (10%) 17 (7%) 33 (14%) Decrease in PEF on two consecutive days Decrease in AM or PM peak expiratory flow (PEF) below the treatment period stability limit (defined as 70% of the AM or PM PEF obtained over the last 7 days of the run-in period).

2 (1%) 4 (2%) 3 (1%) Emergency treatment 2 (1%) 1 (<1%) 1 (<1%) Hospitalization 0 1 (<1%) 0 Treatment with excluded asthma medication Twenty four patients received glucocorticosteroids; 1 patient received albuterol in the DULERA 200 mcg / 5 mcg group.

5 (2%) 8 (3%) 12 (5%) Other Studies in Adults In addition to Trial 1 and Trial 2, the safety and efficacy of the individual components, mometasone furoate MDI 100 mcg and 200 mcg (each administered as 2 inhalations by MDI), in comparison to placebo were demonstrated in two other, 12-week, placebo-controlled trials that evaluated the mean change in FEV 1 from baseline as a primary endpoint.

The safety and efficacy of formoterol MDI 5 mcg (administered as 2 inhalations twice daily) alone in comparison to placebo was replicated in another 26-week trial (NCT00383552) that also evaluated the same endpoint with a lower dose of mometasone furoate MDI in combination with formoterol.

Pediatric Patients Aged 5 to Less Than 12 Years The efficacy of DULERA 50 mcg/5 mcg in children aged 5 to less than 12 years was demonstrated in a randomized, active-controlled, multicenter clinical trial (NCT02741271) in which DULERA 50 mcg/5 mcg (administered as two inhalations, twice daily; n=91) was compared with mometasone furoate MDI 50 mcg (administered as two inhalations, twice daily; n=90), in 181 asthma patients aged 5 to less than 12 years.

These participants had been adequately controlled on an ICS/LABA for at least 4 weeks and had no symptoms of asthma worsening during a 2-week run-in on mometasone furoate MDI 50 mcg (administered as two inhalations twice daily).

Primary endpoint results showed that patients receiving DULERA 50 mcg/5 mcg had a statistically significant change from baseline to Week 12 in 60-min AM post-dose % predicted FEV 1 compared to mometasone furoate MDI 50 mcg (5.21, 95% CI: 3.22, 7.20) [Figure 2].

Bronchodilatory improvement with DULERA 50 mcg/5 mcg relative to mometasone furoate MDI 50 mcg was significant from the first assessment at 5 minutes and was sustained through 4 hours post-dose.

These improvements were evident as early as the first dose and were maintained through Week 12.

Figure 2: Change from Baseline AM Post-Dose in % Predicted FEV 1 Through Week 12 MF = mometasone furoate; MF/F = mometasone furoate/formoterol fumarate Population consists of all subjects who received at least one dose of blinded treatment and completed at least one efficacy evaluation.

Treatment Effects were estimated using the Primary Analysis Method [i.e., a cLDA model with control-based multiple imputation, including terms for treatment, time, age strata (5-7 yrs., 8-11 yrs.), treatment by time interaction, and region (US, ex-US)].

Figure 2 Postmarketing Safety and Efficacy Trial with DULERA This 26-week double-blind, randomized control trial evaluated 11,729 patients, 12 years of age and older, who received at least one dose of DULERA (100 mcg/5 mcg or 200 mcg/5 mcg, n=5868) or mometasone furoate monotherapy (100 mcg or 200 mcg, n=5861) each administered as 2 inhalations twice daily by metered dose inhalation aerosols (NCT01471340).

The primary safety objective was to evaluate whether the addition of formoterol to mometasone furoate (DULERA) was non-inferior to mometasone furoate in risk of serious asthma-related events (adjudicated hospitalization, intubation, and death).

A blinded adjudication committee determined whether events were asthma-related.

The study was designed to rule out a pre-defined risk margin of 2.0.

Enrolled patients had a diagnosis of persistent asthma, had been receiving a stable dose of asthma maintenance therapy for at least 4 weeks and had a history of one to four asthma exacerbations requiring hospitalization or systemic corticosteroid use in the previous year.

The assigned dose level of inhaled corticosteroid was based on the patients’ disease severity, considering their prior asthma medication and current level of asthma control.

The study included patients ranging in age from 12 to 88 years (median age 47 years), and were 66% female and 77% Caucasian.

DULERA was non-inferior to mometasone furoate in terms of time to first serious asthma-related event based on the pre-specified risk margin with an estimated hazard ratio of 1.22 [95% CI: 0.76, 1.94].

Table 7: Serious Asthma-Related Event (Postmarketing Trial) DULERA Actual treatment used for analysis.

n (%) Mometasone Furoate n (%) Total n (%) DULERA vs.

Mometasone Furoate Patients in population 5868 5861 11,729 Hazard Ratio The hazard ratio for time to first event was based on a Cox proportional hazard model with covariates of treatment (DULERA vs.

mometasone furoate) and inhaled corticosteroid dose level (100 mcg vs.

200 mcg), as treated.

(95% CI) Serious Asthma-related Event Results provided for all randomized patients who received at least one dose of DULERA (100 mcg/5 mcg and 200 mcg/5 mcg, two inhalations, prescribed twice daily) or mometasone furoate (100 mcg and 200 mcg, two inhalations, prescribed twice daily).

, Number of patients with an event that occurred within 6 months after the first use of study drug or 7 days after the last date of study drug, whichever date was later.

Patients can have one or more events, but only the first event was counted for analysis.

A blinded adjudication committee determined whether events were asthma related.

39 (0.66) 32 (0.55) 71 (0.6) 1.22 (0.76, 1.94) Asthma-Related Hospitalization (≥24 hr stay) 39 (0.66) 32 (0.55) 71 (0.6) Asthma-Related Intubation (Endotracheal) 0 0 0 Asthma-Related Death 0 0 0 The key efficacy endpoint was time to first asthma exacerbation [defined as a clinical deterioration of asthma associated with systemic corticosteroid use for ≥3 consecutive days (or ≥1 depot injectable), emergency department visits <24 hours requiring systemic corticosteroid, or hospital stays of ≥24 hours].

The estimated hazard ratio for time to first exacerbation for DULERA relative to mometasone furoate was 0.89 [95% CI: 0.8, 0.98].

