Bisoprolol Fumarate 10 MG / Hydrochlorothiazide 6.25 MG Oral Tablet [Ziac]

Generic Name: BISOPROLOL FUMARATE AND HYDROCHLOROTHIAZIDE
Brand Name: Ziac
  • Substance Name(s):
  • BISOPROLOL FUMARATE
  • HYDROCHLOROTHIAZIDE

WARNINGS

Cardiac Failure In general, beta-blocking agents should be avoided in patients with overt congestive failure.

However, in some patients with compensated cardiac failure, it may be necessary to utilize these agents.

In such situations, they must be used cautiously.

Patients Without a History of Cardiac Failure Continued depression of the myocardium with beta-blockers can, in some patients, precipitate cardiac failure.

At the first signs or symptoms of heart failure, discontinuation of ZIAC should be considered.

In some cases ZIAC therapy can be continued while heart failure is treated with other drugs.

Abrupt Cessation of Therapy Exacerbations of angina pectoris and, in some instances, myocardial infarction or ventricular arrhythmia, have been observed in patients with coronary artery disease following abrupt cessation of therapy with beta-blockers.

Such patients should, therefore, be cautioned against interruption or discontinuation of therapy without the physician’s advice.

Even in patients without overt coronary artery disease, it may be advisable to taper therapy with ZIAC (bisoprolol fumarate and hydrochlorothiazide) over approximately 1 week with the patient under careful observation.

If withdrawal symptoms occur, beta-blocking agent therapy should be reinstituted, at least temporarily.

Peripheral Vascular Disease Beta-blockers can precipitate or aggravate symptoms of arterial insufficiency in patients with peripheral vascular disease.

Caution should be exercised in such individuals.

Bronchospastic Disease PATIENTS WITH BRONCHOSPASTIC PULMONARY DISEASE SHOULD, IN GENERAL, NOT RECEIVE BETA-BLOCKERS.

Because of the relative beta 1 -selectivity of bisoprolol fumarate, ZIAC may be used with caution in patients with bronchospastic disease who do not respond to, or who cannot tolerate other antihypertensive treatment.

Since beta 1 -selectivity is not absolute, the lowest possible dose of ZIAC should be used.

A beta 2 agonist (bronchodilator) should be made available.

Major Surgery Chronically administered beta-blocking therapy should not be routinely withdrawn prior to major surgery; however, the impaired ability of the heart to respond to reflex adrenergic stimuli may augment the risks of general anesthesia and surgical procedures.

Hypoglycemia Beta-blockers may prevent early warning signs of hypoglycemia, such as tachycardia, and increase the risk for severe or prolonged hypoglycemia at any time during treatment, especially in patients with diabetes mellitus or children and patients who are fasting (i.e., surgery, not eating regularly, or are vomiting).

If severe hypoglycemia occurs, patients should be instructed to seek emergency treatment.

Also, latent diabetes mellitus may become manifest and diabetic patients given thiazides may require adjustment of their insulin dose.

Thyrotoxicosis Beta-adrenergic blockade may mask clinical signs of hyperthyroidism, such as tachycardia.

Abrupt withdrawal of beta-blockade may be followed by an exacerbation of the symptoms of hyperthyroidism or may precipitate thyroid storm.

Renal Disease Cumulative effects of the thiazides may develop in patients with impaired renal function.

In such patients, thiazides may precipitate azotemia.

In subjects with creatinine clearance less than 40 mL/min, the plasma half-life of bisoprolol fumarate is increased up to threefold, as compared to healthy subjects.

If progressive renal impairment becomes apparent, ZIAC should be discontinued (See Pharmacokinetics and Metabolism ).

Hepatic Disease ZIAC should be used with caution in patients with impaired hepatic function or progressive liver disease.

Thiazides may alter fluid and electrolyte balance, which may precipitate hepatic coma.

Also, elimination of bisoprolol fumarate is significantly slower in patients with cirrhosis than in healthy subjects (See Pharmacokinetics and Metabolism ).

Acute Angle-Closure Glaucoma with or without Acute Myopia and Choroidal Effusions Hydrochlorothiazide, a sulfonamide, can cause an idiosyncratic reaction, resulting in acute angle-closure glaucoma and elevated intraocular pressure with or without a noticeable acute myopic shift and/or choroidal effusions.

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

Untreated, the acute angle-closure glaucoma may result in permanent visual field loss.

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

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

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

DRUG INTERACTIONS

Drug Interactions ZIAC may potentiate the action of other antihypertensive agents used concomitantly.

ZIAC should not be combined with other beta-blocking agents.

Patients receiving catecholamine-depleting drugs, such as reserpine or guanethidine, should be closely monitored because the added beta-adrenergic blocking action of bisoprolol fumarate may produce excessive reduction of sympathetic activity.

In patients receiving concurrent therapy with clonidine, if therapy is to be discontinued, it is suggested that ZIAC be discontinued for several days before the withdrawal of clonidine.

ZIAC should be used with caution when myocardial depressants or inhibitors of AV conduction, such as certain calcium antagonists (particularly of the phenylalkylamine [verapamil] and benzothiazepine [diltiazem] classes), or antiarrhythmic agents, such as disopyramide, are used concurrently.

Both digitalis glycosides and beta-blockers slow atrioventricular conduction and decrease heart rate.

Concomitant use can increase the risk of bradycardia.

OVERDOSAGE

There are limited data on overdose with ZIAC.

However, several cases of overdose with bisoprolol fumarate have been reported (maximum: 2000 mg).

Bradycardia and/or hypotension were noted.

Sympathomimetic agents were given in some cases, and all patients recovered.

The most frequently observed signs expected with overdosage of a beta-blocker are bradycardia and hypotension.

Lethargy is also common, and with severe overdoses, delirium, coma, convulsions, and respiratory arrest have been reported to occur.

Congestive heart failure, bronchospasm, and hypoglycemia may occur, particularly in patients with underlying conditions.

With thiazide diuretics, acute intoxication is rare.

The most prominent feature of overdose is acute loss of fluid and electrolytes.

Signs and symptoms include cardiovascular (tachycardia, hypotension, shock), neuromuscular (weakness, confusion, dizziness, cramps of the calf muscles, paresthesia, fatigue, impairment of consciousness), gastrointestinal (nausea, vomiting, thirst), renal (polyuria, oliguria, or anuria [due to hemoconcentration]), and laboratory findings (hypokalemia, hyponatremia, hypochloremia, alkalosis, increased BUN [especially in patients with renal insufficiency]).

If overdosage of ZIAC (bisoprolol fumarate and hydrochlorothiazide) is suspected, therapy with ZIAC should be discontinued and the patient observed closely.

Treatment is symptomatic and supportive; there is no specific antidote.

Limited data suggest bisoprolol fumarate is not dialyzable; similarly, there is no indication that hydrochlorothiazide is dialyzable.

Suggested general measures include induction of emesis and/or gastric lavage, administration of activated charcoal, respiratory support, correction of fluid and electrolyte imbalance, and treatment of convulsions.

Based on the expected pharmacologic actions and recommendations for other beta-blockers and hydrochlorothiazide, the following measures should be considered when clinically warranted: Bradycardia Administer IV atropine.

If the response is inadequate, isoproterenol or another agent with positive chronotropic properties may be given cautiously.

Under some circumstances, transvenous pacemaker insertion may be necessary.

Hypotension, Shock The patient’s legs should be elevated.

IV fluids should be administered and lost electrolytes (potassium, sodium) replaced.

Intravenous glucagon may be useful.

Vasopressors should be considered.

Heart Block (second or third degree) Patients should be carefully monitored and treated with isoproterenol infusion or transvenous cardiac pacemaker insertion, as appropriate.

Congestive Heart Failure Initiate conventional therapy (ie, digitalis, diuretics, vasodilating agents, inotropic agents).

Bronchospasm Administer a bronchodilator such as isoproterenol and/or aminophylline.

Hypoglycemia Administer IV glucose.

Surveillance Fluid and electrolyte balance (especially serum potassium) and renal function should be monitored until normalized.

DESCRIPTION

ZIAC ® (bisoprolol fumarate and hydrochlorothiazide) is indicated for the treatment of hypertension.

It combines two antihypertensive agents in a once-daily dosage: a synthetic beta 1 -selective (cardioselective) adrenoceptor blocking agent (bisoprolol fumarate) and a benzothiadiazine diuretic (hydrochlorothiazide).

Bisoprolol fumarate is chemically described as (±)-1-[4-[[2-(1-methylethoxy)ethoxy]methyl]phenoxy]-3-[(1-methylethyl)amino]-2-propanol( E )-2-butenedioate (2:1) (salt).

It possesses an asymmetric carbon atom in its structure and is provided as a racemic mixture.

The S(-) enantiomer is responsible for most of the beta-blocking activity.

Its empirical formula is (C 18 H 31 NO 4 ) 2 •C 4 H 4 O 4 and it has a molecular weight of 766.97.

Its structural formula is: Bisoprolol fumarate is a white crystalline powder, approximately equally hydrophilic and lipophilic, and readily soluble in water, methanol, ethanol, and chloroform.

Hydrochlorothiazide (HCTZ) is 6-Chloro-3,4-dihydro-2 H -1,2,4-benzothiadiazine-7-sulfonamide 1,1-dioxide.

It is a white, or practically white, practically odorless crystalline powder.

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

Its empirical formula is C 7 H 8 ClN 3 O 4 S 2 and it has a molecular weight of 297.73.

Its structural formula is: Each ZIAC ® -2.5 mg/6.25 mg tablet for oral administration contains: Bisoprolol fumarate…………………………………………..2.5 mg Hydrochlorothiazide………………………………………..6.25 mg Each ZIAC ® -5 mg/6.25 mg tablet for oral administration contains: Bisoprolol fumarate……………………………………………5 mg Hydrochlorothiazide………………………………………..6.25 mg Each ZIAC ® -10 mg/6.25 mg tablet for oral administration contains: Bisoprolol fumarate……………………………………………10 mg Hydrochlorothiazide………………………………………..6.25 mg Inactive ingredients include Corn Starch, Dibasic Calcium Phosphate, Hypromellose, Magnesium Stearate, Microcrystalline Cellulose, Polyethylene Glycol, Polysorbate 80, and Titanium Dioxide.

The 10 mg/6.25mg tablet also contains Colloidal Silicon Dioxide.

The 5 mg/6.25 mg tablet also contains Colloidal Silicon Dioxide, and Red and Yellow Iron Oxide.

The 2.5 mg/6.25 mg tablet also contains Crospovidone, Pregelatinized Starch, and Yellow Iron Oxide.

Bisoprolol Fumarate Structural Formula Hydrochlorothiazide Structural Formula

CLINICAL STUDIES

In controlled clinical trials, bisoprolol fumarate/hydrochlorothiazide 6.25 mg has been shown to reduce systolic and diastolic blood pressure throughout a 24-hour period when administered once daily.

The effects on systolic and diastolic blood pressure reduction of the combination of bisoprolol fumarate and hydrochlorothiazide were additive.

Further, treatment effects were consistent across age groups (<60, ≥ 60 years), racial groups (black, nonblack), and gender (male, female).

In two randomized, double-blind, placebo-controlled trials conducted in the U.S., reductions in systolic and diastolic blood pressure and heart rate 24 hours after dosing in patients with mild-to-moderate hypertension are shown below.

In both studies mean systolic/diastolic blood pressure and heart rate at baseline were approximately 151/101 mm Hg and 77 bpm.

Sitting Systolic/Diastolic Pressure (BP) and Heart Rate (HR) Mean Decrease (Δ) After 3-4 Weeks Study 1 Study 2 Placebo B5/H6.25 mg Placebo H6.25 mg B2.5/H6.25 mg B10/H6.25 mg n= 75 150 56 23 28 25 Total ΔBP (mm Hg) -2.9/-3.9 -15.8/-12.6 -3.0/-3.7 -6.6/-5.8 -14.1/-10.5 -15.3/-14.3 Drug Effect a -/- -12.9/-8.7 -/- -3.6/-2.1 -11.1/-6.8 -12.3/-10.6 Total ΔHR (bpm) -0.3 -6.9 -1.6 -0.8 -3.7 -9.8 Drug Effect a – -6.6 – +0.8 -2.1 -8.2 a Observed mean change from baseline minus placebo.

Blood pressure responses were seen within 1 week of treatment but the maximum effect was apparent after 2 to 3 weeks of treatment.

Overall, significantly greater blood pressure reductions were observed on ZIAC than on placebo.

Further, blood pressure reductions were significantly greater for each of the bisoprolol fumarate plus hydrochlorothiazide combinations than for either of the components used alone regardless of race, age, or gender.

There were no significant differences in response between black and nonblack patients.

HOW SUPPLIED

ZIAC ® -2.5 mg/6.25 mg Tablets (bisoprolol fumarate 2.5 mg and hydrochlorothiazide 6.25 mg): Yellow, round, film-coated, unscored tablets.

Debossed with stylized b within an engraved heart shape on one side and 47 on the other side, supplied as follows: Bottle of 100 Tablets NDC 51285-047-02 ZIAC ® -5 mg/6.25 mg Tablets (bisoprolol fumarate 5 mg and hydrochlorothiazide 6.25 mg): Pink, round, film-coated, unscored tablets.

Debossed with stylized b within an engraved heart shape on one side and 50 on the other side, supplied as follows: Bottle of 100 Tablets NDC 51285-050-02 ZIAC ® -10 mg/6.25 mg Tablets (bisoprolol fumarate 10 mg and hydrochlorothiazide 6.25 mg): White, round, film-coated, unscored tablets.

Debossed with stylized b within an engraved heart shape on one side and 40 on the other side, supplied as follows: Bottle of 30 Tablets with child resistant closure NDC 51285-040-01 Store at 20°C to 25°C (68°F to 77°F) [See USP Controlled Room Temperature].

Dispense in a tight container.

Manufactured for: Teva Pharmaceuticals Parsippany, NJ 07054 ZIAC-004 Rev.

12/2023

GERIATRIC USE

Geriatric Use In clinical trials, at least 270 patients treated with bisoprolol fumarate plus HCTZ were 60 years of age or older.

HCTZ added significantly to the antihypertensive effect of bisoprolol in elderly hypertensive patients.

No overall differences in effectiveness or safety were observed between these patients and younger patients.

Other reported clinical experience has not identified differences in responses between the elderly and younger patients, but greater sensitivity of some older individuals cannot be ruled out.

INDICATIONS AND USAGE

ZIAC (bisoprolol fumarate and hydrochlorothiazide) is indicated in the management of hypertension.

PEDIATRIC USE

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

PREGNANCY

Pregnancy Teratogenic Effects ZIAC In rats, the bisoprolol fumarate/hydrochlorothiazide (B/H) combination was not teratogenic at doses up to 51.4 mg/kg/day of bisoprolol fumarate in combination with 128.6 mg/kg/day of hydrochlorothiazide.

Bisoprolol fumarate and hydrochlorothiazide doses used in the rat study are, as multiples of the MRHD in the combination, 129 and 514 times greater, respectively, on a body weight basis, and 26 and 106 times greater, respectively, on the basis of body surface area.

The drug combination was maternotoxic (decreased body weight and food consumption) at B5.7/H14.3 (mg/kg/day) and higher, and fetotoxic (increased late resorptions) at B17.1/H42.9 (mg/kg/day) and higher.

Maternotoxicity was present at 14/57 times the MRHD of B/H, respectively, on a body weight basis, and 3/12 times the MRHD of B/H doses, respectively, on the basis of body surface area.

Fetotoxicity was present at 43/172 times the MRHD of B/H, respectively, on a body weight basis, and 9/35 times the MRHD of B/H doses, respectively, on the basis of body surface area.

In rabbits, the B/H combination was not teratogenic at doses of B10/H25 (mg/kg/day).

Bisoprolol fumarate and hydrochlorothiazide used in the rabbit study were not teratogenic at 25/100 times the B/H MRHD, respectively, on a body weight basis, and 10/40 times the B/H MRHD, respectively, on the basis of body surface area.

The drug combination was maternotoxic (decreased body weight) at B1/H2.5 (mg/kg/day) and higher, and fetotoxic (increased resorptions) at B10/H25 (mg/kg/day).

The multiples of the MRHD for the B/H combination that were maternotoxic are, respectively, 2.5/10 (on the basis of body weight) and 1/4 (on the basis of body surface area), and for fetotoxicity were, respectively 25/100 (on the basis of body weight) and 10/40 (on the basis of body surface area).

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

ZIAC (bisoprolol fumarate and hydrochlorothiazide) should be used during pregnancy only if the potential benefit justifies the risk to the fetus.

Bisoprolol Fumarate In rats, bisoprolol fumarate was not teratogenic at doses up to 150 mg/kg/day, which were 375 and 77 times the MRHD on the basis of body weight and body surface area, respectively.

Bisoprolol fumarate was fetotoxic (increased late resorptions) at 50 mg/kg/day and maternotoxic (decreased food intake and body weight gain) at 150 mg/kg/day.

The fetotoxicity in rats occurred at 125 times the MRHD on a body weight basis and 26 times the MRHD on the basis of body surface area.

The maternotoxicity occurred at 375 times the MRHD on a body weight basis and 77 times the MRHD on the basis of body surface area.