This outcome was primarily driven by a reduction in those events requiring systemic corticosteroid use, which accounted for 87% of the total number of first asthma exacerbations.

HOW SUPPLIED

16 /STORAGE AND HANDLING 16.1 How Supplied DULERA is available in three strengths and supplied in the following package sizes ( Table 8 ): Table 8 Package NDC Strength Identifier (Color Band) Included on the outer carton, actuator, and canister labels DULERA 50 mcg/5 mcg 120 inhalations 0085-2223-01 Blue DULERA 100 mcg/5 mcg 120 inhalations 0085-7206-01 Yellow DULERA 100 mcg/5 mcg 60 inhalations (institutional pack) 0085-7206-07 Yellow DULERA 200 mcg/5 mcg 120 inhalations 0085-4610-01 Purple DULERA 200 mcg/5 mcg 60 inhalations (institutional pack) 0085-4610-05 Purple Each strength is supplied as a pressurized aluminum canister that has a blue plastic actuator integrated with a dose counter and a green dust cap.

Each 120-inhalation canister has a net fill weight of 13 grams and each 60-inhalation canister has a net fill weight of 8.8 grams.

Each canister is placed into a carton.

Each carton contains 1 canister and a Patient Information leaflet.

Initially the dose counter will display “64” or “124” actuations.

After the initial priming with 4 actuations, the dose counter will read “60” or “120” and the inhaler is now ready for use.

16.2 Storage and Handling Only use the DULERA canister with the DULERA actuator.

Do not use the DULERA actuator with any other inhalation drug product.

Do not use actuators from other products with the DULERA canister.

Do not remove the canister from the actuator because the correct amount of medication may not be discharged; the dose counter may not function properly; reinsertion may cause the dose counter to count down by 1 and discharge a puff.

The correct amount of medication in each inhalation cannot be ensured after the labeled number of actuations from the canister has been used, even though the inhaler may not feel completely empty and may continue to operate.

Discard the inhaler when the labeled number of actuations has been used (the dose counter will read “0”).

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

The 120-inhalation inhaler does not require specific storage orientation.

For the 60-inhalation inhaler, after priming, store the inhaler with the mouthpiece down or in a horizontal position.

For best results, keep the canister at room temperature before use.

Shake well and remove the cap from the mouthpiece of the actuator before using.

Keep out of reach of children.

Avoid spraying in eyes.

Contents Under Pressure: Do not puncture.

Do not use or store near heat or open flame.

Exposure to temperatures above 120°F may cause bursting.

Never throw container into fire or incinerator.

GERIATRIC USE

8.5 Geriatric Use A total of 77 patients 65 years of age and older (11 of whom were 75 years and older) have been treated with DULERA in 3 clinical trials up to 52 weeks in duration.

Similar efficacy and safety results were observed in an additional 28 patients 65 years of age and older who were treated with DULERA in another clinical trial.

No overall differences in safety or effectiveness were observed between these patients and younger patients, but greater sensitivity of some older individuals cannot be ruled out.

As with other products containing beta 2 -agonists, special caution should be observed when using DULERA in geriatric patients who have concomitant cardiovascular disease that could be adversely affected by beta 2 -agonists.

Based on available data for DULERA or its active components, no adjustment of dosage of DULERA in geriatric patients is warranted.

DOSAGE FORMS AND STRENGTHS

3 DULERA is a pressurized metered dose inhaler (MDI) that is available in 2 strengths (100 mcg/5 mcg or 200 mcg/5 mcg) for adult and adolescent patients aged 12 and older and 1 strength (50 mcg/5 mcg) for pediatric patients aged 5 to less than 12 years.

DULERA 50 mcg/5 mcg delivers 50 mcg of mometasone furoate and 5 mcg of formoterol fumarate dihydrate per actuation.

DULERA 100 mcg/5 mcg delivers 100 mcg of mometasone furoate and 5 mcg of formoterol fumarate dihydrate per actuation.

DULERA 200 mcg/5 mcg delivers 200 mcg of mometasone furoate and 5 mcg of formoterol fumarate dihydrate per actuation.

Each strength of DULERA is supplied with a blue colored actuator and green dust cap [see How Supplied/Storage and Handling (16.1) ].

Inhalation aerosol containing a combination of mometasone furoate (50, 100, or 200 mcg) and formoterol fumarate dihydrate (5 mcg) per actuation.

( 3 )

MECHANISM OF ACTION

12.1 Mechanism of Action DULERA: DULERA contains both mometasone furoate and formoterol fumarate; therefore, the mechanisms of actions described below for the individual components apply to DULERA.

These drugs represent two different classes of medications (a synthetic corticosteroid and a selective long-acting beta 2 -adrenergic receptor agonist) that have different effects on clinical, physiological, and inflammatory indices of asthma.

Mometasone furoate : Mometasone furoate is a corticosteroid demonstrating potent anti-inflammatory activity.

The precise mechanism of corticosteroid action on asthma is not known.

Inflammation is an important component in the pathogenesis of asthma.

Corticosteroids have been shown to have a wide range of inhibitory effects on multiple cell types (e.g., mast cells, eosinophils, neutrophils, macrophages, and lymphocytes) and mediators (e.g., histamine, eicosanoids, leukotrienes, and cytokines) involved in inflammation and in the asthmatic response.

These anti-inflammatory actions of corticosteroids may contribute to their efficacy in asthma.

Mometasone furoate has been shown in vitro to exhibit a binding affinity for the human glucocorticoid receptor, which is approximately 12 times that of dexamethasone, 7 times that of triamcinolone acetonide, 5 times that of budesonide, and 1.5 times that of fluticasone.

The clinical significance of these findings is unknown.

Formoterol fumarate : Formoterol fumarate is a long-acting selective beta 2 -adrenergic receptor agonist (beta 2 -agonist).

Inhaled formoterol fumarate acts locally in the lung as a bronchodilator.

In vitro studies have shown that formoterol has more than 200-fold greater agonist activity at beta 2 -receptors than at beta 1 -receptors.

Although beta 2 -receptors are the predominant adrenergic receptors in bronchial smooth muscle and beta 1 -receptors are the predominant receptors in the heart, there are also beta 2 -receptors in the human heart comprising 10% to 50% of the total beta-adrenergic receptors.

The precise function of these receptors has not been established, but they raise the possibility that even highly selective beta 2 -agonists may have cardiac effects.

The pharmacologic effects of beta 2 -adrenoceptor agonist drugs, including formoterol, are at least in part attributable to stimulation of intracellular adenyl cyclase, the enzyme that catalyzes the conversion of adenosine triphosphate (ATP) to cyclic-3′, 5′-adenosine monophosphate (cyclic AMP).

Increased cyclic AMP levels cause relaxation of bronchial smooth muscle and inhibition of release of mediators of immediate hypersensitivity from cells, especially from mast cells.

In vitro tests show that formoterol is an inhibitor of the release of mast cell mediators, such as histamine and leukotrienes, from the human lung.

Formoterol also inhibits histamine-induced plasma albumin extravasation in anesthetized guinea pigs and inhibits allergen-induced eosinophil influx in dogs with airway hyper-responsiveness.

The relevance of these in vitro and animal findings to humans is unknown.

INDICATIONS AND USAGE

1 DULERA is a combination product containing a corticosteroid and a long-acting beta 2 -adrenergic agonist (LABA) indicated for: Treatment of asthma in patients 5 years of age and older.

( 1.1 ) Important Limitation of Use: Not indicated for the relief of acute bronchospasm.

( 1.1 ) 1.1 Treatment of Asthma DULERA is indicated for the twice-daily treatment of asthma in patients 5 years of age and older.

DULERA should be used for patients not adequately controlled on a long-term asthma-control medication such as an inhaled corticosteroid (ICS) or whose disease warrants initiation of treatment with both an ICS and long-acting beta 2 -adrenergic agonist (LABA).

Important Limitation of Use: DULERA is NOT indicated for the relief of acute bronchospasm.

PEDIATRIC USE

8.4 Pediatric Use The safety and effectiveness of DULERA have been established in patients 12 years of age and older in 3 clinical trials up to 52 weeks in duration.

In the 3 clinical trials, 101 patients 12 to 17 years of age were treated with DULERA.

Patients in this age-group demonstrated efficacy results similar to those observed in patients 18 years of age and older.

There were no obvious differences in the type or frequency of adverse reactions reported in this age group compared to patients 18 years of age and older.

Similar efficacy and safety results were observed in an additional 22 patients 12 to 17 years of age who were treated with DULERA in another clinical trial.

The safety and effectiveness of DULERA 50 mcg/5 mcg, two inhalations twice daily, have been established in patients with asthma aged 5 to less than 12 years in clinical trials up to 24 weeks of treatment duration.

Patients in this age group demonstrated efficacy and safety results similar to those observed in patients aged 12 years and older who were treated with DULERA [see Adverse Reactions (6.1) and Clinical Studies (14.1) ].

The safety and effectiveness of DULERA have not been established in children younger than 5 years of age.

Controlled clinical studies have shown that inhaled corticosteroids may cause a reduction in growth velocity in pediatric patients.

In these studies, the mean reduction in growth velocity was approximately 1 cm per year (range 0.3 to 1.8 per year) and appears to depend upon dose and duration of exposure.

This effect was observed in the absence of laboratory evidence of hypothalamic-pituitary-adrenal (HPA) axis suppression, suggesting that growth velocity is a more sensitive indicator of systemic corticosteroid exposure in pediatric patients than some commonly used tests of HPA axis function.

The long-term effects of this reduction in growth velocity associated with orally inhaled corticosteroids, including the impact on final adult height, are unknown.

The potential for “catch up” growth following discontinuation of treatment with orally inhaled corticosteroids has not been adequately studied.

The growth of children and adolescents receiving orally inhaled corticosteroids, including DULERA, should be monitored routinely (e.g., via stadiometry).

If a child or adolescent on any corticosteroid appears to have growth suppression, the possibility that he/she is particularly sensitive to this effect should be considered.

The potential growth effects of prolonged treatment should be weighed against clinical benefits obtained and the risks associated with alternative therapies.

To minimize the systemic effects of orally inhaled corticosteroids, including DULERA, each patient should be titrated to his/her lowest effective dose [see Dosage and Administration (2.2) ] .

PREGNANCY

8.1 Pregnancy Risk Summary There are no randomized clinical studies of DULERA, mometasone furoate, or formoterol fumarate in pregnant women.

There are clinical considerations with the use of DULERA in pregnant women [see Clinical Considerations ] .

Animal reproduction studies with DULERA are not available; however, studies are available with its individual components, mometasone furoate and formoterol fumarate.

In animal reproduction studies, subcutaneous administration of mometasone furoate to pregnant mice, rats, or rabbits caused increased fetal malformations and decreased fetal survival and growth following administration of doses that produced exposures approximately 1/3 to 8 times the maximum recommended human dose (MRHD) on a mcg/m 2 or AUC basis [see Data ] .

However, experience with oral corticosteroids suggests that rodents are more prone to teratogenic effects from corticosteroid exposure than humans.

In animal reproduction studies, oral administration of formoterol fumarate to pregnant rats and rabbits caused increased fetal malformations (rats and rabbits), decreased fetal weight (rats), and increased neonatal mortality (rats) following administration of doses that produced exposures approximately 1200 to 49,000 times the MRHD on a mg/m 2 or AUC basis [see Data ] .

These adverse effects generally occurred at large multiples of the MRHD when formoterol fumarate was administered by the oral route to achieve high systemic exposures.

No effects were observed in a study with rats that received formoterol fumarate by the inhalation route at an exposure approximately 500 times the MRHD.

The estimated background risk of major birth defects and miscarriage for the indicated population is unknown.

In the U.S.

general population, the estimated risk of major birth defects and miscarriage in clinically recognized pregnancies is 2 to 4% and 15 to 20%, respectively.

Clinical Considerations Disease-associated maternal and/or embryo/fetal risk In women with poorly or moderately controlled asthma, there is an increased risk of several perinatal adverse outcomes such as preeclampsia in the mother and prematurity, low birth weight, and small for gestational age in the neonate.

Pregnant women with asthma should be closely monitored and medication adjusted as necessary to maintain optimal asthma control.

Labor or delivery There are no adequate and well-controlled human studies that have studied the effects of DULERA during labor and delivery.