In rabbits, bisoprolol fumarate was not teratogenic at doses up to 12.5 mg/kg/day, which is 31 and 12 times the MRHD based on body weight and body surface area, respectively, but was embryolethal (increased early resorptions) at 12.5 mg/kg/day.

Hydrochlorothiazide Hydrochlorothiazide was orally administered to pregnant mice and rats during respective periods of major organogenesis at doses up to 3000 and 1000 mg/kg/day, respectively.

At these doses, which are multiples of the MRHD equal to 12,000 for mice and 4000 for rats, based on body weight, and equal to 1129 for mice and 824 for rats, based on body surface area, there was no evidence of harm to the fetus.

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

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

Nonteratogenic Effects Thiazides cross the placental barrier and appear in the cord blood.

The use of thiazides in pregnant women requires that the anticipated benefit be weighed against possible hazards to the fetus.

These hazards include fetal or neonatal jaundice, pancreatitis, thrombocytopenia, and possibly other adverse reactions that have occurred in the adult.

NUSRING MOTHERS

Nursing Mothers Bisoprolol fumarate alone or in combination with HCTZ has not been studied in nursing mothers.

Thiazides are excreted in human breast milk.

Small amounts of bisoprolol fumarate (<2% of the dose) have been detected in the milk of lactating rats.

Because of the potential for serious adverse reactions in nursing infants, a decision should be made whether to discontinue nursing or to discontinue the drug, taking into account the importance of the drug to the mother.

INFORMATION FOR PATIENTS

Warn patients, especially those with coronary artery disease, against discontinuing use of ZIAC without a physician’s supervision.

Patients should also be advised to consult a physician if any difficulty in breathing occurs, or if they develop other signs or symptoms of congestive heart failure or excessive bradycardia.

Inform patients or caregivers that there is a risk of hypoglycemia when Ziac is given to patients who are fasting or who are vomiting.

Monitor for symptoms of hypoglycemia.

Patients should know how they react to this medicine before they operate automobiles and machinery or engage in other tasks requiring alertness.

Advise patients that photosensitivity reactions have been reported with thiazides.

Non-melanoma Skin Cancer Instruct patients taking hydrochlorothiazide to protect skin from the sun and undergo regular skin cancer screening.

Acute Angle-Closure Glaucoma Instruct patients taking hydrochlorothiazide to immediately consult their healthcare provider if visual field defects, decreased visual acuity, or ocular pain occur.

DOSAGE AND ADMINISTRATION

Bisoprolol is an effective treatment of hypertension in once-daily doses of 2.5 to 40 mg, while hydrochlorothiazide is effective in doses of 12.5 to 50 mg.

In clinical trials of bisoprolol/hydrochlorothiazide combination therapy using bisoprolol doses of 2.5 to 20 mg and hydrochlorothiazide doses of 6.25 to 25 mg, the antihypertensive effects increased with increasing doses of either component.

The adverse effects (see WARNINGS ) of bisoprolol are a mixture of dose-dependent phenomena (primarily bradycardia, diarrhea, asthenia, and fatigue) and dose-independent phenomena (eg, occasional rash); those of hydrochlorothiazide are a mixture of dose-dependent phenomena (primarily hypokalemia) and dose-independent phenomena (eg, possibly pancreatitis); the dose-dependent phenomena for each being much more common than the dose-independent phenomena.

The latter consist of those few that are truly idiosyncratic in nature or those that occur with such low frequency that a dose relationship may be difficult to discern.

Therapy with a combination of bisoprolol and hydrochlorothiazide will be associated with both sets of dose-independent adverse effects, and to minimize these, it may be appropriate to begin combination therapy only after a patient has failed to achieve the desired effect with monotherapy.

On the other hand, regimens that combine low doses of bisoprolol and hydrochlorothiazide should produce minimal dose-dependent adverse effects, eg, bradycardia, diarrhea, asthenia and fatigue, and minimal dose-dependent adverse metabolic effects, ie, decreases in serum potassium (see CLINICAL PHARMACOLOGY ).

Therapy Guided by Clinical Effect A patient whose blood pressure is not adequately controlled with 2.5-20 mg bisoprolol daily may instead be given ZIAC.

Patients whose blood pressures are adequately controlled with 50 mg of hydrochlorothiazide daily, but who experience significant potassium loss with this regimen, may achieve similar blood pressure control without electrolyte disturbance if they are switched to ZIAC.

Initial Therapy Antihypertensive therapy may be initiated with the lowest dose of ZIAC, one 2.5/6.25 mg tablet once daily.

Subsequent titration (14-day intervals) may be carried out with ZIAC tablets up to the maximum recommended dose 20/12.5 mg (two 10/6.25 mg tablets) once daily, as appropriate.

Replacement Therapy The combination may be substituted for the titrated individual components.

Cessation of Therapy If withdrawal of ZIAC therapy is planned, it should be achieved gradually over a period of about 2 weeks.

Patients should be carefully observed.

Patients with Renal or Hepatic Impairment: As noted in the WARNINGS section, caution must be used in dosing/titrating patients with hepatic impairment or renal dysfunction.

Since there is no indication that hydrochlorothiazide is dialyzable, and limited data suggest that bisoprolol is not dialyzable, drug replacement is not necessary in patients undergoing dialysis.

Geriatric Patients: Dosage adjustment on the basis of age is not usually necessary, unless there is also significant renal or hepatic dysfunction (see above and WARNINGS section).

Pediatric Patients: There is no pediatric experience with ZIAC.

Dextromethorphan 10 MG Oral Lozenge

WARNINGS

Warnings Do not use if taking a prescription monoamine oxidase inhibitor (MAOI) (certain drugs for depression, psychiatric, or emotional conditions, or Parkinson’s disease), or for 2 weeks after stopping the MAOI drug.

If you do not know if your prescription drug contains an MAOI, ask a doctor or pharmacist before taking this product.

Ask a doctor before use if you have chronic cough that lasts or occurs with smoking, asthma, chronic bronchitis or emphysema cough that occurs with too much phlegm (mucus) Stop use and ask a doctor if cough lasts more than 7 days, comes back, or occurs with fever, rash or headache that lasts.

These could be signs of a serious condition.

If pregnant or breast-feeding, ask a health professional before use.

Keep out of reach of children.

In case of overdose, get medical help or contact a Poison Control Center right away.

INDICATIONS AND USAGE

Uses temporarily suppresses coughs due to minor throat and bronchial irritation associated with a cold or inhaled irritants

INACTIVE INGREDIENTS

Inactive ingredients Corn Syrup, FD&C Red 40, FD&C Blue 1, Flavors, Menthol, Purified Water, Sucrose, Titanium Dioxide.

PURPOSE

Purpose Cough Suppressant

KEEP OUT OF REACH OF CHILDREN

Keep out of reach of children.

In case of overdose, get medical help or contact a Poison Control Center right away.

ASK DOCTOR

Ask a doctor before use if you have chronic cough that lasts or occurs with smoking, asthma, chronic bronchitis or emphysema cough that occurs with too much phlegm (mucus)

DOSAGE AND ADMINISTRATION

Directions adults and children 12 years of age and over: take 3 lozenges every 6-8 hours, do not take more than 12 lozenges per day children 6 to under 12 years of age: take 1 lozenge every 4 hours, do not take more than 6 lozenges per day children 4 to under 6 years of age: ask a doctor do not use in children under 4 years of age

PREGNANCY AND BREAST FEEDING

If pregnant or breast-feeding, ask a health professional before use.

DO NOT USE

Do not use if taking a prescription monoamine oxidase inhibitor (MAOI) (certain drugs for depression, psychiatric, or emotional conditions, or Parkinson’s disease), or for 2 weeks after stopping the MAOI drug.

If you do not know if your prescription drug contains an MAOI, ask a doctor or pharmacist before taking this product.

STOP USE

Stop use and ask a doctor if cough lasts more than 7 days, comes back, or occurs with fever, rash or headache that lasts.

These could be signs of a serious condition.

ACTIVE INGREDIENTS

Active ingredient (per lozenge) Dextromethorphan Hydrobromide 10mg

Sudafed PE Children’s Cold & Cough 5 MG / 2.5 MG per 5 ML Oral Solution

Generic Name: DEXTROMETHORPHAN HYDROBROMIDE AND PHENYLEPHRINE HYDROCHLORIDE
Brand Name: Childrens SUDAFED PE Cold plus Cough
  • Substance Name(s):
  • DEXTROMETHORPHAN HYDROBROMIDE
  • PHENYLEPHRINE HYDROCHLORIDE

WARNINGS

Warnings Do not use in a child who is taking a prescription monoamine oxidase inhibitor (MAOI) (certain drugs for depression, psychiatric or emotional conditions, or Parkinson’s disease), or for 2 weeks after stopping the MAOI drug.

If you do not know if your child’s prescription drug contains an MAOI, ask a doctor or pharmacist before giving this product.

Ask a doctor before use if the child has heart disease high blood pressure thyroid disease diabetes persistent or chronic cough such as occurs with asthma cough that occurs with too much phlegm (mucus) a sodium-restricted diet When using this product do not exceed recommended dose Stop use and ask a doctor if nervousness, dizziness, or sleeplessness occur symptoms do not improve within 7 days or occur with a fever cough gets worse or lasts for more than 7 days cough tends to come back or occurs with fever, rash or headache that lasts These could be signs of a serious condition.

Keep out of reach of children.

In case of overdose, get medical help or contact a Poison Control Center right away.

(1-800-222-1222)

INDICATIONS AND USAGE

Uses temporarily relieves these symptoms due to the common cold, hay fever, or other upper respiratory allergies: cough nasal congestion sinus congestion and pressure

INACTIVE INGREDIENTS

Inactive ingredients anhydrous citric acid, carboxymethylcellulose sodium, edetate disodium, FD&C blue no.

1, FD&C red no.

40, flavors, glycerin, purified water, sodium benzoate, sodium citrate, sorbitol solution, sucralose

PURPOSE

Active ingredients (in each 5 mL) Purposes Dextromethorphan HBr 5 mg Cough suppressant Phenylephrine HCl 2.5 mg Nasal decongestant

KEEP OUT OF REACH OF CHILDREN

Keep out of reach of children.

In case of overdose, get medical help or contact a Poison Control Center right away.

(1-800-222-1222)

ASK DOCTOR

Ask a doctor before use if the child has heart disease high blood pressure thyroid disease diabetes persistent or chronic cough such as occurs with asthma cough that occurs with too much phlegm (mucus) a sodium-restricted diet

DOSAGE AND ADMINISTRATION

Directions find right dose on chart below mL = milliliters repeat dose every 4 hours do not give more than 6 times in 24 hours Age (yr) Dose (mL) under 4 years do not use 4 to 5 years 5 mL 6 to 11 years 10 mL Attention: use only enclosed dosing cup specifically designed for use with this product.

Do not use any other dosing device.

DO NOT USE

Do not use in a child who is taking a prescription monoamine oxidase inhibitor (MAOI) (certain drugs for depression, psychiatric or emotional conditions, or Parkinson’s disease), or for 2 weeks after stopping the MAOI drug.

If you do not know if your child’s prescription drug contains an MAOI, ask a doctor or pharmacist before giving this product.

STOP USE

Stop use and ask a doctor if nervousness, dizziness, or sleeplessness occur symptoms do not improve within 7 days or occur with a fever cough gets worse or lasts for more than 7 days cough tends to come back or occurs with fever, rash or headache that lasts These could be signs of a serious condition.

ACTIVE INGREDIENTS

Active ingredients (in each 5 mL) Purposes Dextromethorphan HBr 5 mg Cough suppressant Phenylephrine HCl 2.5 mg Nasal decongestant

cefTRIAXone 2 GM Injection

Generic Name: CEFTRIAXONE SODIUM
Brand Name: Ceftriaxone Sodium
  • Substance Name(s):
  • CEFTRIAXONE SODIUM

WARNINGS

Hypersensitivity Reactions Before therapy with ceftriaxone for injection is instituted, careful inquiry should be made to determine whether the patient has had previous hypersensitivity reactions to cephalosporins, penicillins and other beta-lactam agents or other drugs.

This product should be given cautiously to penicillin and other beta-lactam agent-sensitive patients.

Antibacterial drugs should be administered with caution to any patient who has demonstrated some form of allergy, particularly to drugs.

Serious acute hypersensitivity reactions may require the use of subcutaneous epinephrine and other emergency measures.

As with all beta-lactam antibacterial agents, serious and occasionally fatal hypersensitivity reactions (i.e., anaphylaxis) have been reported.

In case of severe hypersensitivity reactions, treatment with ceftriaxone must be discontinued immediately and adequate emergency measures must be initiated.

Interaction with Calcium-Containing Products Do not use diluents containing calcium, such as Ringer’s solution or Hartmann’s solution, to reconstitute ceftriaxone vials or to further dilute a reconstituted vial for IV administration because a precipitate can form.

Precipitation of ceftriaxone-calcium can also occur when ceftriaxone is mixed with calcium-containing solutions in the same IV administration line.

Ceftriaxone must not be administered simultaneously with calcium-containing IV solutions, including continuous calcium-containing infusions such as parenteral nutrition via a Y-site.

However, in patients other than neonates, ceftriaxone and calcium-containing solutions may be administered sequentially of one another if the infusion lines are thoroughly flushed between infusions with a compatible fluid.

In vitro studies using adult and neonatal plasma from umbilical cord blood demonstrated that neonates have an increased risk of precipitation of ceftriaxone-calcium (see CLINICAL PHARMACOLOGY , CONTRAINDICATIONS and DOSAGE AND ADMINISTRATION ).

Neurological Adverse Reactions Serious neurological adverse reactions have been reported during postmarketing surveillance with ceftriaxone use.

These reactions include encephalopathy (disturbance of consciousness including somnolence, lethargy, and confusion), seizures, myoclonus, and non-convulsive status epilepticus (see ADVERSE REACTIONS ).

Some cases occurred in patients with severe renal impairment who did not receive appropriate dosage adjustment.

However, in other cases, neurological adverse reactions occurred in patients receiving an appropriate dosage adjustment.

The neurological adverse reactions were reversible and resolved after discontinuation.

If neurological adverse reactions associated with ceftriaxone for injection therapy occur, discontinue ceftriaxone for injection and institute appropriate supportive measures.

Make appropriate dosage adjustments in patients with severe renal impairment (see DOSAGE AND ADMINISTRATION ).

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

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

difficile .

C.

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

Hypertoxin producing strains of C.

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

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

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

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

difficile may need to be discontinued.

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

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

Hemolytic Anemia An immune mediated hemolytic anemia has been observed in patients receiving cephalosporin class antibacterials including ceftriaxone.

Severe cases of hemolytic anemia, including fatalities, have been reported during treatment in both adults and children.

If a patient develops anemia while on ceftriaxone, the diagnosis of a cephalosporin associated anemia should be considered and ceftriaxone stopped until the etiology is determined.

OVERDOSAGE

Ceftriaxone overdosage has been reported in patients with severe renal impairment.

Reactions have included neurological outcomes, including encephalopathy, seizures, myoclonus, and non-convulsive status epilepticus.

In the event of overdosage, discontinue ceftriaxone for injection therapy and provide general supportive treatment (see DOSAGE AND ADMINISTRATION , WARNINGS and PRECAUTIONS ).

In the case of overdosage, drug concentration would not be reduced by hemodialysis or peritoneal dialysis.

There is no specific antidote.

Treatment of overdosage should be symptomatic.

DESCRIPTION

Ceftriaxone for injection, USP is a sterile, semisynthetic, broad-spectrum cephalosporin antibiotic for intravenous or intramuscular administration.

Ceftriaxone sodium is (6 R , 7 R )-7-[2-(2-Amino-4-thiazolyl) glyoxylamido]-8-oxo-3-[[(1,2,5,6-tetrahydro-2-methyl-5,6-dioxo- as -triazin-3-yl)thio]methyl]-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid, 7 2 -( Z )-( O -methyloxime), disodium salt, sesquaterhydrate.

The chemical formula of ceftriaxone sodium is C 18 H 16 N 8 Na 2 O 7 S 3 •3.5H 2 O.

It has a calculated molecular weight of 661.60 and the following structural formula: Ceftriaxone sodium is a white to yellowish crystalline powder which is readily soluble in water, sparingly soluble in methanol and very slightly soluble in ethanol.

The pH of a 1% aqueous solution is approximately 6.7.

The color of ceftriaxone sodium solutions ranges from light yellow to amber, depending on the length of storage, concentration and diluent used.

Each vial contains ceftriaxone sodium equivalent to 250 mg, 500 mg, 1 gram or 2 grams of ceftriaxone activity.

Ceftriaxone sodium contains approximately 83 mg (3.6 mEq) of sodium per gram of ceftriaxone activity.

ceftriaxone-chemical-structure

CLINICAL STUDIES

Clinical Trials in Pediatric Patients with Acute Bacterial Otitis Media In two adequate and well-controlled US clinical trials a single IM dose of ceftriaxone was compared with a 10 day course of oral antibiotic in pediatric patients between the ages of 3 months and 6 years.

The clinical cure rates and statistical outcome appear in the table below: Table 5.