Because of the potential for beta-agonist interference with uterine contractility, use of DULERA during labor should be restricted to those patients in whom the benefits clearly outweigh the risk.

Data Animal Data Mometasone Furoate In an embryofetal development study with pregnant mice dosed throughout the period of organogenesis, mometasone furoate produced cleft palate at an exposure approximately one-third of the MRHD (on a mcg/m 2 basis with maternal subcutaneous doses of 60 mcg/kg and above) and decreased fetal survival at an exposure approximately equivalent to the MRHD (on a mcg/m 2 basis with a maternal subcutaneous dose of 180 mcg/kg).

No toxicity was observed with a dose that produced an exposure approximately one-tenth of the MRHD (on a mcg/m 2 basis with maternal topical dermal doses of 20 mcg/kg and above).

In an embryofetal development study with pregnant rats dosed throughout the period of organogenesis, mometasone furoate produced fetal umbilical hernia at exposures approximately 6 times the MRHD (on a mcg/m 2 basis with maternal topical dermal doses of 600 mcg/kg and above) and delays in fetal ossification at exposures approximately 3 times the MRHD (on a mcg/m 2 basis with maternal topical dermal doses of 300 mcg/kg and above).

In another reproductive toxicity study, pregnant rats were dosed with mometasone furoate throughout pregnancy or late in gestation.

Treated animals had prolonged and difficult labor, fewer live births, lower birth weight, and reduced early pup survival at an exposure that was approximately 8 times the MRHD (on an area under the curve (AUC) basis with a maternal subcutaneous dose of 15 mcg/kg).

There were no findings with an exposure approximately 4 times the MRHD (on an AUC basis with a maternal subcutaneous dose of 7.5 mcg/kg).

Embryofetal development studies were conducted with pregnant rabbits dosed with mometasone furoate by either the topical dermal route or oral route throughout the period of organogenesis.

In the study using the topical dermal route, mometasone furoate caused multiple malformations in fetuses (e.g., flexed front paws, gallbladder agenesis, umbilical hernia, hydrocephaly) at an exposure approximately 3 times the MRHD (on a mcg/m 2 basis with maternal topical dermal doses of 150 mcg/kg and above).

In the study using the oral route, mometasone furoate caused increased fetal resorptions and cleft palate and/or head malformations (hydrocephaly and domed head) at an exposure approximately 1/2 of the MRHD (on AUC basis with a maternal oral dose of 700 mcg/kg).

At an exposure approximately 2 times the MRHD (on an AUC basis with a maternal oral dose of 2800 mcg/kg), most litters were aborted or resorbed.

No effects were observed at an exposure approximately 1/10 of the MRHD (on an AUC basis with a maternal oral dose of 140 mcg/kg).

Formoterol Fumarate In embryofetal development studies with pregnant rats and rabbits dosed throughout the period of organogenesis, formoterol fumarate did not cause malformations in either species.

However, for pregnant rats dosed throughout organogenesis, formoterol fumarate caused delayed fetal ossification at an exposure approximately 80 times the MRHD (on a mcg/m 2 basis with maternal oral doses of 200 mcg/kg and higher) and decreased fetal weight at an exposure approximately 2400 times the MRHD (on a mcg/m 2 basis with maternal oral doses of 6000 mcg/kg and above).

In a pre- and post-natal development study with rats dosed during the late stage of pregnancy, formoterol fumarate caused stillbirth and neonatal mortality at an exposure approximately 2400 times the MRHD (on a mcg/m 2 basis with maternal oral doses of 6000 mcg/kg and above).

However, no effects were observed in this study at an exposure approximately 80 times the MRHD (on a mcg/m 2 basis with a maternal oral dose of 200 mcg/kg).

In embryofetal development studies, conducted by another testing laboratory, with pregnant rats and rabbits dosed throughout the period of organogenesis, formoterol fumarate was teratogenic in both species.

Umbilical hernia, a malformation, was observed in rat fetuses at exposures approximately 1200 times the MRHD (on a mcg/m 2 basis with maternal oral doses of 3000 mcg/kg/day and above).

Brachygnathia, a skeletal malformation, was observed in rat fetuses at an exposure approximately 6100 times the MRHD (on a mcg/m 2 basis with a maternal oral dose of 15,000 mcg/kg/day).

In another study with rats, no teratogenic effects were observed with exposures up to approximately 500 times the MRHD (on a mcg/m 2 basis with a maternal inhalation dose of 1200 mcg/kg/day).

Subcapsular cysts on the liver were observed in rabbit fetuses at an exposure approximately 49,000 times the MRHD (on a mcg/m 2 basis with a maternal oral dose of 60,000 mcg/kg/day).

No teratogenic effects were observed with exposures up to approximately 3000 times the MRHD (on a mcg/m 2 basis with a maternal oral dose of 3500 mcg/kg).

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS LABA monotherapy increases the risk of serious asthma-related events.

( 5.1 ) Deterioration of disease and acute episodes: Do not initiate in acutely deteriorating asthma or to treat acute symptoms.

( 5.2 ) Use with additional long-acting beta 2 -agonist: Do not use in combination because of risk of overdose.

( 5.3 ) Localized infections: Candida albicans infection of the mouth and throat may occur.

Monitor patients periodically for signs of adverse effects on the oral cavity.

After dosing, advise patients to rinse their mouth with water and spit out contents without swallowing.

( 5.4 ) Immunosuppression: Potential worsening of existing tuberculosis, fungal, bacterial, viral, or parasitic infection; or ocular herpes simplex infections.

More serious or even fatal course of chickenpox or measles can occur in susceptible patients.

Use with caution in patients with these infections because of the potential for worsening of these infections.

( 5.5 ) Transferring patients from systemic corticosteroids: Risk of impaired adrenal function when transferring from oral steroids.

Taper patients slowly from systemic corticosteroids if transferring to DULERA.

( 5.6 ) Hypercorticism and adrenal suppression: May occur with very high dosages or at the regular dosage in susceptible individuals.

If such changes occur, discontinue DULERA slowly.

( 5.7 ) Strong cytochrome P450 3A4 inhibitors (e.g., ritonavir): Risk of increased systemic corticosteroid effects.

Exercise caution when used with DULERA.

( 5.8 ) Paradoxical bronchospasm: Discontinue DULERA and institute alternative therapy if paradoxical bronchospasm occurs.

( 5.9 ) Patients with cardiovascular disorders: Use with caution because of beta-adrenergic stimulation.

( 5.11 ) Decreases in bone mineral density: Monitor patients with major risk factors for decreased bone mineral content.

( 5.12 ) Effects on growth: Monitor growth of pediatric patients.

( 5.13 ) Glaucoma and cataracts: Consider referral to an ophthalmologist in patients who develop ocular symptoms or use DULERA long term.

( 5.14 ) Coexisting conditions: Use with caution in patients with aneurysm, pheochromocytoma, convulsive disorders, thyrotoxicosis, diabetes mellitus, and ketoacidosis.

( 5.15 ) Hypokalemia and hyperglycemia: Be alert to hypokalemia and hyperglycemia.

( 5.16 ) 5.1 Serious Asthma-Related Events – Hospitalizations, Intubations, and Death Use of LABA as monotherapy (without ICS) for asthma is associated with an increased risk of asthma-related death [see Salmeterol Multicenter Asthma Research Trial (SMART) ] .

Available data from controlled clinical trials also suggest that use of LABA as monotherapy increases the risk of asthma-related hospitalization in pediatric and adolescent patients.

These findings are considered a class effect of LABA monotherapy.

When LABA are used in fixed-dose combination with ICS, data from large clinical trials do not show a significant increase in the risk of serious asthma-related events (hospitalizations, intubations, death) compared to ICS alone [see Serious Asthma-Related Events with ICS/LABA ] .

Serious Asthma-Related Events with ICS/LABA Four large, 26-week, randomized, blinded, active-controlled clinical safety trials were conducted to evaluate the risk of serious asthma-related events when LABA were used in fixed-dose combination with ICS compared to ICS alone in patients with asthma.

Three trials included adult and adolescent patients aged ≥12 years: one trial compared mometasone furoate/formoterol (DULERA) to mometasone furoate [see Clinical Studies (14.1) ] ; one trial compared fluticasone propionate/salmeterol inhalation powder to fluticasone propionate inhalation powder; and one trial compared budesonide/formoterol to budesonide.

The fourth trial included pediatric patients 4 to 11 years of age and compared fluticasone propionate/salmeterol inhalation powder to fluticasone propionate inhalation powder.

The primary safety endpoint for all four trials was serious asthma-related events (hospitalizations, intubations and death).

A blinded adjudication committee determined whether events were asthma-related.

The three adult and adolescent trials were designed to rule out a risk margin of 2.0, and the pediatric trial was designed to rule out a risk of 2.7.

Each individual trial met its pre-specified objective and demonstrated non-inferiority of ICS/LABA to ICS alone.

A meta-analysis of the three adult and adolescent trials did not show a significant increase in risk of a serious asthma-related event with ICS/LABA fixed-dose combination compared with ICS alone (Table 1).

These trials were not designed to rule out all risk for serious asthma-related events with ICS/LABA compared with ICS.

Table 1: Meta-Analysis of Serious Asthma-Related Events in Patients with Asthma Aged 12 Years and Older ICS/LABA (N=17,537) Randomized patients who had taken at least 1 dose of study drug.

Planned treatment used for analysis.

ICS (N=17,552) ICS/LABA vs.

ICS Hazard ratio (95% CI) Estimated using a Cox proportional hazards model for time to first event with baseline hazards stratified by each of the 3 trials.

ICS = Inhaled Corticosteroid, LABA = Long-acting Beta 2 -adrenergic Agonist.

Serious asthma-related event Number of patients with events that occurred within 6 months after the first use of study drug or 7 days after the last date of study drug, whichever date was later.

Patients can have one or more events, but only the first event was counted for analysis.

A single, blinded, independent adjudication committee determined whether events were asthma-related.

116 105 1.10 (0.85, 1.44) Asthma-related death 2 0 Asthma-related intubation (endotracheal) 1 2 Asthma-related hospitalization (≥24 hour stay) 115 105 The pediatric safety trial included 6208 pediatric patients 4 to 11 years of age who received ICS/LABA (fluticasone propionate/salmeterol inhalation powder) or ICS (fluticasone propionate inhalation powder).

In this trial, 27/3107 (0.9%) patients randomized to ICS/LABA and 21/3101 (0.7%) patients randomized to ICS experienced a serious asthma-related event.

There were no asthma-related deaths or intubations.

ICS/LABA did not show a significantly increased risk of a serious asthma-related event compared to ICS based on the pre-specified risk margin (2.7), with an estimated hazard ratio of time to first event of 1.29 (95% CI: 0.73, 2.27).

Salmeterol Multicenter Asthma Research Trial (SMART) A 28-week, placebo-controlled U.S.

trial that compared the safety of salmeterol with placebo, each added to usual asthma therapy, showed an increase in asthma-related deaths in patients receiving salmeterol (13/13,176 in patients treated with salmeterol vs.

3/13,179 in patients treated with placebo; relative risk: 4.37 [95% CI: 1.25, 15.34]).

Use of background ICS was not required in SMART.

The increased risk of asthma-related death is considered a class effect of LABA monotherapy.

Formoterol Monotherapy Studies Clinical studies with formoterol used as monotherapy suggested a higher incidence of serious asthma exacerbation in patients who received formoterol than in those who received placebo.

The sizes of these studies were not adequate to precisely quantify the difference in serious asthma exacerbations between treatment groups.

5.2 Deterioration of Disease and Acute Episodes DULERA should not be initiated in patients during rapidly deteriorating or potentially life-threatening episodes of asthma.

DULERA has not been studied in patients with acutely deteriorating asthma.

The initiation of DULERA in this setting is not appropriate.

Increasing use of inhaled, short-acting beta 2 -agonists is a marker of deteriorating asthma.

In this situation, the patient requires immediate re-evaluation with reassessment of the treatment regimen, giving special consideration to the possible need for replacing the current strength of DULERA with a higher strength, adding additional inhaled corticosteroid, or initiating systemic corticosteroids.

Patients should not use more than 2 inhalations twice daily (morning and evening) of DULERA.