Clinical Efficacy in Pediatric Patients with Acute Bacterial Otitis Media Clinical Efficacy in Evaluable Population Study Day Ceftriaxone Single-Dose Comparator- 10 Days of Oral Therapy 95% Confidence Interval Statistical Outcome Study 1 – US amoxicillin/ clavulanate Ceftriaxone is lower than control at study day 14 and 28.

14 74% (220/296) 82% (247/302) (-14.4%, -0.5%) 28 58% (167/288) 67% (200/297) (-17.5%, -1.2%) Study 2 – US 1 TMP-SMZ Ceftriaxone is equivalent to control at study day 14 and 28.

14 54% (113/210) 60% (124/206) (-16.4%, 3.6%) 28 35% (73/206) 45% (93/205) (-19.9%, 0.0%) An open-label bacteriologic study of ceftriaxone without a comparator enrolled 108 pediatric patients, 79 of whom had positive baseline cultures for one or more of the common pathogens.

The results of this study are tabulated as follows: Week 2 and 4 Bacteriologic Eradication Rates in the Per Protocol Analysis in the Roche Bacteriologic Study by pathogen: Table 6.

Bacteriologic Eradication Rates by Pathogen Organism Study Day 13 to 15 Study Day 30+2 No.

Analyzed No.

Erad.

(%) No.

Analyzed No.

Erad.

(%) Streptococcus pneumoniae 38 32 (84) 35 25 (71) Haemophilus influenzae 33 28 (85) 31 22 (71) Moraxella catarrhalis 15 12 (80) 15 9 (60)

HOW SUPPLIED

Ceftriaxone for injection, USP is supplied as a sterile crystalline powder in glass vials as follows: NDC Vials containing 250 mg equivalent to ceftriaxone.

Package of 10 0409-7337-01 Vials containing 500 mg equivalent to ceftriaxone.

Package of 10 0409-7338-01 Vials containing 1 g equivalent to ceftriaxone.

Package of 10 0409-7332-01 Vials containing 2 g equivalent to ceftriaxone.

Package of 10 0409-7335-03 Storage Prior to Reconstitution Store at 20° to 25°C (68° to 77°F) [see USP Controlled Room Temperature].

Protect from light.

GERIATRIC USE

Geriatric Use Of the total number of subjects in clinical studies of ceftriaxone, 32% were 60 and over.

No overall differences in safety or effectiveness were observed between these subjects and younger subjects, and other reported clinical experience has not identified differences in responses between the elderly and younger patients, but greater sensitivity of some older individuals cannot be ruled out.

The pharmacokinetics of ceftriaxone were only minimally altered in geriatric patients compared to healthy adult subjects and dosage adjustments are not necessary for geriatric patients with ceftriaxone dosages up to 2 grams per day provided there is no severe renal and hepatic impairment (see CLINICAL PHARMACOLOGY ).

INDICATIONS AND USAGE

Before instituting treatment with ceftriaxone, appropriate specimens should be obtained for isolation of the causative organism and for determination of its susceptibility to the drug.

Therapy may be instituted prior to obtaining results of susceptibility testing.

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

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

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

Ceftriaxone for injection, USP is indicated for the treatment of the following infections when caused by susceptible organisms: Lower Respiratory Tract Infections Caused by Streptococcus pneumoniae , Staphylococcus aureus, Haemophilus influenzae, Haemophilus parainfluenzae, Klebsiella pneumoniae, Escherichia coli, Enterobacter aerogenes, Proteus mirabilis or Serratia marcescens .

Acute Bacterial Otitis Media Caused by Streptococcus pneumoniae, Haemophilus influenzae (including beta-lactamase producing strains) or Moraxella catarrhalis (including beta-lactamase producing strains).

NOTE: In one study lower clinical cure rates were observed with a single dose of ceftriaxone compared to 10 days of oral therapy.

In a second study comparable cure rates were observed between single dose ceftriaxone and the comparator.

The potentially lower clinical cure rate of ceftriaxone should be balanced against the potential advantages of parenteral therapy (see CLINICAL STUDIES ).

Skin and Skin Structure Infections Caused by Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes , Viridans group streptococci, Escherichia coli, Enterobacter cloacae, Klebsiella oxytoca, Klebsiella pneumoniae, Proteus mirabilis, Morganella morganii*, Pseudomonas aeruginosa, Serratia marcescens, Acinetobacter calcoaceticus, Bacteroides fragilis * or Peptostreptococcus species.

Urinary Tract Infections (complicated and uncomplicated) Caused by Escherichia coli, Proteus mirabilis, Proteus vulgaris, Morganella morganii or Klebsiella pneumoniae .

Uncomplicated Gonorrhea (cervical/urethral and rectal) Caused by Neisseria gonorrhoeae , including both penicillinase- and nonpenicillinase-producing strains, and pharyngeal gonorrhea caused by nonpenicillinase-producing strains of Neisseria gonorrhoeae .

Pelvic Inflammatory Disease Caused by Neisseria gonorrhoeae .

Ceftriaxone sodium, like other cephalosporins, has no activity against Chlamydia trachomatis .

Therefore, when cephalosporins are used in the treatment of patients with pelvic inflammatory disease and Chlamydia trachomatis is one of the suspected pathogens, appropriate antichlamydial coverage should be added.

Bacterial Septicemia Caused by Staphylococcus aureus, Streptococcus pneumoniae, Escherichia coli, Haemophilus influenzae or Klebsiella pneumoniae .

Bone and Joint Infections Caused by Staphylococcus aureus, Streptococcus pneumoniae, Escherichia coli, Proteus mirabilis, Klebsiella pneumoniae or Enterobacter species.

Intra-abdominal Infections Caused by Escherichia coli, Klebsiella pneumoniae, Bacteroides fragilis, Clostridium species (Note: most strains of Clostridium difficile are resistant) or Peptostreptococcus species.

Meningitis Caused by Haemophilus influenzae, Neisseria meningitidis or Streptococcus pneumoniae .

Ceftriaxone has also been used successfully in a limited number of cases of meningitis and shunt infection caused by Staphylococcus epidermidis * and Escherichia coli* .

*Efficacy for this organism in this organ system was studied in fewer than ten infections.

Surgical Prophylaxis The preoperative administration of a single 1 g dose of ceftriaxone may reduce the incidence of postoperative infections in patients undergoing surgical procedures classified as contaminated or potentially contaminated (e.g., vaginal or abdominal hysterectomy or cholecystectomy for chronic calculous cholecystitis in high-risk patients, such as those over 70 years of age, with acute cholecystitis not requiring therapeutic antimicrobials, obstructive jaundice or common duct bile stones) and in surgical patients for whom infection at the operative site would present serious risk (e.g., during coronary artery bypass surgery).

Although ceftriaxone has been shown to have been as effective as cefazolin in the prevention of infection following coronary artery bypass surgery, no placebo-controlled trials have been conducted to evaluate any cephalosporin antibiotic in the prevention of infection following coronary artery bypass surgery.

When administered prior to surgical procedures for which it is indicated, a single 1 g dose of ceftriaxone provides protection from most infections due to susceptible organisms throughout the course of the procedure.

PEDIATRIC USE

Pediatric Use Safety and effectiveness of ceftriaxone in neonates, infants and pediatric patients have been established for the dosages described in the DOSAGE AND ADMINISTRATION section.

In vitro studies have shown that ceftriaxone, like some other cephalosporins, can displace bilirubin from serum albumin.

Ceftriaxone should not be administered to hyperbilirubinemic neonates, especially prematures (see CONTRAINDICATIONS ).

PREGNANCY

Pregnancy Teratogenic Effects Reproductive studies have been performed in mice and rats at doses up to 20 times the usual human dose and have no evidence of embryotoxicity, fetotoxicity or teratogenicity.

In primates, no embryotoxicity or teratogenicity was demonstrated at a dose approximately 3 times the human dose.

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

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

Nonteratogenic Effects In rats, in the Segment I (fertility and general reproduction) and Segment III (perinatal and postnatal) studies with intravenously administered ceftriaxone, no adverse effects were noted on various reproductive parameters during gestation and lactation, including postnatal growth, functional behavior and reproductive ability of the offspring, at doses of 586 mg/kg/day or less.

NUSRING MOTHERS

Nursing Mothers Low concentrations of ceftriaxone are excreted in human milk.

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

INFORMATION FOR PATIENTS

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

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

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

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

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

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

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

DOSAGE AND ADMINISTRATION

Ceftriaxone may be administered intravenously or intramuscularly.

Do not use diluents containing calcium, such as Ringer’s solution or Hartmann’s solution, to reconstitute ceftriaxone vials or to further dilute a reconstituted vial for IV administration because a precipitate can form.

Precipitation of ceftriaxone-calcium can also occur when ceftriaxone is mixed with calcium-containing solutions in the same IV administration line.

Ceftriaxone must not be administered simultaneously with calcium-containing IV solutions, including continuous calcium-containing infusions such as parenteral nutrition via a Y-site.

However, in patients other than neonates, ceftriaxone and calcium-containing solutions may be administered sequentially of one another if the infusion lines are thoroughly flushed between infusions with a compatible fluid (see WARNINGS ).

There have been no reports of an interaction between ceftriaxone and oral calcium-containing products or interaction between intramuscular ceftriaxone and calcium-containing products (IV or oral).

Neonates Hyperbilirubinemic neonates, especially prematures, should not be treated with ceftriaxone for injection.

Ceftriaxone is contraindicated in premature neonates (see CONTRAINDICATIONS ).

Ceftriaxone is contraindicated in neonates (≤ 28 days) if they require (or are expected to require) treatment with calcium-containing IV solutions, including continuous calcium-containing infusions such as parenteral nutrition because of the risk of precipitation of ceftriaxone-calcium (see CONTRAINDICATIONS ).

Intravenous doses should be given over 60 minutes in neonates to reduce the risk of bilirubin encephalopathy.

Pediatric Patients For the treatment of skin and skin structure infections, the recommended total daily dose is 50 to 75 mg/kg given once a day (or in equally divided doses twice a day).

The total daily dose should not exceed 2 grams.

For the treatment of acute bacterial otitis media, a single intramuscular dose of 50 mg/kg (not to exceed 1 gram) is recommended (see INDICATIONS AND USAGE ).

For the treatment of serious miscellaneous infections other than meningitis, the recommended total daily dose is 50 to 75 mg/kg, given in divided doses every 12 hours.

The total daily dose should not exceed 2 grams.

In the treatment of meningitis, it is recommended that the initial therapeutic dose be 100 mg/kg (not to exceed 4 grams).

Thereafter, a total daily dose of 100 mg/kg/day (not to exceed 4 grams daily) is recommended.

The daily dose may be administered once a day (or in equally divided doses every 12 hours).

The usual duration of therapy is 7 to 14 days.

Adults The usual adult daily dose is 1 to 2 grams given once a day (or in equally divided doses twice a day) depending on the type and severity of infection.

The total daily dose should not exceed 4 grams.

If Chlamydia trachomatis is a suspected pathogen, appropriate antichlamydial coverage should be added, because ceftriaxone sodium has no activity against this organism.

For the treatment of uncomplicated gonococcal infections, a single intramuscular dose of 250 mg is recommended.

For preoperative use (surgical prophylaxis), a single dose of 1 gram administered intravenously 1/2 to 2 hours before surgery is recommended.

Generally, ceftriaxone therapy should be continued for at least 2 days after the signs and symptoms of infection have disappeared.

The usual duration of therapy is 4 to 14 days; in complicated infections, longer therapy may be required.

When treating infections caused by Streptococcus pyogenes , therapy should be continued for at least 10 days.

No dosage adjustment is necessary for patients with impairment of renal or hepatic function (see PRECAUTIONS ).

The dosages recommended for adults require no modification in elderly patients, up to 2 g per day, provided there is no severe renal and hepatic impairment (see PRECAUTIONS ).

Directions for Use Intramuscular Administration Reconstitute ceftriaxone sodium powder with the appropriate diluent (see : Compatibility and Stability ).

Inject diluent into vial, shake vial thoroughly to form solution.

Withdraw entire contents of vial into syringe to equal total labeled dose.

After reconstitution, each 1 mL of solution contains approximately 250 mg or 350 mg equivalent of ceftriaxone according to the amount of diluent indicated below.

If required, more dilute solutions could be utilized.

A 350 mg/mL concentration is not recommended for the 250 mg vial since it may not be possible to withdraw the entire contents.

As with all intramuscular preparations, ceftriaxone should be injected well within the body of a relatively large muscle; aspiration helps to avoid unintentional injection into a blood vessel.

Vial Dosage Size Amount of Diluent to be Added 250 mg/mL 350 mg/mL 250 mg 0.9 mL – 500 mg 1.8 mL 1 mL 1 g 3.6 mL 2.1 mL 2 g 7.2 mL 4.2 mL Intravenous Administration Ceftriaxone should be administered intravenously by infusion over a period of 30 minutes, except in neonates where administration over 60 minutes is recommended to reduce the risk of bilirubin encephalopathy.

Concentrations between 10 mg/mL and 40 mg/mL are recommended; however, lower concentrations may be used if desired.

Reconstitute vials with an appropriate IV diluent (see : Compatibility and Stability ).

Vial Dosage Size Amount of Diluent to be Added 250 mg 2.4 mL 500 mg 4.8 mL 1 g 9.6 mL 2 g 19.2 mL After reconstitution, each 1 mL of solution contains approximately 100 mg equivalent of ceftriaxone.

Withdraw entire contents and dilute to the desired concentration with the appropriate IV diluent.

Compatibility and Stability Do not use diluents containing calcium, such as Ringer’s solution or Hartmann’s solution, to reconstitute ceftriaxone for injection vials or to further dilute a reconstituted vial for IV administration.

Particulate formation can result.

Ceftriaxone has been shown to be compatible with Flagyl® IV (metronidazole hydrochloride).

The concentration should not exceed 5 to 7.5 mg/mL metronidazole hydrochloride with ceftriaxone 10 mg/mL as an admixture.

The admixture is stable for 24 hours at room temperature only in 0.9% sodium chloride injection or 5% dextrose in water (D5W).

No compatibility studies have been conducted with the Flagyl® IV RTU® (metronidazole) formulation or using other diluents.

Metronidazole at concentrations greater than 8 mg/mL will precipitate.

Do not refrigerate the admixture as precipitation will occur.

Vancomycin, amsacrine, aminoglycosides, and fluconazole are incompatible with ceftriaxone in admixtures.

When any of these drugs are to be administered concomitantly with ceftriaxone by intermittent intravenous infusion, it is recommended that they be given sequentially, with thorough flushing of the intravenous lines (with one of the compatible fluids) between the administrations.

Ceftriaxone for injection solutions should not be physically mixed with or piggybacked into solutions containing other antimicrobial drugs or into diluent solutions other than those listed above, due to possible incompatibility (see WARNINGS ).

Ceftriaxone sodium sterile powder should be stored at 20° to 25°C (68° to 77°F) [see USP Controlled Room Temperature] and protected from light.

After reconstitution, protection from normal light is not necessary.

The color of solutions ranges from light yellow to amber, depending on the length of storage, concentration and diluent used.

Ceftriaxone intramuscular solutions remain stable (loss of potency less than 10%) for the following time periods: Diluent Concentration Storage mg/mL Room Temperature (25°C) Refrigerated (4°C) Sterile Water for Injection 100 250, 350 2 days 24 hours 10 days 3 days 0.9% Sodium Chloride Solution 100 250, 350 2 days 24 hours 10 days 3 days 5% Dextrose Solution 100 250, 350 2 days 24 hours 10 days 3 days Bacteriostatic Water + 0.9% Benzyl Alcohol 100 250, 350 24 hours 24 hours 10 days 3 days 1% Lidocaine Solution (without epinephrine) 100 250, 350 24 hours 24 hours 10 days 3 days Ceftriaxone intravenous solutions, at concentrations of 10, 20 and 40 mg/mL, remain stable (loss of potency less than 10%) for the following time periods stored in glass or PVC containers: Diluent Storage Room Temperature (25°C) Refrigerated (4°C) Sterile Water 2 days 10 days 0.9% Sodium Chloride Solution 2 days 10 days 5% Dextrose Solution 2 days 10 days 10% Dextrose Solution 2 days 10 days 5% Dextrose + 0.9% Sodium Chloride Solution 2 days Incompatible 5% Dextrose + 0.45% Sodium Chloride Solution 2 days Incompatible * Data available for 10 to 40 mg/mL concentrations in this diluent in PVC containers only.

The following intravenous ceftriaxone solutions are stable at room temperature (25°C) for 24 hours, at concentrations between 10 mg/mL and 40 mg/mL: Sodium Lactate (PVC container), 10% Invert Sugar (glass container), 5% Sodium Bicarbonate (glass container), Freamine III (glass container), Normosol-M in 5% Dextrose (glass and PVC containers), Ionosol-B in 5% Dextrose (glass container), 5% Mannitol (glass container), 10% Mannitol (glass container).

After the indicated stability time periods, unused portions of solutions should be discarded.

NOTE: Parenteral drug products should be inspected visually for particulate matter before administration.

Ceftriaxone reconstituted with 5% Dextrose or 0.9% Sodium Chloride solution at concentrations between 10 mg/mL and 40 mg/mL, and then stored in frozen state (-20°C) in PVC or polyolefin containers, remains stable for 26 weeks.

Frozen solutions of ceftriaxone for injection should be thawed at room temperature before use.