DULERA is not indicated for the relief of acute symptoms, i.e., as rescue therapy for the treatment of acute episodes of bronchospasm.

An inhaled, short-acting beta 2 -agonist, not DULERA, should be used to relieve acute symptoms such as shortness of breath.

When beginning treatment with DULERA, patients who have been taking oral or inhaled, short-acting beta 2 -agonists on a regular basis (e.g., 4 times a day) should be instructed to discontinue the regular use of these drugs.

5.3 Excessive Use of DULERA and Use with Other Long-Acting Beta 2 -Agonists As with other inhaled drugs containing beta 2 -adrenergic agents, DULERA should not be used more often than recommended, at higher doses than recommended, or in conjunction with other medications containing long-acting beta 2 -agonists, as an overdose may result.

Clinically significant cardiovascular effects and fatalities have been reported in association with excessive use of inhaled sympathomimetic drugs.

Patients using DULERA should not use an additional long-acting beta 2 -agonist (e.g., salmeterol, formoterol fumarate, arformoterol tartrate) for any reason, including prevention of exercise-induced bronchospasm (EIB) or the treatment of asthma.

5.4 Local Effects In clinical trials, the development of localized infections of the mouth and pharynx with Candida albicans have occurred in patients treated with DULERA.

If oropharyngeal candidiasis develops, treat with appropriate local or systemic (i.e., oral) antifungal therapy while remaining on treatment with DULERA therapy, but at times therapy with DULERA may need to be interrupted.

To reduce the risk of oropharyngeal candidiasis, after dosing with DULERA, advise patients to rinse their mouth with water and spit out the contents without swallowing.

5.5 Immunosuppression Persons who are using drugs that suppress the immune system are more susceptible to infections than healthy individuals.

Chickenpox and measles, for example, can have a more serious or even fatal course in susceptible children or adults using corticosteroids.

In such children or adults who have not had these diseases or who are not properly immunized, particular care should be taken to avoid exposure.

How the dose, route, and duration of corticosteroid administration affect the risk of developing a disseminated infection is not known.

The contribution of the underlying disease and/or prior corticosteroid treatment to the risk is also not known.

If exposed to chickenpox, prophylaxis with varicella zoster immune globulin (VZIG) or pooled intravenous immunoglobulin (IVIG) may be indicated.

If exposed to measles, prophylaxis with pooled intramuscular immunoglobulin (IG) may be indicated.

(See the respective package inserts for complete VZIG and IG prescribing information.) If chickenpox develops, treatment with antiviral agents may be considered.

DULERA should be used with caution, if at all, in patients with active or quiescent tuberculosis infection of the respiratory tract, untreated systemic fungal, bacterial, viral, or parasitic infections; or ocular herpes simplex.

5.6 Transferring Patients from Systemic Corticosteroid Therapy Particular care is needed for patients who are transferred from systemically active corticosteroids to DULERA because deaths due to adrenal insufficiency have occurred in asthmatic patients during and after transfer from systemic corticosteroids to less systemically available inhaled corticosteroids.

After withdrawal from systemic corticosteroids, a number of months are required for recovery of hypothalamic-pituitary-adrenal (HPA) function.

Patients who have been previously maintained on 20 mg or more per day of prednisone (or its equivalent) may be most susceptible, particularly when their systemic corticosteroids have been almost completely withdrawn.

During this period of HPA suppression, patients may exhibit signs and symptoms of adrenal insufficiency when exposed to trauma, surgery, or infection (particularly gastroenteritis) or other conditions associated with severe electrolyte loss.

Although DULERA may improve control of asthma symptoms during these episodes, in recommended doses it supplies less than normal physiological amounts of corticosteroid systemically and does NOT provide the mineralocorticoid activity necessary for coping with these emergencies.

During periods of stress or severe asthma attack, patients who have been withdrawn from systemic corticosteroids should be instructed to resume oral corticosteroids (in large doses) immediately and to contact their physicians for further instruction.

These patients should also be instructed to carry a medical identification card indicating that they may need supplementary systemic corticosteroids during periods of stress or severe asthma attack.

Patients requiring systemic corticosteroids should be weaned slowly from systemic corticosteroid use after transferring to DULERA.

Lung function (FEV 1 or PEF), beta-agonist use, and asthma symptoms should be carefully monitored during withdrawal of systemic corticosteroids.

In addition to monitoring asthma signs and symptoms, patients should be observed for signs and symptoms of adrenal insufficiency such as fatigue, lassitude, weakness, nausea and vomiting, and hypotension.

Transfer of patients from systemic corticosteroid therapy to DULERA may unmask allergic conditions previously suppressed by the systemic corticosteroid therapy, e.g., rhinitis, conjunctivitis, eczema, arthritis, and eosinophilic conditions.

During withdrawal from oral corticosteroids, some patients may experience symptoms of systemically active corticosteroid withdrawal, e.g., joint and/or muscular pain, lassitude, and depression, despite maintenance or even improvement of respiratory function.

5.7 Hypercorticism and Adrenal Suppression Mometasone furoate, a component of DULERA, will often help control asthma symptoms with less suppression of HPA function than therapeutically equivalent oral doses of prednisone.

Since mometasone furoate is absorbed into the circulation and can be systemically active at higher doses, the beneficial effects of DULERA in minimizing HPA dysfunction may be expected only when recommended dosages are not exceeded and individual patients are titrated to the lowest effective dose.

Because of the possibility of systemic absorption of inhaled corticosteroids, patients treated with DULERA should be observed carefully for any evidence of systemic corticosteroid effects.

Particular care should be taken in observing patients postoperatively or during periods of stress for evidence of inadequate adrenal response.

It is possible that systemic corticosteroid effects such as hypercorticism and adrenal suppression (including adrenal crisis) may appear in a small number of patients, particularly when mometasone furoate is administered at higher than recommended doses over prolonged periods of time.

If such effects occur, the dosage of DULERA should be reduced slowly, consistent with accepted procedures for reducing systemic corticosteroids and for management of asthma symptoms.

5.8 Drug Interactions with Strong Cytochrome P450 3A4 Inhibitors Caution should be exercised when considering the coadministration of DULERA with ketoconazole, and other known strong CYP3A4 inhibitors (e.g., ritonavir, cobicistat-containing products, atazanavir, clarithromycin, indinavir, itraconazole, nefazodone, nelfinavir, saquinavir, telithromycin) because adverse effects related to increased systemic exposure to mometasone furoate may occur [see Drug Interactions (7.1) and Clinical Pharmacology (12.3) ].