After thawing, unused portions should be discarded.

DO NOT REFREEZE .

theophylline 100 MG per 18.75 ML Oral Solution

WARNINGS

Concurrent Illness: Theophylline should be used with extreme caution in patients with the following clinical conditions due to the increased risk of exacerbation of the concurrent condition: Active peptic ulcer disease Seizure disorders Cardiac arrhythmias (not including bradyarrhythmias) Conditions That Reduce Theophylline Clearance: There are several readily identifiable causes of reduced theophylline clearance.

If the total daily dose is not approximately reduced in the presence of these risk factors, sever e and potentially fatal theophylline toxicity can occur .

Careful consideration must be given to the benefits and risks of theophylline use and the need for more intensive monitoring of serum theophylline concentrations in patients with the following risk factors: Age Neonates (term and premature) Children 60 years) Concurrent Diseases Acute pulmonary edema Congestive heart failure Cor pulmonale Fever; ≥102°F for 24 hours or more; or lesser temperature elevations for longer periods Hypothyroidism Liver disease; cirrhosis, acute hepatitis Reduced renal function in infants <3 months of age Sepsis with multi-organ failure Shock Cessation of Smoking Drug Interactions Adding a drug that inhibits theophylline metabolism (e.g., cimetidine, erythromycin, tacrine) or stopping a concurrently administered drug that enhances theophylline metabolism (e.g., carbamazepine, rifampin).

(see PRECAUTIONS, Drug Interactions , Table II ).

When Signs or Symptoms of Theophylline Toxicity Are Present: Whenever a patient receiving theophylline develops nausea or vomiting, particularly repetitive vomiting or other signs or symptoms consistent with theophylline toxicity (even if another cause may be suspected), additional doses of theophylline should be withheld and a serum theophylline concentration measured immediately .

Patients should be instructed not to continue any dosage that causes adverse effects and to withhold subsequent doses until the symptoms have resolved, at which time and clinician may instruct the patient to resume the drug at a lower dosage (see DOSAGE AND ADMINISTRATION, Dosing Guidelines , Table VI ).

Dosage Increases: Increases in the dose of theophylline should not be made in response to an acute exacerbation of symptoms of chronic lung disease since theophylline provides little added benefit to inhaled beta2-selective agonists and systemically administered corticosteroids in this circumstance and increases the risk of adverse effects.

A peak steady state serum theophylline concentration should be measured before increasing the dose in response to persistent chronic symptoms to ascertain whether an increase in dose is safe.

Before increasing the theophylline dose on the basis of a low serum concentration, the clinician should consider whether the blood sample was obtained at an appropriate time in relationship to the dose and whether the patient has adhered to the prescribed regimen (see PRECAUTIONS, Laboratory Tests ).

As the rate of theophylline clearance may be dose-dependent (i.e., steady-state serum concentrations may increase disproportionately to the increase in dose), an increase in dose based upon a sub-therapeutic serum concentration measurement should be conservative.

In general, limiting dose increases to about 25% of the previous total daily dose will reduce the risk of unintended excessive increases in serum theophylline concentration (see DOSAGE AND ADMINISTRATION , Table VI ).

DRUG INTERACTIONS

Drug Interactions: Theophylline interacts with a wide variety of drugs.

The interaction may be pharmacodynamic, i.e., alterations in the therapeutic response to theophylline or another drug or occurrence of adverse effects without a change in serum theophylline concentration.

More frequently, however, the interaction is pharmacokinetic, i.e., the rate of theophylline clearance is altered by another drug resulting in increased or decreased serum theophylline concentrations.

Theophylline only rarely alters the pharmacokinetics of other drugs.

The drugs listed in Table II have the potential to produce clinically significant pharmacodynamics or pharmacokinetic interactions with theophylline.

The information in the “Effect” column of Table II assumes that the interacting drug is being added to a steady-state theophylline regimen.

If theophylline is being initiated in a patient who is already taking a drug that inhibits theophylline clearance (e.g., cimetidine, erythromycin), the dose of theophylline required to achieve a therapeutic serum theophylline concentration will be smaller.

Conversely, if theophylline is being initiated in a patient who is already taking a drug that enhances theophylline clearance (e.g., rifampin), the dose of theophylline required to achieve a therapeutic serum theophylline concentration will be larger.

Discontinuation of a concomitant drug that increases theophylline clearance will result in accumulation of theophylline to potentially toxic levels, unless the theophylline dose is appropriately reduced.

Discontinuation of a concomitant drug that inhibits theophylline clearance will result in decreased serum theophylline concentrations, unless the theophylline dose is appropriately increased.

The drugs listed in Table III have either been documented not to interact with theophylline or do not produce a clinically significant interaction (i.e., <15% change in theophylline clearance).

The listing of drugs in Table II and III are current as of February 9, 1995.

New interactions are continuously being reported for theophylline, especially with new chemical entities.

The clinician should not assume that a drug does not interact with theophylline if it is not listed in Table II .

Before addition of a newly available drug in a patient receiving theophylline, the package insert of the new drug and/or the medical literature should be consulted to determine if an interaction between the new drug and theophylline has been reported.

Table II.

Clinically significant drug interactions with theophylline*.

Drug Type of Interaction Effect** Adenosine Theophylline blocks adenosine receptors.

Higher doses of adenosine may be required to achieve desired effect.

Alcohol A single large dose of alcohol (3 ml/kg of whiskey) decreases theophylline clearance for up to 24 hours.

30% increase Allopurinol Decreases theophylline clearance at allopurinol doses ≥ 600 mg/day.

25% increase Amino glutethimide Increases theophylline clearance by induction of microsomal enzyme activity.

25% increase Carbamazepine Similar to aminoglutethimide 30% increase Cimetidine Decreases theophylline clearance by inhibiting cytochrome P450 1A2.

70% increase Ciprofloxacin Similar to cimetidine.

40% increase Clarithromycin Similar to erythromycin.

25% increase Diazepam Benzodiazepines increase CNS concentrations of adenosine, a potent CNS depressant, while theophylline blocks adenosine receptors.

Larger diazepam doses may be required to produce desired level of sedation.

Discontinuation of theophylline without reduction of diazepam dose may result in respiratory depression.

Disulfiram Decreases theophylline clearance by inhibiting hydroxylation and demethylation.

50% increase Enoxacin Similar to cimetidine.

300% increase Ephedrine Synergistic CNS effect Increased frequency of nausea, nervousness, and insomnia.

Erythromycin Erythromycin metabolite decreases theophylline clearance by inhibiting cytochrome P450 3A3 35% increase.

Erythromycin steady-state serum concentrations decrease by a similar amount.

Estrogen Estrogen containing oral contraceptives decrease theophylline clearance in a dose-dependent fashion.

The effect of progesterone on theophylline clearance is unknown.

30% increase Flurazepam Similar to diazepam.

Similar to diazepam.

Fluvoxamine Similar to cimetidine.

Similar to cimetidine Halothane Halothane sensitizes the myocardium to catecholamines, theophylline increases release of endogenous catecholamines.

Increase risk of ventricular arrhythmias.

Interferon, human recombinant alpha-A Decreases theophylline clearance.

100% increase Isoproterenol (IV) Increases theophylline clearance.

20% decrease Ketamine Pharmacologic May lower theophylline seizure threshold.

Lithium Theophylline increases renal lithium clearance.

Lithium dose required to achieve a therapeutic serum concentration increased an average of 60% Lorazepam Similar to diazepam.

Similar to diazepam.

Methotrexate (MTX) Decreases theophylline clearance.

20% increase after low dose MTX, higher dose MTX may have greater effect.

Mexiletine Similar to disulfiram.

80% increase Midazolam Similar to diazepam.

Similar to diazepam.

Moricizine Increases theophylline clearance.

25% decrease Pancuronium Theophylline may antagonize non-depolarizing neuromuscular blocking effects; possibly due to phosphodiesterase inhibition.

Larger dose of pancuronium may be required to achieve neuromuscular blockade.

Pentoxifylline Decreases theophylline clearance.

30% increase Phenobarbital (PB) Similar to aminoglutethimide.

25% decrease after two weeks of concurrent PB.

Phenytoin Phenytoin increase theophylline clearance by increasing microsomal enzyme activity.

Theophylline decreases phenytoin absorption.

Serum theophylline and phenytoin concentrations decrease about 40%.

Propafenone Decreases theophylline clearance and pharmacologic interaction.

40% increase.

Beta-2 blocking effect may decrease efficacy of theophylline.

Propranolol Similar to cimetidine and pharmacologic interaction.

100% increase.

Beta-2 blocking effect may decrease efficacy of theophylline.

Rifampin Increases theophylline clearance by increasing cytochrome P450 1A2 and 3A3 activity.

20-40% decrease Sulfinpyrazone Increases theophylline clearance by increasing demethylation and hydroxylation.

Decreases renal clearance of theophylline.

20% decrease Tacrine Similar to cimetidine, also increases renal clearance of theophylline.

90% increase Thiabendazole Decreases theophylline clearance.

190% increase Ticlopidine Decreases theophylline clearance.

60% increase Troleandomycin Similar to erythromycin.

33-100% increase depending on troleandomycin dose.

Verapamil Similar to disulfiram.

20% increase *Refer to PRECAUTIONS, Drug Interactions for further information regarding table.

**Average effect on steady s tate theophylline concentrati on or other clinical effect for pharmacologic interactions .

Individual patients may experience larger changes in serum theophylline c oncentration than the value lis ted.

Table III.

Drugs that have been documented not to interact with theophylline or drugs that produce no clinically significant interaction with theophylline.* albuterol, systemic and inhaled felodipinefinasteride nizatidinenorfloxacin amoxicillin hydrocortisone ofloxacin ampicillin, with or without sulbactam isoflurane isoniazid omeprazole prednisone, prednisolone atenolol isradipine ranitidine azithromycin influenze vaccine rifabutin caffeine, dietary ingestion ketoconazo lelomefloxacin Roxithromycin sorbitol (purgative doses do onot inhibit theophylline absorption) cefaclor mebendazole co-trimoxazole (trimethoprim and sulfamethoxazole) medroxyprogesteronemethylprednisolone sucralfate diltiazem metronidazole terbutaline, systemic dirithromycin metoprolol terfenadine enflurane nadolol tetracycline famotidine nifedipine tocainide *Refer to PRECAUTIONS , Drug Interactions for information regarding table.

OVERDOSAGE

General: The chronicity and pattern of theophylline overdosage significantly influences clinical manifestations of toxicity, management and outcome.

There are two common presentations: (1) acute overdose, i.e., ingestion of a single large excessive dose (>10 mg/kg) as occurs in the context of an attempted suicide or isolated medication error, and (2) chronic overdosage, i.e., ingestion of repeated doses that are excessive for the patient’s rate of theophylline clearance.

The most common causes of chronic theophylline overdosage include patient or care giver error in dosing, clinician prescribing of an excessive dose or a normal dose in the presence of factors known to decrease the rate of theophylline clearance, and increasing the dose in response to an exacerbation of symptoms without first measuring the serum theophylline concentration to determine whether a dose increase is safe.

Severe toxicity from theophylline overdose is a relatively rare event.

In one health maintenance organization, the frequency of hospital admissions for chronic overdosage of theophylline was about 1 per 1000 person-years exposure.

In another study, among 6000 blood samples obtained for measurement of serum theophylline concentration, for any reason, from patients treated in an emergency department, 7% were in the 20-30 mcg/mL range and 3% were >30 mcg/mL.

Approximately two-thirds of the patients with serum theophylline concentrations in the 20-30 mcg/mL range had one or more manifestations of toxicity while >90% of patients with serum theophylline concentrations >30mcg/mL were clinically intoxicated.

Similarly, in other reports, serious toxicity from theophylline is seen principally at serum concentrations >30 mcg/mL.

Several studies have described the clinical manifestations of theophylline overdose and attempted to determine the factors that predict life-threatening toxicity.

In general, patients who experience an acute overdose are less likely to experience seizures than patients who have experienced a chronic overdosage, unless the peak serum theophylline concentration is >100 mcg/mL.

After a chronic overdosage, generalized seizures, life-threatening cardiac arrhythmias, and death may occur at serum theophylline concentrations >30 mcg/mL.

The severity of toxicity after chronic overdosage is more strongly correlated with the patient’s age than the peak serum theophylline concentration; patients >60 years are at the greatest risk for severe toxicity and mortality after a chronic overdosage.

Pre-existing or concurrent disease may also significantly increase the susceptibility of a patient to a particular toxic manifestation, e.g., patients with neurologic disorders have an increased risk of seizures and patients with cardiac disease have an increased risk of cardiac arrhythmias for a given serum theophylline concentration compared to patients without the underlying disease.

The frequency of various reported manifestations of theophylline overdose according to the mode of overdose are listed in Table IV.

Other manifestations of theophylline toxicity include increases in serum calcium, creatine kinase, myoglobin and leukocyte count, decreases in serum phosphate and magnesium, acute myocardial infarction, and urinary retention in men with obstructive uropathy.

Seizures associated with serum theophylline concentrations >30 mcg/mL are often resistant to anticonvulsant therapy and may result in irreversible brain injury if not rapidly controlled.

Death from theophylline toxicity is most often secondary to cardiorespiratory arrest and/or hypoxic encephalopathy following prolonged generalized seizures or intractable cardiac arrhythmias causing hemodynamic compromise.

Overdose Management : General Recommendations for Patients with Symptoms of Theophylline Overdose or Serum Theophylline Concentrations >30 mcg/mL (Note: Serum theophylline co ncentrations may continue to increase after presentation of the patient for medical care.) 1.

While simultaneously instituting treatment, contact a regional poison center to obtain updated information and advice on individualizing the recommendations that follow.

2.

Institute supportive care, including establishment of intravenous access, maintenance of the airway, and electrocardiographic monitoring.

3.

Treatment of seizures Because of the high morbidity and mortality associated with theophylline-induced seizures, treatment should be rapid and aggressive.

Anticonvulsant therapy should be initiated with an intravenous benzodiazepine, e.g., diazepam, in increments of 0.1-0.2 mg/kg every 1-3 minutes until seizures are terminated.

Repetitive seizures should be treated with a loading dose of Phenobarbital (20 mg/kg infused over 30-60 minutes).

Case reports of theophylline overdose in humans and animal studies suggest that phenytoin is ineffective in terminating theophylline-induced seizures.

The doses of benzodiazepines and phenobarbital required to terminate theophylline-induced seizures are close to the doses that may cause severe respiratory depression or respiratory arrest; the clinician should therefore be prepared to provide assisted ventilation.

Elderly patients and patients with COPD may be more susceptible to the respiratory depressant effects of anticonvulsants.

Barbiturate-induced coma or administration of general anesthesia may be required to terminate repetitive seizures or status epilepticus.

General anesthesia should be used with caution in patients with theophylline overdose because fluorinated volatile anesthetics may sensitize the myocardium to endogenous catecholamines released by theophylline.

Enflurane appears to less likely to be associated with this effect than halothane and may, therefore, be safer.

Neuromuscular blocking agents alone should not be used to terminate seizures since they abolish the musculoskeletal manifestations without terminating seizure activity in the brain.

4.

Anticipate Need for Anticonvulsants In patients with theophylline overdose who are at high risk for theophylline induced seizures, e.g., patients with acute overdoses and serum theophylline concentrations >100 mcg/mL chronic overdosage in patients >60 years of age with serum theophylline concentrations >30 mcg/mL, the need for anticonvulsant therapy should be anticipated.

A benzodiazepine such as diazepam should be drawn into a syringe and kept at the patient’s bedside and medical personnel qualified to treat seizures should be immediately available.

In selected patients at high risk for theophylline-induced seizures, consideration should be given to the administration of prophylactic anticonvulsant therapy.

Situations where prophylactic anticonvulsant therapy should be considered in high risk patients include anticipated delays in instituting methods for extracorporeal removal of theophylline (e.g., transfer of a high risk patient from one health care facility to another for extracorporeal removal) and clinical circumstances that significantly interfere with efforts to enhance theophylline clearance (e.g., a neonate where dialysis may not be technically feasible or a patient with vomiting unresponsive to antiemetics who is unable to tolerate multiple-dose oral activated charcoal).

In animal studies, prophylactic administration of phenobarbital, but not phenytoin, has been shown to delay the onset of theophylline induced generalized seizures and to increase the dose of theophylline required to induce seizures (i.e., markedly increases the LD50).

Although there are no controlled studies in humans, a loading dose of intravenous Phenobarbital (20 mg/kg infused over 60 minutes) may delay or prevent life-threatening seizures in high risk patients while efforts to enhance theophylline clearance are continued.

Phenobarbital may cause respiratory depression, particularly in elderly patients and patients with COPD.

5.

Treatment of cardiac arrhythmias Sinus tachycardia and simple ventricular premature beats are not harbingers of life-threatening arrhythmias, they do not require treatment in the absence of hemodynamic compromise, and they resolve with declining serum theophylline concentrations.

Other arrhythmias, especially those associated with hemodynamic compromise, should be treated with antiarrhythmic therapy appropriate for the type of arrhythmia.

6.

Gastrointestinal decontamination Oral activated charcoal (0.5 g/kg up to 20 g and repeat at least once 1-2 hours after the first dose) is extremely effective in blocking the absorption of theophylline throughout the gastrointestinal tract, even when administered several hours after ingestion.

If the patient is vomiting, the charcoal should be administered through a nasogastric tube or after administration of an antiemetic.

Phenothiazine antiemetics such as prochlorperazine or perphenazine should be avoided since they can lower the seizure threshold and frequently cause dystonic reactions.

A single dose of sorbitol may be used to promote stooling to facilitate removal of theophylline bound to charcoal from the gastrointestinal tract.

Sorbitol, however, should be dosed with caution since it is a potent purgative which can cause profound fluid and electrolyte abnormalities, particularly after multiple doses.

Commercially available fixed combinations of liquid charcoal and sorbitol should be avoided in young children and after the first dose in adolescents and adults since they do not allow for individualization of charcoal and sorbitol dosing.

Ipecac syrup should be avoided in theophylline overdoses.

Although ipecac induces emesis, it does not reduce the absorption of theophylline unless administered within 5 minutes of ingestion and even then is less effective than oral activated charcoal.

Moreover, ipecac induced emesis may persist for several hours after a single dose and significantly decrease the retention and the effectiveness of oral activated charcoal.

7.

Serum Theophylline Concentration Monitoring The serum theophylline concentration should be measured immediately upon presentation, 2-4 hours later, and then at sufficient intervals, e.g., every 4 hours, to guide treatment decisions and to assess the effectiveness of therapy.

Serum theophylline concentrations may continue to increase after presentation of the patient for medical care as a result of continued absorption of theophylline from the gastrointestinal tract.

Serial monitoring of serum theophylline serum concentrations should be continued until it is clear that the concentration is no longer rising and has returned to non-toxic 8.

General Monitoring Procedures Electrocardiographic monitoring should be initiated on presentation and continued until the serum theophylline level has returned to a non-toxic level.

Serum electrolytes and glucose should be measured on presentation and at appropriate intervals indicated by clinical circumstances.

Fluid and electrolyte abnormalities should be promptly corrected.

Monitoring and treatment should be continued until the serum concentration decreases below 20 mcg/mL.

9.

Enhance clearance of theophylline Multiple-dose oral activated charcoal (e.g., 0.5 mg/kg up to 20 g, every two hours) increases the clearance of theophylline at least twofold by adsorption of theophylline secreted into gastrointestinal fluids.

Charcoal must be retained in, and pass through, the gastrointestinal tract to be effective; emesis should therefore be controlled by administration of appropriate antiemetics.

Alternatively, the charcoal can be administered continuously through a nasogastric tube in conjunction with appropriate antiemetics.

A single dose of sorbitol may be administered with the activated charcoal to promote stooling to facilitate clearance of the adsorbed theophylline from the gastrointestinal tract.

Sorbitol alone does not enhance clearance of theophylline and should be dosed with caution to prevent excessive stooling which can result in severe fluid and electrolyte imbalances.

Commercially available fixed combinations of liquid charcoal and sorbitol should be avoided in young children and after the first dose in adolescents and adults since they do not allow for individualization of charcoal and sorbitol dosing.

In patients with intractable vomiting, extracorporeal methods of theophylline removal should be instituted (see , Extracorporeal Removal ).

Specific Recommendations: Acute Overdose A.

Serum Concentration >20<30 mcg/mL 1.

Administer a single dose of oral activated charcoal.

2.

Monitor the patient and obtain a serum theophylline concentration in 2-4 hours to insure that the concentration is not increasing.

B.Serum Concentration >30<100 mcg/mL 1.

Administer multiple dose oral activated charcoal and measures to control emesis.

2.

Monitor the patient and obtain serial theophylline concentrations every 2-4 hours to gauge the effectiveness of therapy and to guide further treatment decisions 3.

Institute extracorporeal removal if emesis, seizures, or cardiac arrhythmias cannot be adequately controlled (see , Extracorporeal Removal ).

C.

Serum Concentration >100 mcg/mL 1.

Consider prophylactic anticonvulsant therapy.

2.

Administer multiple-dose oral activated charcoal and measures to control emesis.

3.

Consider extracorporeal removal, even if the patient has not experienced a seizure (see , Extracorporeal Removal ).

4.

Monitor the patient and obtain serial theophylline concentrations every 2-4 hours to gauge the effectiveness of therapy and to guide further treatment decisions.

Chronic Overdosage A.

Serum Concentration >20<30 mcg/mL (with manifestations of theophylline toxicity) 1.

Administer a single dose of oral activated charcoal.

2.

Monitor the patient and obtain a serum theophylline concentration in 2-4 hours to insure that the concentration is not increasing.

B.

Serum Concentration >30 mcg/mL in patients <60 years of age 1.

Administer multiple-dose oral activated charcoal and measures to control emesis.

2.

Monitor the patient and obtain serial theophylline concentrations every 2-4 hours to gauge the effectiveness of therapy and to guide further treatment decisions.

3.

Institute extracorporeal removal if emesis, seizures, or cardiac arrhythmias cannot be adequately controlled (see , Extracorporeal Removal ).

C.

Serum Concentration >30 mcg/mL in patients ≥60 years of age.

1.

Consider prophylactic anticonvulsant therapy.

2.

Administer multiple-dose oral activated charcoal and measures to control emesis.

3.

Consider extracorporeal removal even if the patient has not experienced a seizure (see , Extracorporeal Removal ).

4.

Monitor the patient and obtain serial theophylline concentrations every 2-4 hours to gauge the effectiveness of therapy and to guide further treatment decisions.

Extracorporeal Removal: Increasing the rate of theophylline clearance by extracorporeal methods may rapidly decrease serum concentrations, but the risks of the procedure must be weighed against the potential benefit.

Charcoal hemoperfusion is the most effective method of extracorporeal removal, increasing theophylline clearance up to six fold, but serious complications including hypotension, hypocalcemia, platelet consumption and bleeding diatheses may occur.

Hemodialysis is about as efficient as multiple-dose oral activated charcoal and has a lower risk of serious complications than charcoal hemoperfusion.

Hemodialysis should be considered as an alternative when charcoal hemoperfusion is not feasible and multiple-dose oral charcoal is ineffective because of intractable emesis.

Serum theophylline concentrations may rebound 5-10 mcg/mL after discontinuation of charcoal hemoperfusion or hemodialysis due to redistribution of theophylline from the tissue compartment.

Peritoneal dialysis is ineffective for theophylline removal; exchange transfusions in neonates have been minimally effective.

DESCRIPTION

Theophylline is structurally classified as a methylxanthine.

It occurs as a white, odorless, crystalline powder with a bitter taste.

Anhydrous theophylline has the chemical name 1H-Purine- 2,6-dione, 3,7-dihydro-1 ,3 –dimethyl-, and is represented by the following structural formula: The molecular formula of anhydrous theophylline is C 7 H 8 N 4 0 2 with a molecular weight of 180.17.

THEOPHYLLINE ORAL SOLUTION, USP is available as a liquid intended for oral administration, containing 80 mg of theophylline anhydrous in each 15 mL (tablespoonful).

THEOPHYLLINE ORAL SOLUTION, USP also contains the following inactive ingredients: citric acid, sodium saccharin, sodium benzoate, glycerin, propylene glycol, FD and C Red #40, natural and artificial fruity flavor and purified water.

THEOPHYLLINE ORAL SOLUTION, USP has a pH of 4.0 – 5.0.

Theophylline is structurally classified as a methylxanthine.

It occurs as a white, odorless, crystalline powder with a bitter taste.

Anhydrous theophylline has the chemical name 1H-Purine- 2,6-dione, 3,7-dihydro-1 ,3 –dimethyl-, and is represented by the following structural formula:

CLINICAL STUDIES

Clinical Studies: In patients with chronic asthma, including patients with severe asthma requiring inhaled corticosteroids or alternate-day oral corticosteroids, many clinical studies have shown that theophylline decreases the frequency and severity of symptoms, including nocturnal exacerbations, and decreases the “as needed” use of inhaled beta-2 agonists.

Theophylline has also been shown to reduce the need for short courses of daily oral prednisone to relieve exacerbations of airway obstruction that are unresponsive to bronchodilators in asthmatics.

In patients with chronic obstructive pulmonary disease (COPD), clinical studies have shown that theophylline decreases dyspnea, air trapping, the work of breathing, and improves contractility of diaphragmatic muscles with little or no improvement in pulmonary function measurements.

HOW SUPPLIED

THEOPHYLLINE ORAL SOLUTION, USP is a clear red solution with a fruit flavor.

Each tablespoonful (15 mL) contains 80 mg theophylline anhydrous.

THEOPHYLLINE ORAL SOLUTION, USP is available in bottles of 473 mL NDC 10135-0604-08 Bottles of 473mL (16 ounces)

GERIATRIC USE

Geriatric Use: Elderly patients are at significantly greater risk of experiencing serious toxicity from theophylline than younger patients due to pharmacokinetic and pharmacodynamic changes associated with aging.

Theophylline clearance is reduced in patients greater than 60 years of age, resulting in increased serum theophylline concentrations in response to a given theophylline dose.

Protein binding may be decreased in the elderly resulting in a larger proportion of the total serum theophylline concentration in the pharmacologically active unbound form.

Elderly patients also appear to be more sensitive to the toxic effects of theophylline after chronic overdosage than younger patients.

For these reasons, the maximum daily dose of theophylline in patients greater than 60 years of age ordinarily should not exceed 400 mg/day unless the patient continues to be symptomatic and the peak steady state serum theophylline concentration is <10 mcg/mL (see DOSAGE AND ADMINISTRATION ).

Theophylline doses greater than 400 mg/d should be prescribed with caution in elderly patients.

MECHANISM OF ACTION

Mechanism of Action: Theophylline has two distinct actions in the airways of patients with reversible obstruction; smooth muscle relaxation (i.e., bronchodilation) and suppression of the response of the airways to stimuli (i.e., non-bronchodilator prophylactic effects).

While the mechanisms of action of theophylline are not known with certainty, studies in animals suggest that bronchodilatation is mediated by the inhibition of two isozymes of phosphodiesterase (PDE III and, to a lesser extent, PDE IV) while non-bronchodilator prophylactic actions are probably mediated through one or more different molecular mechanisms, that do not involve inhibition of PDE III or antagonism of adenosine receptors.

Some of the adverse effects associated with theophylline appear to be mediated by inhibition of PDE III (e.g., hypotension, tachycardia, headache, and emesis) and adenosine receptor antagonism (e.g., alterations in cerebral blood flow).

Theophylline increases the force of contraction of diaphragmatic muscles.

This action appears to be due to enhancement of calcium uptake through an adenosine-mediated channel.

INDICATIONS AND USAGE

Theophylline is indicated for the treatment of the symptoms and reversible airflow obstruction associated with chronic asthma and other chronic lung diseases, e.g., emphysema and chronic bronchitis.

PEDIATRIC USE

Pediatric Use: Theophylline is safe and effective for the approved indications in pediatric patients (See INDICATIONS AND USAGE ).

The maintenance dose of theophylline must be selected with caution in pediatric patients since the rate of theophylline clearance is highly variable across the age range of neonates to adolescents (see CLINICAL PHARMACOLOGY , Table I , WARNINGS , and DOSAGE AND ADMINISTRATION , Table V ).

Due to the immaturity of theophylline metabolic pathways in infants under the age of one year, particular attention to dosage selection and frequent monitoring of serum theophylline concentrations are required when theophylline is prescribed to pediatric patients in this age group.

PREGNANCY

Pregnancy: CATEGORY C : There are no adequate and well controlled studies in pregnant women.

Additionally, there are no teratogenicity studies in non-rodents (e.g., rabbits).

Theophylline was not shown to be teratogenic in CD-1 mice at oral doses up to 400 mg/kg, approximately 2.0 times the human dose on a mg/m2 basis or in CD-1 rats at oral doses up to 260 mg/kg, approximately 3.0 times the recommended human dose on a mg/m 2 basis.

At a dose of 220 mg/kg, embryotoxicity was observed in rats in the absence of maternal toxicity.

NUSRING MOTHERS

Nursing Mothers: Theophylline is excreted into breast milk and may cause irritability or other signs of mild toxicity in nursing human infants.

The concentration of theophylline in breast milk is about equivalent to the maternal serum concentration.

An infant ingesting a liter of breast milk containing 10-20 mcg/mL of theophylline per day is likely to receive 10-20 mg of theophylline per day.

Serious adverse effects in the infant are unlikely unless the mother has toxic serum theophylline concentrations.

INFORMATION FOR PATIENTS

Information for Patients: The patient (or parent/care giver) should be instructed to seek medical advice whenever nausea, vomiting, persistent headache, insomnia or rapid heart beat occurs during treatment with theophylline, even if another cause is suspected.

The patient should be instructed to contact their clinician if they develop a new illness, especially if accompanied by a persistent fever, if they experience worsening of a chronic illness, if they start or stop smoking cigarettes or marijuana, or if another clinician adds a new medication or discontinues a previously prescribed medication.

Patients should be instructed to inform all clinicians involved in their care that they are taking theophylline, especially when a medication is being added or deleted from their treatment.

Patients should be instructed to not alter the dose, timing of the dose, or frequency of administration without first consulting their clinician.

If a dose is missed, the patient should be instructed to take the next dose at the usually scheduled time and to not attempt to make up for the missed dose.

DOSAGE AND ADMINISTRATION

General Considerations: The steady-state peak serum theophylline concentration is a function of the dose, the dosing interval, and the rate of theophylline absorption and clearance in the individual patient.

Because of marked individual differences in the rate of theophylline clearance, the dose required to achieve a peak serum theophylline concentration in the 10-20 mcg/mL range varies fourfold among otherwise similar patients in the absence of factors known to alter theophylline clearance (e.g., 400- 1600 mg/day in adults <60 years old and 10-36 mg/kg/day in children 1-9 years old).

For a given population there is no single theophylline dose that will provide both safe and effective serum concentrations for all patients.

Administration of the median theophylline dose required to achieve a therapeutic serum theophylline concentration in a given population may result in either sub-therapeutic or potentially toxic serum theophylline concentrations in individual patients.

For example, at a dose of 900 mg/d in adults <60 years or 22 mg/kg/d in children 1-9 years, the steady state peak serum theophylline concentration will be <10 mcg/mL in about 30% of patients, 10-20 mcg/mL in about 50% and 20-30 mcg/mL in about 20% of patients.

The dos e of theophylline mus t be individ ualized on the bas is of peak s erum theophylline concentration meas urements in order to achieve a dos e that will provide maximum potential benefit with minimal risk to adverse effects .

Transient caffeine-like adverse effects and excessive serum concentrations in slow metabolizers can be avoided in most patients by starting with a sufficiently low dose and slowly increasing the dose, if judged to be clinically indicated, in small increments (See Table V ).

Dose increases should only be made if the previous dosage is well tolerated and at intervals of no less than 3 days to allow serum theophylline concentrations to reach the new steady state.

Dosage adjustment should be guided by serum theophylline concentration measurement (see PRECAUTIONS, Laboratory Tests and , Table VI ).

Health care providers should instruct patients and care givers to discontinue any dosage that causes adverse effects, to withhold the medication until these symptoms are gone and to then resume therapy at a lower, previously tolerated dosage (see WARNINGS ).

If the patient’s symptoms are well controlled, there are no apparent adverse effects, and no intervening factors that might alter dosage requirements (see WARNINGS and PRECAUTIONS ), serum theophylline concentrations should be monitored at 6 month intervals for rapidly growing children and at yearly intervals for all others.

In acutely ill patients, serum theophylline concentrations should be monitored at frequent intervals, e.g., every 24 hours.

Theophylline distributes poorly into body fat, therefore, mg/kg dose should be calculated on the basis of ideal body weight.

Table V contains theophylline dosing titration schema recommended for patients in various age groups and clinical circumstances.

Table VI contains recommendations for theophylline dosage adjustment based upon serum theophylline concentrations.

Application of these general dos ing recommendat ions to individual patients mus t take into account the unique clinical characteris tics of each patient.

In general, these recommendations should serve as the upper limit for dos age adj ustments in order to decrease the risk of potentially serious adverse events ass ociated with unexpected large increases in s erum theophylline concentration.

A.

Infants <1 year old.

1.

Initial Dosage.

a.

Premature Neonates: i.

<24 days postnatal age; 1.0 mg/kg every 12 hr ii.

≥ 24 days postnatal age; 1.5 mg/kg every 12 hr b.

Full term infants and infants up to 52 weeks of age: Total daily dose (mg) = [(0.2 x age in weeks)+5.0] x (Kg body Wt).

i.

up to age 26 weeks; divide dose into 3 equal amounts administered at 8 hour intervals ii.

≥26 weeks of age; divide dose into 4 equal amounts administered at 6 hour intervals.

2.

Final Dosage.

Adjusted to maintain a peak steady state serum theophylline concentration of 5-10 mcg/ml in neonates and 10-15 mcg/mL in older infants (see Table VI ).

Since the time required to reach steady-state is a function of theophylline half-life, up to 5 days may be required to achieve steady state in a premature neonate while only 2-3 days may be required in a 6 month old infant without other risk factors for impaired clearance in the absence of a loading dose.