5.9 Paradoxical Bronchospasm and Upper Airway Symptoms DULERA may produce inhalation induced bronchospasm with an immediate increase in wheezing after dosing that may be life-threatening.

If inhalation induced bronchospasm occurs, it should be treated immediately with an inhaled, short-acting bronchodilator.

DULERA should be discontinued immediately and alternative therapy instituted.

5.10 Immediate Hypersensitivity Reactions Immediate hypersensitivity reactions may occur after administration of DULERA, as demonstrated by cases of urticaria, flushing, allergic dermatitis, and bronchospasm.

5.11 Cardiovascular and Central Nervous System Effects Excessive beta-adrenergic stimulation has been associated with seizures, angina, hypertension or hypotension, tachycardia with rates up to 200 beats/min, arrhythmias, nervousness, headache, tremor, palpitation, nausea, dizziness, fatigue, malaise, and insomnia.

Therefore, DULERA should be used with caution in patients with cardiovascular disorders, especially coronary insufficiency, cardiac arrhythmias, and hypertension.

Formoterol fumarate, a component of DULERA, can produce a clinically significant cardiovascular effect in some patients as measured by pulse rate, blood pressure, and/or symptoms.

Although such effects are uncommon after administration of DULERA at recommended doses, if they occur, the drug may need to be discontinued.

In addition, beta-agonists have been reported to produce ECG changes, such as flattening of the T wave, prolongation of the QTc interval, and ST segment depression.

The clinical significance of these findings is unknown.

Fatalities have been reported in association with excessive use of inhaled sympathomimetic drugs.

5.12 Reduction in Bone Mineral Density Decreases in bone mineral density (BMD) have been observed with long-term administration of products containing inhaled corticosteroids, including mometasone furoate, one of the components of DULERA.

The clinical significance of small changes in BMD with regard to long-term outcomes, such as fracture, is unknown.

Patients with major risk factors for decreased bone mineral content, such as prolonged immobilization, family history of osteoporosis, or chronic use of drugs that can reduce bone mass (e.g., anticonvulsants and corticosteroids) should be monitored and treated with established standards of care.

In a 2-year double-blind study in 103 male and female asthma patients 18 to 50 years of age previously maintained on bronchodilator therapy (Baseline FEV 1 85%–88% predicted), treatment with mometasone furoate dry powder inhaler (DPI) 200 mcg twice daily resulted in significant reductions in lumbar spine (LS) BMD at the end of the treatment period compared to placebo.

The mean change from Baseline to Endpoint in the lumbar spine BMD was -0.015 (-1.43%) for the mometasone furoate group compared to 0.002 (0.25%) for the placebo group.

In another 2-year double-blind study in 87 male and female asthma patients 18 to 50 years of age previously maintained on bronchodilator therapy (Baseline FEV 1 82%–83% predicted), treatment with mometasone furoate 400 mcg twice daily demonstrated no statistically significant changes in lumbar spine BMD at the end of the treatment period compared to placebo.

The mean change from Baseline to Endpoint in the lumbar spine BMD was -0.018 (-1.57%) for the mometasone furoate group compared to -0.006 (-0.43%) for the placebo group.

5.13 Effect on Growth Orally inhaled corticosteroids, including DULERA, may cause a reduction in growth velocity when administered to pediatric patients.

Monitor the growth of pediatric patients receiving DULERA routinely (e.g., via stadiometry).

To minimize the systemic effects of orally inhaled corticosteroids, including DULERA, titrate each patient’s dose to the lowest dosage that effectively controls his/her symptoms [see Use in Specific Populations (8.4) ] .

5.14 Glaucoma and Cataracts Glaucoma, increased intraocular pressure, and cataracts have been reported following the use of long-term administration of inhaled corticosteroids, including mometasone furoate, a component of DULERA.

Consider referral to an ophthalmologist in patients who develop ocular symptoms or use DULERA long term [see Adverse Reactions (6) ] .

5.15 Coexisting Conditions DULERA, like other medications containing sympathomimetic amines, should be used with caution in patients with aneurysm, pheochromocytoma, convulsive disorders, or thyrotoxicosis; and in patients who are unusually responsive to sympathomimetic amines.

Doses of the related beta 2 -agonist albuterol, when administered intravenously, have been reported to aggravate preexisting diabetes mellitus and ketoacidosis.

5.16 Hypokalemia and Hyperglycemia Beta 2 -agonist medications may produce significant hypokalemia in some patients, possibly through intracellular shunting, which has the potential to produce adverse cardiovascular effects.

The decrease in serum potassium is usually transient, not requiring supplementation.

Clinically significant changes in blood glucose and/or serum potassium were seen infrequently during clinical studies with DULERA at recommended doses.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION Advise the patient to read the FDA-approved patient labeling ( Patient Information and Instructions for Use ).

Serious Asthma-Related Events Inform patients with asthma that LABA when used alone increases the risk of asthma-related hospitalization, or asthma-related death.

Available data show that when ICS and LABA are used together, such as with DULERA, there is not a significant increase in risk of these events.

Not for Acute Symptoms DULERA is not indicated to relieve acute asthma symptoms and extra doses should not be used for that purpose.

Treat acute symptoms with an inhaled, short-acting, beta 2 -agonist (the health care provider should prescribe the patient with such medication and instruct the patient in how it should be used).

Instruct patients to seek medical attention immediately if they experience any of the following: If their symptoms worsen Significant decrease in lung function as outlined by the physician If they need more inhalations of a short-acting beta 2 -agonist than usual Advise patients not to increase the dose or frequency of DULERA.

Do not exceed the daily dosage of DULERA of two inhalations twice daily.

If they miss a dose, instruct patients to take their next dose at the same time they normally do.

DULERA provides bronchodilation for up to 12 hours.

Instruct patients not to stop or reduce DULERA therapy without physician/provider guidance since symptoms may recur after discontinuation [see Warnings and Precautions (5.2) ] .

Do Not Use Additional Long-Acting Beta 2 -Agonists When patients are prescribed DULERA, other long-acting beta 2 -agonists should not be used [see Warnings and Precautions (5.3) ] .

Risks Associated With Corticosteroid Therapy Local Effects: Advise patients that localized infections with Candida albicans occurred in the mouth and pharynx in some patients.

If oropharyngeal candidiasis develops, treat with appropriate local or systemic (i.e., oral) antifungal therapy while still continuing with DULERA therapy, but at times therapy with DULERA may need to be temporarily interrupted under close medical supervision.

Rinsing the mouth after inhalation is advised [see Warnings and Precautions (5.4) ].

Immunosuppression: Warn patients who are on immunosuppressant doses of corticosteroids to avoid exposure to chickenpox or measles and, if exposed, to consult their physician without delay.

Inform patients of potential worsening of existing tuberculosis, fungal, bacterial, viral, or parasitic infections, or ocular herpes simplex [see Warnings and Precautions (5.5) ].

Hypercorticism and Adrenal Suppression: Advise patients that DULERA may cause systemic corticosteroid effects of hypercorticism and adrenal suppression.

Additionally, instruct patients that deaths due to adrenal insufficiency have occurred during and after transfer from systemic corticosteroids.

Instruct patients to taper slowly from systemic corticosteroids if transferring to DULERA [see Warnings and Precautions (5.7) ].

Reduction in Bone Mineral Density: Advise patients who are at an increased risk for decreased BMD that the use of corticosteroids may pose an additional risk and that they should be monitored and, where appropriate, be treated for this condition [see Warnings and Precautions (5.12) ].

Reduced Growth Velocity: Inform patients that orally inhaled corticosteroids, a component of DULERA, may cause a reduction in growth velocity when administered to pediatric patients.

Physicians should closely follow the growth of pediatric patients taking corticosteroids by any route [see Warnings and Precautions (5.13) ].

Glaucoma and Cataracts: Long-term use of inhaled corticosteroids may increase the risk of some eye problems (glaucoma or cataracts); consider regular eye examinations [see Warnings and Precautions (5.14) ].

Risks Associated With Beta-Agonist Therapy Inform patients that treatment with beta 2 -agonists may lead to adverse events which include palpitations, chest pain, rapid heart rate, tremor or nervousness [see Warnings and Precautions (5.11) ].

Instructions for Use Instruct patients regarding the following: Read the Patient Information before use and follow the Instructions for Use carefully.

Remind patients to: Remove the cap from the mouthpiece of the actuator before use.

After dosing, rinse their mouth with water without swallowing and spit out after breathing in the medicine.

This will help reduce the risk of oropharyngeal candidiasis.

Not remove the canister from the actuator.

Not wash inhaler in water.

The mouthpiece should be cleaned using a dry wipe after every 7 days of use.

DOSAGE AND ADMINISTRATION

2 For oral inhalation only.

( 2.1 ) Treatment of asthma in patients ≥12 years: 2 inhalations twice daily of DULERA 100 mcg/5 mcg or 200 mcg/5 mcg.

Starting dosage is based on disease severity.

( 2.2 ) Treatment of asthma in patients aged 5 to less than 12 years: 2 inhalations twice daily of DULERA 50 mcg/5 mcg.

( 2.2 ) 2.1 Administration Information Administer DULERA as two inhalations twice daily every day (morning and evening) by the orally inhaled route (see Patient Instructions for Use in the Patient Information leaflet).

Do not use more than two inhalations twice daily of the prescribed strength of DULERA as some patients are more likely to experience adverse effects with higher doses of formoterol.

If symptoms arise between doses, an inhaled short-acting beta 2 -agonist should be taken for immediate relief.

Shake well prior to each inhalation.

After each dose, advise patients to rinse their mouth with water and, without swallowing, spit out the contents to help reduce the risk of oropharyngeal candidiasis.

Remove the cap from the mouthpiece of the actuator before using DULERA.

Prime DULERA before using for the first time by releasing 4 test sprays into the air, away from the face, shaking well before each spray.

In cases where the inhaler has not been used for more than 5 days, prime the inhaler again by releasing 4 test sprays into the air, away from the face, shaking well before each spray.

Only use the DULERA canister with the DULERA actuator.

Do not use the DULERA actuator with any other inhalation drug product.

Do not use actuators from other products with the DULERA canister.

2.2 Recommended Dosage Administer DULERA as two inhalations twice daily every day (morning and evening) by the orally inhaled route.

Shake well prior to each inhalation.

Individual patients may experience a variable time to onset and degree of symptom relief.

If symptoms arise between doses, use an inhaled short-acting beta 2 -agonist for immediate relief.

Improvement in lung function following administration of DULERA can occur within 5 minutes of treatment, although the maximum benefit may not be achieved for 1 week or longer after beginning treatment.

Adult and Adolescent Patients Aged 12 Years and Older For patients 12 years and older, the dosage is either 2 inhalations twice daily of DULERA 100 mcg/5 mcg or DULERA 200 mcg/5 mcg.

When choosing the starting dosage strength of DULERA, consider the patients’ disease severity, based on their previous asthma therapy, including the inhaled corticosteroid dosage, as well as the patients’ current control of asthma symptoms and risk of future exacerbation.

For patients who do not respond adequately after 2 weeks of therapy with two inhalations of DULERA 100 mcg/5 mcg twice daily (morning and evening), increasing the dosage to two inhalations of DULERA 200 mcg/5 mcg twice daily (morning and evening) may provide additional asthma control.

The maximum recommended dosage is two inhalations of DULERA 200 mcg/5 mcg twice daily (maximum daily dosage 800 mcg/20 mcg).

After asthma stability has been achieved, it may be desirable to titrate to the lowest effective dosage to reduce the possibility of side effects.

If a previously effective dosage regimen of DULERA fails to provide adequate control of asthma, re-evaluate the therapeutic regimen and consider additional therapeutic options, e.g., replacing the current strength of DULERA with a higher strength, adding additional inhaled corticosteroid, or initiating oral corticosteroids.

Pediatric Patients Aged 5 to Less Than 12 Years For patients aged 5 to less than 12 years, the dosage is 2 inhalations of DULERA 50 mcg/5 mcg twice daily.

The maximum daily dosage is 200 mcg/20 mcg.