If a s erum theophylline concentrati on is obtained before steady s tate is achieved, the maintena nce dose s hould not be increased, even if the s erum theophylline concentration is <10 mcg/mL.

B.

Children (1-15 years ) and ad ults (16-60 years) without ris k factors for impaired clearance.

Table V.

Dosing initation and titration (as anhydrous theophylline).* Titration Step Children 45 kg and adults 1.

Starting Dosage 12-14 mg/kg/day up to a maximum of 300 mg/day divided Q4-6 hrs* 300 mg/day divided Q6-8 hrs* 2.

After 3 days, if tolerated, increase dose to: 16 mg/kg/day up to a maximum of 400 mg/day divided Q4-6 hrs* 400 mg/day divided Q6-8 hrs* 3.

After 3 more days, if tolerated , increase dose to: 20 mg/kg/day up to a maximum of 600 mg/day divided Q4-6 hrs* 600 mg/day divided Q6-8 hrs* C.

Patients With Risk Factors For Impaired Clearance, The Elderly (>60 Years), And Those In Whom It Is Not Feasible To Monitor Serum Theophylline Concentrations: In children 1-15 years of age, the final theophylline dose should not exceed 16 mg/kg/day up to a maximum of 400 mg/day in the presence of risk factors for reduced theophylline clearance (see WARNINGS ) or if it is not feasible to monitor serum theophylline concentrations.

In adolescents ≥16 years and adults, including the elderly, the final theophylline dose should not exceed 400 mg/day in the presence of risk factors for reduced theophylline clearance (see WARNINGS ) or if it is not feasible to monitor serum theophylline concentrations.

D.

Loading Dose for Acute Bronchodilatation : An inhaled beta-2 selective agonist, alone or in combination with a systemically administered corticosteroid, is the most effective treatment for acute exacerbations of reversible airways obstruction.

Theophylline is a relatively weak bronchodilator, is less effective than an inhaled beta-2 selective agonist and provides no added benefit in the treatment of acute bronchospasm.

If an inhaled or parenteral beta agonist is not available, a loading dose of an oral immediate release theophylline can be used as a temporary measure.

A single 5 mg/kg dose of theophylline, in a patient who has not received any theophylline in the previous 24 hours, will produce an average peak serum theophylline concentration of 10 mcg/mL (range 5-15 mcg/mL).

If dosing with theophylline is to be continued beyond the loading dose, the guidelines in Sections A.1.b., B.3, or C., above, should be utilized and serum theophylline concentration monitored at 24 hour intervals to adjust final dosage.

* Patients with more rapid metabolism, clinically identified by higher than average dose requirements, should receive a smaller dose more frequently to prevent breakthrough symptoms resulting from low trough concentrations before the next dose.

A reliably absorbed slow-release formulation will decrease fluctuations and permit longer dosing intervals.

VI Table.

Dosage adjustment guided by serum theophylline concentration.

Peak Serum Concentration Dosage Adjustment <9.9 mcg/mL If symptoms are not controlled and current dosage is tolerated, increase dose about 25%.

Recheck serum concentration after three days for further dosage adjustment.

10 to 14.9 mcg/mL If symptoms are controlled and current dosage is tolerated, maintain dose and recheck serum concentration at 6-12 month intervals.

¶ If symptoms are not controlled and current dosage is tolerated consider adding additional medication(s) to treatment regimen.

15-19.9 mcg/mL Consider 10% decrease in dose to provide greater margin of safety even if current dosage is tolerated.¶ 20-24.9 mcg/mL Decrease dose by 25% even if no adverse effects are present.

Recheck serum concentration after 3 days to guide further dosage adjustment.

25-30 mcg/mL Skip next dose and decrease subsequent doses at least 25% even if no adverse effects are present.

Recheck serum concentration after 3 days to guide further dosage adjustment.

If symptomatic, consider whether overdose treatment is indicated (see recommendations for chronic overdosage).

>30 mcg/mL Treat overdose as indicated (see recommendations for chronic overdosage).

If theophylline is subsequently resumed, decrease dose by at least 50% and recheck serum concentration after 3 days to guide further dosage adjustment.

¶ Dose reduction and/or serum theophylline concentration measurement is indicated whenever adverse effects are present, physiologic abnormalities that can reduce theophylline clearance occur (e.g., sustained fever), or a drug that interacts with theophylline is added or discontinued (see WARNINGS ).

CortiSil MDX 1 % Topical Gel

WARNINGS

Warnings For external use only

INDICATIONS AND USAGE

INACTIVE INGREDIENTS

Inactive ingredients Cyclopentasiloxane, Dimethicone Crosspolymer, Cyclomethicone, PEG-12 Glyceryl Dimyristate, Disodium EDTA.

PURPOSE

Use For the temporary relief of itching associated with minor skin irritations, inflammation and rashes.

KEEP OUT OF REACH OF CHILDREN

Keep out of reach of children.

If swallowed, get medical help or contact a Poison Control Center immediately.

DOSAGE AND ADMINISTRATION

Directions Adults and children 2 years of age and older: Apply to the affected area not more than 3 to 4 times daily.

Children under 2 year of age: consult a doctor.

DO NOT USE

Do not use hydrocortisone product unless you have consulted a doctor.

on children under 2 years of age.

on mucous membranes

STOP USE

Stop use and ask a doctor if condition worsens symptoms persist for more than 7 days or clear up and occur again within a few days.

ACTIVE INGREDIENTS

Active ingredient Hydrocortisone Acetate 1.0% Purpose Anti-Pruritic

Tetrahydrocannabinol 10 MG Oral Capsule

WARNINGS

Patients receiving treatment with Dronabinol Capsules should be specifically warned not to drive, operate machinery, or engage in any hazardous activity until it is established that they are able to tolerate the drug and to perform such tasks safely.

DRUG INTERACTIONS

Drug Interactions In studies involving patients with AIDS and/or cancer, Dronabinol Capsules has been co-administered with a variety of medications (e.g., cytotoxic agents, anti-infective agents, sedatives, or opioid analgesics) without resulting in any clinically significant drug/drug interactions.

Although no drug/drug interactions were discovered during the clinical trials of Dronabinol Capsules, cannabinoids may interact with other medications through both metabolic and pharmacodynamic mechanisms.

Dronabinol is highly protein bound to plasma proteins, and therefore, might displace other protein-bound drugs.

Although this displacement has not been confirmed in vivo , practitioners should monitor patients for a change in dosage requirements when administering dronabinol to patients receiving other highly protein-bound drugs.

Published reports of drug/drug interactions involving cannabinoids are summarized in the following table.

CONCOMITANT DRUG CLINICAL EFFECT(S) Amphetamines, cocaine, other sympathomimetic agents Additive hypertension, tachycardia, possibly cardiotoxicity Atropine, scopolamine, antihistamines, other anticholinergic agents Additive or super-additive tachycardia, drowsiness Amitriptyline, amoxapine, desipramine, other tricyclic antidepressants Additive tachycardia, hypertension, drowsiness Barbiturates, benzodiazepines, ethanol, lithium, opioids, buspirone, antihistamines, muscle relaxants, other CNS depressants Additive drowsiness and CNS depression Disulfiram A reversible hypomanic reaction was reported in a 28 y/o man who smoked marijuana; confirmed by dechallenge and rechallenge Fluoxetine A 21 y/o female with depression and bulimia receiving 20 mg/day fluoxetine × 4 wks became hypomanic after smoking marijuana; symptoms resolved after 4 days Antipyrine, barbiturates Decreased clearance of these agents, presumably via competitive inhibition of metabolism Theophylline Increased theophylline metabolism reported with smoking of marijuana; effect similar to that following smoking tobacco

OVERDOSAGE

Signs and symptoms following MILD Dronabinol Capsules intoxication include drowsiness, euphoria, heightened sensory awareness, altered time perception, reddened conjunctiva, dry mouth and tachycardia; following MODERATE intoxication include memory impairment, depersonalization, mood alteration, urinary retention, and reduced bowel motility; and following SEVERE intoxication include decreased motor coordination, lethargy, slurred speech, and postural hypotension.

Apprehensive patients may experience panic reactions and seizures may occur in patients with existing seizure disorders.

The estimated lethal human dose of intravenous dronabinol is 30 mg/kg (2100 mg/ 70 kg).

Significant CNS symptoms in antiemetic studies followed oral doses of 0.4 mg/kg (28 mg/70 kg) of Dronabinol Capsules.

Management A potentially serious oral ingestion, if recent, should be managed with gut decontamination.

In unconscious patients with a secure airway, instill activated charcoal (30 to 100 g in adults, 1 to 2 g/kg in infants) via a nasogastric tube.

A saline cathartic or sorbitol may be added to the first dose of activated charcoal.

Patients experiencing depressive, hallucinatory or psychotic reactions should be placed in a quiet area and offered reassurance.

Benzodiazepines (5 to 10 mg diazepam po ) may be used for treatment of extreme agitation.

Hypotension usually responds to Trendelenburg position and IV fluids.

Pressors are rarely required.

DESCRIPTION

Dronabinol is a cannabinoid designated chemically as ( 6aR-trans )-6a,7,8,10a-tetrahydro-6,6,9-trimethyl-3-pentyl- 6H -dibenzo[ b,d ]pyran-1-ol.

Dronabinol has the following empirical and structural formulas: Dronabinol, the active ingredient in Dronabinol Capsules, is synthetic delta-9-tetrahydrocannabinol (delta-9-THC).

Delta-9-tetrahydrocannabinol is also a naturally occurring component of Cannabis sativa L.

(Marijuana).

Dronabinol is a light yellow resinous oil that is sticky at room temperature and hardens upon refrigeration.

Dronabinol is insoluble in water and is formulated in sesame oil.

It has a pKa of 10.6 and an octanol-water partition coefficient: 6,000:1 at pH 7.

Capsules for oral administration: Dronabinol Capsules are supplied as oval, soft gelatin capsules containing either 2.5 mg, 5 mg, or 10 mg dronabinol.

Each Dronabinol Capsule strength is formulated with the following inactive ingredients: 2.5 mg capsule contains gelatin, glycerin, sesame oil, titanium dioxide, SDA 35A alcohol, propylene glycol, black iron oxide, polyvinyl acetate phthalate, purified water, isopropyl alcohol, polyethylene glycol, and ammonium hydroxide; 5 mg capsule contains iron oxide red, iron oxide black, gelatin, glycerin, sesame oil, titanium dioxide, SDA 35A alcohol, propylene glycol, polyvinyl acetate phthalate, purified water, isopropyl alcohol, polyethylene glycol, and ammonium hydroxide; 10 mg capsule contains iron oxide red, iron oxide yellow, gelatin, glycerin, sesame oil, titanium dioxide, SDA 35A alcohol, propylene glycol, black iron oxide, polyvinyl acetate phthalate, purified water, isopropyl alcohol, polyethylene glycol, and ammonium hydroxide.

Chemical Structure

CLINICAL STUDIES

Clinical Trials Appetite Stimulation The appetite stimulant effect of Dronabinol Capsules in the treatment of AIDS-related anorexia associated with weight loss was studied in a randomized, double-blind, placebo-controlled study involving 139 patients.

The initial dosage of Dronabinol Capsules in all patients was 5 mg/day, administered in doses of 2.5 mg one hour before lunch and one hour before supper.

In pilot studies, early morning administration of Dronabinol Capsules appeared to have been associated with an increased frequency of adverse experiences, as compared to dosing later in the day.

The effect of Dronabinol Capsules on appetite, weight, mood, and nausea was measured at scheduled intervals during the six-week treatment period.

Side effects (feeling high, dizziness, confusion, somnolence) occurred in 13 of 72 patients (18%) at this dosage level and the dosage was reduced to 2.5 mg/day, administered as a single dose at supper or bedtime.

Of the 112 patients that completed at least 2 visits in the randomized, double-blind, placebo-controlled study, 99 patients had appetite data at 4-weeks (50 received Dronabinol Capsules and 49 received placebo) and 91 patients had appetite data at 6-weeks (46 received Dronabinol Capsules and 45 received placebo).

A statistically significant difference between Dronabinol Capsules and placebo was seen in appetite as measured by the visual analog scale at weeks 4 and 6 (see figure ).

Trends toward improved body weight and mood, and decreases in nausea were also seen.

After completing the 6-week study, patients were allowed to continue treatment with Dronabinol Capsules in an open-label study, in which there was a sustained improvement in appetite.

Figure Antiemetic Dronabinol Capsules treatment of chemotherapy-induced emesis was evaluated in 454 patients with cancer, who received a total of 750 courses of treatment of various malignancies.

The antiemetic efficacy of Dronabinol Capsules was greatest in patients receiving cytotoxic therapy with MOPP for Hodgkin’s and non-Hodgkin’s lymphomas.

Dronabinol Capsule dosages ranged from 2.5 mg/day to 40 mg/day, administered in equally divided doses every four to six hours (four times daily).

As indicated in the following table, escalating the Dronabinol Capsules dose above 7 mg/m 2 increased the frequency of adverse experiences, with no additional antiemetic benefit.

Dronabinol Capsules Dose: Response Frequency and Adverse Experiences Nondysphoric events consisted of drowsiness, tachycardia, etc.

(N = 750 treatment courses) Dronabinol Capsules Dose Response Frequency (%) Adverse Events Frequency (%) Complete Partial Poor None Nondysphoric Dysphoric 7 mg/m 2 33 31 36 13 58 28 Combination antiemetic therapy with Dronabinol Capsules and a phenothiazine (prochlorperazine) may result in synergistic or additive antiemetic effects and attenuate the toxicities associated with each of the agents.

HOW SUPPLIED

Dronabinol Capsules are available containing 2.5 mg, 5 mg or 10 mg of dronabinol.

The 2.5 mg capsule is an opaque off-white soft gelatin capsule printed with INS in black ink.

They are available as follows: NDC 0378-8170-91 bottles of 60 capsules The 5 mg capsule is an opaque maroon or brown soft gelatin capsule printed with INS in white ink.

They are available as follows: NDC 0378-8171-91 bottles of 60 capsules The 10 mg capsule is an opaque tan to tan-orange soft gelatin capsule printed with INS in black ink.

They are available as follows: NDC 0378-8172-91 bottles of 60 capsules

GERIATRIC USE

Geriatric Use Clinical studies of Dronabinol Capsules in AIDS and cancer patients did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects.

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

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

INDICATIONS AND USAGE

Dronabinol Capsules is indicated for the treatment of: 1.

anorexia associated with weight loss in patients with AIDS; and 2.

nausea and vomiting associated with cancer chemotherapy in patients who have failed to respond adequately to conventional antiemetic treatments.

PREGNANCY

Pregnancy Pregnancy Category C Reproduction studies with dronabinol have been performed in mice at 15 to 450 mg/m 2 , equivalent to 0.2 to 5 times maximum recommended human dose (MRHD) of 90 mg/m 2 /day in cancer patients or 1 to 30 times MRHD of 15 mg/m 2 /day in AIDS patients, and in rats at 74 to 295 mg/m 2 (equivalent to 0.8 to 3 times MRHD of 90 mg/m 2 in cancer patients or 5 to 20 times MRHD of 15 mg/ m 2 /day in AIDS patients).

These studies have revealed no evidence of teratogenicity due to dronabinol.

At these dosages in mice and rats, dronabinol decreased maternal weight gain and number of viable pups and increased fetal mortality and early resorptions.

Such effects were dose dependent and less apparent at lower doses which produced less maternal toxicity.

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

Dronabinol should be used only if the potential benefit justifies the potential risk to the fetus.

NUSRING MOTHERS

Nursing Mothers Use of Dronabinol Capsules is not recommended in nursing mothers since, in addition to the secretion of HIV virus in breast milk, dronabinol is concentrated in and secreted in human breast milk and is absorbed by the nursing baby.

INFORMATION FOR PATIENTS

Information for Patients Patients receiving treatment with Dronabinol Capsules should be alerted to the potential for additive central nervous system depression if Dronabinol Capsules is used concomitantly with alcohol or other CNS depressants such as benzodiazepines and barbiturates.

Patients receiving treatment with Dronabinol Capsules should be specifically warned not to drive, operate machinery, or engage in any hazardous activity until it is established that they are able to tolerate the drug and to perform such tasks safely.

Patients using Dronabinol Capsules should be advised of possible changes in mood and other adverse behavioral effects of the drug so as to avoid panic in the event of such manifestations.

Patients should remain under the supervision of a responsible adult during initial use of Dronabinol Capsules and following dosage adjustments.

DOSAGE AND ADMINISTRATION

Appetite Stimulation Initially, 2.5 mg Dronabinol Capsules should be administered orally twice daily (b.i.d.), before lunch and supper.

For patients unable to tolerate this 5 mg/day dosage of Dronabinol Capsules, the dosage can be reduced to 2.5 mg/day, administered as a single dose in the evening or at bedtime.

If clinically indicated and in the absence of significant adverse effects, the dosage may be gradually increased to a maximum of 20 mg/day Dronabinol Capsules, administered in divided oral doses.

Caution should be exercised in escalating the dosage of Dronabinol Capsules because of the increased frequency of dose-related adverse experiences at higher dosages.

(See PRECAUTIONS .

) Antiemetic Dronabinol Capsules are best administered at an initial dose of 5 mg/m 2 , given 1 to 3 hours prior to the administration of chemotherapy, then every 2 to 4 hours after chemotherapy is given, for a total of 4 to 6 doses/day.

Should the 5 mg/m 2 dose prove to be ineffective, and in the absence of significant side effects, the dose may be escalated by 2.5 mg/m 2 increments to a maximum of 15 mg/m 2 per dose.

Caution should be exercised in dose escalation, however, as the incidence of disturbing psychiatric symptoms increases significantly at maximum dose.

(See PRECAUTIONS .

)

ERYTHROPOIETIN 10000 UNT/ML Injectable Solution [Procrit]

Generic Name: ERYTHROPOIETIN
Brand Name: PROCRIT
  • Substance Name(s):
  • EPOETIN

OVERDOSAGE

10 PROCRIT overdosage can cause hemoglobin levels above the desired level, which should be managed with discontinuation or reduction of PROCRIT dosage and/or with phlebotomy, as clinically indicated [see Clinical Pharmacology (12.2) ] .

Cases of severe hypertension have been observed following overdose with ESAs [see Warnings and Precautions (5.3) ].

DESCRIPTION

11 Epoetin alfa is a 165-amino acid erythropoiesis-stimulating glycoprotein manufactured by recombinant DNA technology.

It has a molecular weight of approximately 30,400 daltons and is produced by mammalian cells into which the human erythropoietin gene has been introduced.

The product contains the identical amino acid sequence of isolated natural erythropoietin.

PROCRIT (epoetin alfa) injection for intravenous or subcutaneous administration is formulated as a sterile, clear, colorless liquid in vials in multiple formulations.

Single-dose vials, formulated with an isotonic sodium chloride/sodium citrate-buffered solution, are supplied in multiple strengths.

Each single-dose 1 mL vial contains 2,000, 3,000, 4,000, or 10,000 Units of epoetin alfa, Albumin (Human) (2.5 mg), citric acid (0.06 mg), sodium chloride (5.9 mg), and sodium citrate (5.8 mg) in Water for Injection, USP (pH 6.9 ± 0.3).

Single-dose 1 mL vials formulated with an isotonic sodium chloride/sodium phosphate buffer contain 40,000 Units of epoetin alfa albumin (human) (2.5 mg), citric acid (0.0068 mg), sodium chloride (5.8 mg), sodium citrate (0.7 mg), sodium phosphate dibasic anhydrate (1.8 mg), and sodium phosphate monobasic monohydrate (1.2 mg) in Water for Injection, USP (pH 6.9 ± 0.3).

Multiple-dose, 2 mL vials contain 10,000 Units epoetin alfa, albumin (human) (2.5 mg), benzyl alcohol (1%), sodium chloride (8.2 mg), citric acid (0.11 mg), and sodium citrate (1.3 mg) per 1 mL Water for Injection, USP (pH 6.1 ± 0.3).

Multiple-dose 1 mL vials contain 20,000 Units epoetin alfa, albumin (human) (2.5 mg), benzyl alcohol (1%), sodium chloride (8.2 mg), citric acid (0.11 mg), and sodium citrate (1.3 mg), per 1 mL in Water for Injection, USP (pH 6.1 ± 0.3).

CLINICAL STUDIES

14 14.1 Patients with Chronic Kidney Disease Adult Patients on Dialysis Patients with chronic kidney disease on dialysis: ESA effects on rates of transfusion In clinical studies of patients with CKD on dialysis, PROCRIT increased hemoglobin levels and decreased the need for RBC transfusion.

Overall, more than 95% of patients were RBC transfusion-independent after receiving PROCRIT for 3 months.

In clinical studies at starting doses of 50 to 150 Units/kg 3 times weekly, adult patients responded with an average rate of hemoglobin rise as presented in Table 8.

Table 8: Average Rate of Hemoglobin Rise in 2 Weeks Starting Dose (3 Times Weekly Intravenously) Hemoglobin Increase in 2 Weeks 50 Units/kg 0.5 g/dL 100 Units/kg 0.8 g/dL 150 Units/kg 1.2 g/dL The safety and efficacy of PROCRIT were evaluated in 13 clinical studies involving intravenous administration to a total of 1010 patients on dialysis with anemia.

Overall, more than 90% of the patients treated with PROCRIT experienced improvement in hemoglobin concentrations.

In the 3 largest of these clinical studies, the median maintenance dose necessary to maintain the hemoglobin between 10 to 12 g/dL was approximately 75 Units/kg 3 times weekly.

More than 95% of patients were able to avoid RBC transfusions.

In the largest US multicenter study, approximately 65% of the patients received doses of 100 Units/kg 3 times weekly or less to maintain their hemoglobin at approximately 11.7 g/dL.

Almost 10% of patients received a dose of 25 Units/kg or less, and approximately 10% received a dose of more than 200 Units/kg 3 times weekly to maintain their hemoglobin at this level.

In the Normal Hematocrit Study, the yearly transfusion rate was 51.5% in the lower hemoglobin group (10 g/dL) and 32.4% in the higher hemoglobin group (14 g/dL).

Other ESA trials In a 26-week, double-blind, placebo-controlled study, 118 patients on dialysis with an average hemoglobin of approximately 7 g/dL were randomized to either PROCRIT or placebo.

By the end of the study, average hemoglobin increased to approximately 11 g/dL in the PROCRIT-treated patients and remained unchanged in patients receiving placebo.

PROCRIT-treated patients experienced improvements in exercise tolerance and patient-reported physical functioning at month 2 that were maintained throughout the study.

A multicenter, unit-dose study was also conducted in 119 patients receiving peritoneal dialysis who self-administered PROCRIT subcutaneously.

Patients responded to PROCRIT administered subcutaneously in a manner similar to patients receiving intravenous administration.

Pediatric Patients with CKD on Dialysis The safety and efficacy of PROCRIT were studied in a placebo-controlled, randomized study of 113 pediatric patients with anemia (hemoglobin ≤ 9 g/dL) undergoing peritoneal dialysis or hemodialysis.

The initial dose of PROCRIT was 50 Units/kg intravenously or subcutaneously 3 times weekly.

The dose of study drug was titrated to achieve either a hemoglobin of 10 to 12 g/dL or an absolute increase in hemoglobin of 2 g/dL over baseline.

At the end of the initial 12 weeks, a statistically significant rise in mean hemoglobin (3.1 g/dL vs.

0.3 g/dL) was observed only in the PROCRIT arm.

The proportion of pediatric patients achieving a hemoglobin of 10 g/dL, or an increase in hemoglobin of 2 g/dL over baseline, at any time during the first 12 weeks was higher in the PROCRIT arm (96% vs.

58%).

Within 12 weeks of initiating PROCRIT therapy, 92.3% of the pediatric patients were RBC transfusion independent as compared to 65.4% who received placebo.

Among patients who received 36 weeks of PROCRIT, hemodialysis patients received a higher median maintenance dose [167 Units/kg/week (n=28) vs.

76 Units/kg/week (n=36)] and took longer to achieve a hemoglobin of 10 to 12 g/dL (median time to response 69 days vs.

32 days) than patients undergoing peritoneal dialysis.

Adult Patients with CKD Not Requiring Dialysis Four clinical studies were conducted in patients with CKD not on dialysis involving 181 patients treated with PROCRIT.

These patients responded to PROCRIT therapy in a manner similar to that observed in patients on dialysis.

Patients with CKD not on dialysis demonstrated a dose-dependent and sustained increase in hemoglobin when PROCRIT was administered by either an intravenous or subcutaneous route, with similar rates of rise of hemoglobin when PROCRIT was administered by either route.

Patients with chronic kidney disease not on dialysis: ESA effects on rates of transfusion In TREAT, a randomized, double-blind trial of 4038 patients with CKD and type 2 diabetes not on dialysis, a post-hoc analysis showed that the proportion of patients receiving RBC transfusions was lower in patients administered an ESA to target a hemoglobin of 13 g/dL compared to the control arm in which an ESA was administered intermittently if hemoglobin concentration decreased to less than 9 g/dL (15% versus 25%, respectively).

In CHOIR, a randomized open-label study of 1432 patients with CKD not on dialysis, use of epoetin alfa to target a higher (13.5 g/dL) versus lower (11.3 g/dL) hemoglobin goal did not reduce the use of RBC transfusions.

In each trial, no benefits occurred for the cardiovascular or end-stage renal disease outcomes.

In each trial, the potential benefit of ESA therapy was offset by worse cardiovascular safety outcomes resulting in an unfavorable benefit-risk profile [see Warnings and Precautions (5.1) ] .

ESA Effects on Rates of Death and Other Serious Cardiac Adverse Reactions Three randomized outcome trials (Normal Hematocrit Study [NHS], Correction of Anemia with Epoetin Alfa in Chronic Kidney Disease [CHOIR], and Trial of Darbepoetin Alfa in Type 2 Diabetes and CKD [TREAT]) have been conducted in patients with CKD using Epogen/PROCRIT/Aranesp to target higher vs.

lower hemoglobin levels.

Though these trials were designed to establish a cardiovascular or renal benefit of targeting higher hemoglobin levels, in all 3 studies, patients randomized to the higher hemoglobin target experienced worse cardiovascular outcomes and showed no reduction in progression to ESRD.

In each trial, the potential benefit of ESA therapy was offset by worse cardiovascular safety outcomes resulting in an unfavorable benefit-risk profile [see Warnings and Precautions (5.1) ] .

14.2 Zidovudine-treated Patients with HIV Infection The safety and efficacy of PROCRIT were evaluated in 4 placebo-controlled studies enrolling 297 anemic patients (hemoglobin < 10 g/dL) with HIV Infection receiving concomitant therapy with zidovudine.

In the subgroup of patients (89/125 PROCRIT and 88/130 placebo) with pre-study endogenous serum erythropoietin levels ≤ 500 mUnits/mL, PROCRIT reduced the mean cumulative number of units of blood transfused per patient by approximately 40% as compared to the placebo group.

Among those patients who required RBC transfusions at baseline, 43% of patients treated with PROCRIT versus 18% of placebo-treated patients were RBC transfusion-independent during the second and third months of therapy.

PROCRIT therapy also resulted in significant increases in hemoglobin in comparison to placebo.

When examining the results according to the weekly dose of zidovudine received during month 3 of therapy, there was a statistically significant reduction (p < 0.003) in RBC transfusion requirements in patients treated with PROCRIT (n=51) compared to placebo-treated patients (n=54) whose mean weekly zidovudine dose was ≤ 4200 mg/week.

Approximately 17% of the patients with endogenous serum erythropoietin levels ≤ 500 mUnits/mL receiving PROCRIT in doses from 100 to 200 Units/kg 3 times weekly achieved a hemoglobin of 12.7 g/dL without administration of RBC transfusions or significant reduction in zidovudine dose.

In the subgroup of patients whose pre-study endogenous serum erythropoietin levels were > 500 mUnits/mL, PROCRIT therapy did not reduce RBC transfusion requirements or increase hemoglobin compared to the corresponding responses in placebo-treated patients.

14.3 Patients with Cancer on Chemotherapy The safety and effectiveness of PROCRIT was assessed in two multicenter, randomized (1:1), placebo-controlled, double-blind studies (Study C1 and Study C2) and a pooled analysis of six additional randomized (1:1), multicenter, placebo-controlled, double-blind studies.

All studies were conducted in patients with anemia due to concomitantly administered cancer chemotherapy.

Study C1 enrolled 344 adult patients, Study C2 enrolled 222 pediatric patients, and the pooled analysis contained 131 patients randomized to epoetin alfa or placebo.

In Studies C1 and C2, efficacy was demonstrated by a reduction in the proportion of patients who received an RBC transfusion, from week 5 through end of the study, with the last-known RBC transfusion status carried forward for patients who discontinued treatment.

In the pooled analysis, efficacy was demonstrated by a reduction in the proportion of patients who received an RBC transfusion from week 5 through end of the study in the subset of patients who were remaining on therapy for 6 or more weeks.

Study C1 Study C1 was conducted in patients with anemia (hemoglobin < 11.5 g/dL for males; < 10.5 g/dL for females) with non-myeloid malignancies receiving myelosuppressive chemotherapy.

Randomization was stratified by type of malignancy (lung vs.

breast vs.

other), concurrent radiation therapy planned (yes or no), and baseline hemoglobin (< 9 g/dL vs.

≥ 9 g/dL); patients were randomized to epoetin alfa 40,000 Units (n=174) or placebo (n=170) as a weekly subcutaneous injection commencing on the first day of the chemotherapy cycle.

Ninety-one percent of patients were white, 44% were male, and the median age of patients was 66 years (range: 20 to 88 years).

The proportion of patients withdrawn from the study prior to week 5 was less than 10% for placebo-treated or epoetin-treated patients.

Per protocol, the last available hemoglobin values from patients who dropped out were included in the efficacy analyses.

Efficacy results are shown in Table 9.

Table 9: Study C1: Proportion of Patients Transfused Week 5 Through Week 16 or End of Study Last-known RBC transfusion status carried forward for patients who discontinued treatment.

Chemotherapy Regimen PROCRIT (n=174) Placebo (n=170) All Regimens 14% (25/174) Two-sided p < 0.001, logistic regression analysis adjusting for accrual rate and stratification variables.

28% (48/170) Regimens without cisplatin 14% (21/148) 26% (35/137) Regimens containing cisplatin 15% (4/26) 39% (13/33) Study C2 Study C2 was conducted in 222 patients with anemia, ages 5 to 18, receiving chemotherapy for the treatment of various childhood malignancies.

Randomization was stratified by cancer type (solid tumors, Hodgkin’s disease, acute lymphocytic leukemia, vs.

non-Hodgkin’s lymphoma); patients were randomized to receive epoetin alfa at 600 Units/kg maximum 40,000 Units (n=111) or placebo (n=111) as a weekly intravenous injection.

Sixty-nine percent of patients were white, 55% were male, and the median age of patients was 12 years (range: 5 to 18 years).

Two (2%) of placebo-treated patients and 3 (3%) of epoetin alfa-treated patients dropped out of the study prior to week 5.

There were fewer RBC transfusions from week 5 through the end-of-study in epoetin-alfa treated patients [51% (57/111)] compared to placebo-treated patients [69% (77/111)].

There was no evidence of an improvement in health-related quality of life, including no evidence of an effect on fatigue, energy, or strength in patients receiving PROCRIT as compared to those receiving placebo.

Pooled Analysis (Three Times Per Week Dosing) The results of 6 studies of similar design and that randomized 131 patients to epoetin alfa or placebo were pooled to assess the safety and effectiveness of epoetin alfa.

Patients were randomized to receive epoetin alfa at 150 Units/kg (n=63) or placebo (n=68), subcutaneously three times per week for 12 weeks in each study.

Across all studies, 72 patients were treated with concomitant non cisplatin-containing chemotherapy regimens and 59 patients were treated with concomitant cisplatin-containing chemotherapy regimens.

Twelve patients (19%) in the epoetin alfa arm and 10 patients (15%) in the placebo-arm dropped out prior to week 6 and are excluded from efficacy analyses.

Table 10: Proportion of Patients Transfused in the Pooled Analysis for Three Times Per Week Dosing Week 5 Through Week 12 or End of Study Limited to patients remaining on study beyond week 6 and includes only RBC transfusions during weeks 5–12.

Chemotherapy Regimen PROCRIT Placebo All Regimens 22% (11/51) Two-sided p < 0.05, unadjusted.

43% (25/58) Regimens without cisplatin 21% (6/29) 33% (11/33) Regimens containing cisplatin 23% (5/22) 56% (14/25) 14.4 Surgery Patients The safety and efficacy of PROCRIT were evaluated in a placebo-controlled, double-blind study (S1) enrolling 316 patients scheduled for major, elective orthopedic hip or knee surgery who were expected to require ≥ 2 units of blood and who were not able or willing to participate in an autologous blood donation program.

Patients were stratified into 1 of 3 groups based on their pretreatment hemoglobin [≤ 10 g/dL (n=2), > 10 to ≤ 13 g/dL (n=96), and > 13 to ≤ 15 g/dL (n=218)] and then randomly assigned to receive 300 Units/kg PROCRIT, 100 Units/kg PROCRIT, or placebo by subcutaneous injection for 10 days before surgery, on the day of surgery, and for 4 days after surgery.

All patients received oral iron and a low-dose, postoperative warfarin regimen.

Treatment with PROCRIT 300 Units/kg significantly (p=0.024) reduced the risk of allogeneic RBC transfusion in patients with a pretreatment hemoglobin of > 10 to ≤ 13 g/dL; 5/31 (16%) of patients treated with PROCRIT 300 Units/kg, 6/26 (23%) of patients treated with PROCRIT 100 Units/kg, and 13/29 (45%) of placebo-treated patients were transfused.

There was no significant difference in the number of patients transfused between PROCRIT (9% 300 Units/kg, 6% 100 Units/kg) and placebo (13%) in the > 13 to ≤ 15 g/dL hemoglobin stratum.

There were too few patients in the ≤ 10 g/dL group to determine if PROCRIT is useful in this hemoglobin strata.

In the > 10 to ≤ 13 g/dL pretreatment stratum, the mean number of units transfused per PROCRIT-treated patient (0.45 units blood for 300 Units/kg, 0.42 units blood for 100 Units/kg) was less than the mean transfused per placebo-treated patient (1.14 units) (overall p=0.028).

In addition, mean hemoglobin, hematocrit, and reticulocyte counts increased significantly during the presurgery period in patients treated with PROCRIT.

PROCRIT was also evaluated in an open-label, parallel-group study (S2) enrolling 145 patients with a pretreatment hemoglobin level of ≥ 10 to ≤ 13 g/dL who were scheduled for major orthopedic hip or knee surgery and who were not participating in an autologous program.

Patients were randomly assigned to receive 1 of 2 subcutaneous dosing regimens of PROCRIT (600 Units/kg once weekly for 3 weeks prior to surgery and on the day of surgery, or 300 Units/kg once daily for 10 days prior to surgery, on the day of surgery, and for 4 days after surgery).

All patients received oral iron and appropriate pharmacologic anticoagulation therapy.

From pretreatment to presurgery, the mean increase in hemoglobin in the 600 Units/kg weekly group (1.44 g/dL) was greater than that observed in the 300 Units/kg daily group.

The mean increase in absolute reticulocyte count was smaller in the weekly group (0.11 × 10 6 /mm 3 ) compared to the daily group (0.17 × 10 6 /mm 3 ).

Mean hemoglobin levels were similar for the 2 treatment groups throughout the postsurgical period.

The erythropoietic response observed in both treatment groups resulted in similar RBC transfusion rates [11/69 (16%) in the 600 Units/kg weekly group and 14/71 (20%) in the 300 Units/kg daily group].

The mean number of units transfused per patient was approximately 0.3 units in both treatment groups.

HOW SUPPLIED

16 /STORAGE AND HANDLING PROCRIT (epoetin alfa) injection is a sterile, clear, and colorless solution available as: Preservative-free, single-dose vials (in citrate-buffered formulation) : 2,000 Units/mL (NDC 59676-302-01), 3,000 Units/mL (NDC 59676-303-01), 4,000 Units/mL (NDC 59676-304-01), or 10,000 Units/mL (NDC 59676-310-01) supplied in cartons, each carton containing six 1 mL single-dose vials.

Preservative-free, single-dose vials (in citrate-buffered formulation) : 10,000 Units/mL (NDC 59676-310-02) supplied in dispensing packs (tray) containing 25 single-dose 1 mL vials.

Preservative-free, single-dose vials (in phosphate-buffered formulation) : 40,000 Units/mL (NDC 59676-340-01) supplied in dispensing packs containing four 1 mL single-dose vials.

Preserved, multiple-dose vials : 20,000 Units/2mL (10,000 Units/mL) (NDC 59676-312-04) supplied in dispensing packs containing four 2 mL multiple-dose vials.

Preserved, multiple-dose vials : 20,000 Units/mL (NDC 59676-320-04) supplied in dispensing packs containing four 1 mL multiple-dose vials.

Store at 36°F to 46°F (2°C to 8°C).

Do not freeze.

Do not shake.

Do not use PROCRIT that has been shaken or frozen.

Store PROCRIT vials in the original carton until use to protect from light.

GERIATRIC USE

8.5 Geriatric Use Of the 4553 patients who received PROCRIT in the 6 studies for treatment of anemia due to CKD not receiving dialysis, 2726 (60%) were age 65 years and over, while 1418 (31%) were 75 years and over.

Of the 757 patients who received PROCRIT in the 3 studies of CKD patients on dialysis, 361 (47%) were age 65 years and over, while 100 (13%) were 75 years and over.

No differences in safety or effectiveness were observed between geriatric and younger patients.

Dose selection and adjustment for an elderly patient should be individualized to achieve and maintain the target hemoglobin [see Dosage and Administration (2) ] .

Among 778 patients enrolled in the 3 clinical studies of PROCRIT for the treatment of anemia due to concomitant chemotherapy, 419 received PROCRIT and 359 received placebo.

Of the 419 who received PROCRIT, 247 (59%) were age 65 years and over, while 78 (19%) were 75 years and over.

No overall differences in safety or effectiveness were observed between geriatric and younger patients.

The dose requirements for PROCRIT in geriatric and younger patients within the 3 studies were similar.

Among 1731 patients enrolled in the 6 clinical studies of PROCRIT for reduction of allogeneic RBC transfusions in patients undergoing elective surgery, 1085 received PROCRIT and 646 received placebo or standard of care treatment.

Of the 1085 patients who received PROCRIT, 582 (54%) were age 65 years and over, while 245 (23%) were 75 years and over.

No overall differences in safety or effectiveness were observed between geriatric and younger patients.

The dose requirements for PROCRIT in geriatric and younger patients within the 4 studies using the 3 times weekly schedule and 2 studies using the weekly schedule were similar.

Insufficient numbers of patients age 65 years or older were enrolled in clinical studies of PROCRIT for the treatment of patients treated with zidovudine for HIV Infection to determine whether they respond differently from younger patients .

DOSAGE FORMS AND STRENGTHS

3 Injection: 2,000 Units/mL, 3,000 Units/mL, 4,000 Units/mL, 10,000 Units/mL, and 40,000 Units/mL of PROCRIT as a clear and colorless liquid in single-dose vials 20,000 Units/2 mL (10,000 Units/mL) and 20,000 Units/mL of PROCRIT as a clear and colorless liquid in multiple-dose vials (contains benzyl alcohol) Injection 2,000 Units/mL, 3,000 Units/mL, 4,000 Units/mL, 10,000 Units/mL, and 40,000 Units/mL in single-dose vials ( 3 ) 20,000 Units/2 mL (10,000 Units/mL) and 20,000 Units/mL in multiple-dose vials containing benzyl alcohol ( 3 )

MECHANISM OF ACTION

12.1 Mechanism of Action PROCRIT stimulates erythropoiesis by the same mechanism as endogenous erythropoietin.

INDICATIONS AND USAGE

1 PROCRIT is an erythropoiesis-stimulating agent (ESA) indicated for: Treatment of anemia due to Chronic Kidney Disease (CKD) in patients on dialysis and not on dialysis ( 1.1 ).

Zidovudine in patients with Human Immunodeficiency Virus (HIV) infection ( 1.2 ).

The effects of concomitant myelosuppressive chemotherapy, and upon initiation, there is a minimum of two additional months of planned chemotherapy ( 1.3 ).

Reduction of allogeneic red blood cell (RBC) transfusions in patients undergoing elective, noncardiac, nonvascular surgery ( 1.4 ).

Limitations of Use PROCRIT has not been shown to improve quality of life, fatigue, or patient well-being ( 1.5 ).

PROCRIT is not indicated for use: In patients with cancer receiving hormonal agents, biologic products, or radiotherapy, unless also receiving concomitant myelosuppressive chemotherapy ( 1.5 ).

In patients with cancer receiving myelosuppressive chemotherapy when the anticipated outcome is cure ( 1.5 ).

In patients with cancer receiving myelosuppressive chemotherapy in whom the anemia can be managed by transfusion ( 1.5 ).

In patients scheduled for surgery who are willing to donate autologous blood ( 1.5 ).

In patients undergoing cardiac or vascular surgery ( 1.5 ).

As a substitute for RBC transfusions in patients who require immediate correction of anemia ( 1.5 ).

1.1 Anemia Due to Chronic Kidney Disease PROCRIT is indicated for the treatment of anemia due to chronic kidney disease (CKD), including patients on dialysis and not on dialysis to decrease the need for red blood cell (RBC) transfusion.

1.2 Anemia Due to Zidovudine in Patients with HIV Infection PROCRIT is indicated for the treatment of anemia due to zidovudine administered at ≤ 4200 mg/week in patients with HIV Infection with endogenous serum erythropoietin levels of ≤ 500 mUnits/mL.

1.3 Anemia Due to Chemotherapy in Patients with Cancer PROCRIT is indicated for the treatment of anemia in patients with non-myeloid malignancies where anemia is due to the effect of concomitant myelosuppressive chemotherapy, and upon initiation, there is a minimum of two additional months of planned chemotherapy.

1.4 Reduction of Allogeneic Red Blood Cell Transfusions in Patients Undergoing Elective, Noncardiac, Nonvascular Surgery PROCRIT is indicated to reduce the need for allogeneic RBC transfusions among patients with perioperative hemoglobin > 10 to ≤ 13 g/dL who are at high risk for perioperative blood loss from elective, noncardiac, nonvascular surgery.

PROCRIT is not indicated for patients who are willing to donate autologous blood pre-operatively.

1.5 Limitations of Use PROCRIT has not been shown to improve quality of life, fatigue, or patient well-being.

PROCRIT is not indicated for use: In patients with cancer receiving hormonal agents, biologic products, or radiotherapy, unless also receiving concomitant myelosuppressive chemotherapy.

In patients with cancer receiving myelosuppressive chemotherapy when the anticipated outcome is cure.

In patients with cancer receiving myelosuppressive chemotherapy in whom the anemia can be managed by transfusion.

In patients scheduled for surgery who are willing to donate autologous blood.

In patients undergoing cardiac or vascular surgery.

As a substitute for RBC transfusions in patients who require immediate correction of anemia.

PEDIATRIC USE

8.4 Pediatric Use The multiple-dose vials are formulated with benzyl alcohol and are contraindicated for use in neonates and infants [see Contraindications (4) , Warnings and Precautions (5.9) ] .

When therapy with PROCRIT is needed in neonates and infants, use the single-dose vial, which is a benzyl alcohol-free formulation.

Do not mix the single-dose vials with bacteriostatic saline when administering PROCRIT to neonates or infants because it contains benzyl alcohol [see Dosage and Administration (2.6) ] .

Serious adverse reactions including fatal reactions and the “gasping syndrome” occurred in premature neonates and infants in the neonatal intensive care unit who received drugs containing benzyl alcohol as a preservative.

In these cases, benzyl alcohol dosages of 99 to 234 mg/kg/day produced high levels of benzyl alcohol and its metabolites in the blood and urine (blood levels of benzyl alcohol were 0.61 to 1.378 mmol/L).

Additional adverse reactions included gradual neurological deterioration, seizures, intracranial hemorrhage, hematologic abnormalities, skin breakdown, hepatic and renal failure, hypotension, bradycardia, and cardiovascular collapse.

Preterm, low birth weight infants may be more likely to develop these reactions because they may be less able to metabolize benzyl alcohol.

The minimum amount of benzyl alcohol at which serious adverse reactions may occur is not known [see Warnings and Precautions (5.9) ] .

Pediatric Patients with CKD PROCRIT is indicated in pediatric patients, ages 1 month to 16 years of age, for the treatment of anemia associated with CKD requiring dialysis.

Safety and effectiveness in pediatric patients less than 1 month old have not been established [see Clinical Studies (14.1) ] .

Use of PROCRIT in pediatric patients with CKD not requiring dialysis is supported by efficacy in pediatric patients requiring dialysis.

The mechanism of action of PROCRIT is the same for these two populations.

Published literature also has reported the use of PROCRIT in pediatric patients with CKD not requiring dialysis.

Dose-dependent increases in hemoglobin and hematocrit were observed with reductions in transfusion requirements.

The safety data from the pediatric studies and postmarketing reports are similar to those obtained from the studies of PROCRIT in adult patients with CKD [see Warnings and Precautions (5) and Adverse Reactions (6.1) ] .

Postmarketing reports do not indicate a difference in safety profiles in pediatric patients with CKD requiring dialysis and not requiring dialysis.

Pediatric Patients with Cancer on Chemotherapy PROCRIT is indicated in patients 5 to 18 years old for the treatment of anemia due to concomitant myelosuppressive chemotherapy.

Safety and effectiveness in pediatric patients less than 5 years of age have not been established [see Clinical Studies (14.3) ].

The safety data from these studies are similar to those obtained from the studies of PROCRIT in adult patients with cancer [see Warnings and Precautions (5.1 , 5.2) and Adverse Reactions (6.1) ].

Pediatric Patients with HIV Infection Receiving Zidovudine Published literature has reported the use of PROCRIT in 20 zidovudine-treated, anemic, pediatric patients with HIV Infection, ages 8 months to 17 years, treated with 50 to 400 Units/kg subcutaneously or intravenously 2 to 3 times per week.

Increases in hemoglobin levels and in reticulocyte counts and decreases in or elimination of RBC transfusions were observed.

Pharmacokinetics in Neonates Limited pharmacokinetic data from a study of 7 preterm, very low birth weight neonates and 10 healthy adults given intravenous erythropoietin suggested that distribution volume was approximately 1.5 to 2 times higher in the preterm neonates than in the healthy adults, and clearance was approximately 3 times higher in the preterm neonates than in the healthy adults.

PREGNANCY

8.1 Pregnancy Risk Summary PROCRIT from multiple-dose vials contains benzyl alcohol and is contraindicated in pregnant women [see Contraindications (4) ] .

When therapy with PROCRIT is needed during pregnancy, use a benzyl alcohol-free formulation (i.e., single-dose vial).

Do not mix PROCRIT with bacteriostatic saline when administering to pregnant women because it contains benzyl alcohol (see Clinical Considerations ) [see Dosage and Administration (2.1) ] .

The limited available data on PROCRIT use in pregnant women are insufficient to determine a drug-associated risk of adverse developmental outcomes.

In animal reproductive and developmental toxicity studies, adverse fetal effects including embryo-fetal death, skeletal anomalies, and growth defects occurred when pregnant rats received epoetin alfa at doses approximating the clinical recommended starting doses (see Data ) .

Consider the benefits and risks of PROCRIT single-dose vials for the mother and possible risks to the fetus when prescribing PROCRIT to a pregnant woman.

The estimated background risk of major birth defects and miscarriage for the indicated population 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 risks of major birth defects and miscarriage in clinically recognized pregnancies is 2–4% and 15–20%, respectively.

Clinical Considerations Fetal/Neonatal Adverse Reactions The multiple-dose vials of PROCRIT contain benzyl alcohol.

The preservative benzyl alcohol has been associated with serious adverse reactions and death when administered intravenously to neonates and infants [see Warnings and Precautions (5.9) , Use in Specific Populations (8.4) ].

There is a potential for similar risks to fetuses exposed to benzyl alcohol in utero .

Data Human Data There are reports of pregnant women with anemia alone or anemia associated with severe renal disease and other hematologic disorders who received PROCRIT.

Polyhydramnios and intrauterine growth restriction were reported in women with chronic renal disease, which is associated with an increased risk for these adverse pregnancy outcomes.

Due to the limited number of exposed pregnancies and multiple confounding factors (such as underlying maternal conditions, other maternal medications, and gestational timing of exposure), these published case reports and studies do not reliably estimate the frequency, presence or absence of adverse outcomes.

Animal Data When rats received PROCRIT at doses greater than or equal to 100 Units/kg/day during mating and through early pregnancy (dosing stopped prior to organogenesis), there were slight increases in the incidences of pre- and post-implantation loss, and a decrease in live fetuses in the presence of maternal toxicity (red limbs/pinna, focal splenic capsular toxicity, increased organ weights).

This animal dose level of 100 Units/kg/day may approximate the clinical recommended starting dose, depending on the treatment indication.

When pregnant rats and rabbits received intravenous doses of up to 500 mg/kg/day of PROCRIT only during organogenesis (gestational days 7 to 17 in rats and gestational days 6 to 18 in rabbits), no teratogenic effects were observed in the offspring.

The offspring (F1 generation) of the treated rats were observed postnatally; rats from the F1 generation reached maturity and were mated; no PROCRIT-related effects were apparent for their offspring (F2 generation fetuses).

When pregnant rats received PROCRIT at doses of 500 Units/kg/day late in pregnancy (after the period of organogenesis from day 17 of gestation through day 21 of lactation), pups exhibited decreased number of caudal vertebrae, decreased body weight gain, and delayed appearance of abdominal hair, eyelid opening, and ossification in the presence of maternal toxicity (red limbs/pinna, increased organ weights).

This animal dose level of 500 U/kg/day is approximately five times the clinical recommended starting dose depending on the patient’s treatment indication.

BOXED WARNING

WARNING: ESAs INCREASE THE RISK OF DEATH, MYOCARDIAL INFARCTION, STROKE, VENOUS THROMBOEMBOLISM, THROMBOSIS OF VASCULAR ACCESS AND TUMOR PROGRESSION OR RECURRENCE WARNING: ESAs INCREASE THE RISK OF DEATH, MYOCARDIAL INFARCTION, STROKE, VENOUS THROMBOEMBOLISM, THROMBOSIS OF VASCULAR ACCESS AND TUMOR PROGRESSION OR RECURRENCE See full prescribing information for complete boxed warning.

Chronic Kidney Disease: In controlled trials, patients experienced greater risks for death, serious adverse cardiovascular reactions, and stroke when administered erythropoiesis-stimulating agents (ESAs) to target a hemoglobin level of greater than 11 g/dL ( 5.1 ).

No trial has identified a hemoglobin target level, ESA dose, or dosing strategy that does not increase these risks ( 2.2 ).

Use the lowest PROCRIT dose sufficient to reduce the need for red blood cell (RBC) transfusions ( 5.1 ).

Cancer: ESAs shortened overall survival and/or increased the risk of tumor progression or recurrence in clinical studies of patients with breast, non-small cell lung, head and neck, lymphoid, and cervical cancers ( 5.2 ).

Use the lowest dose to avoid RBC transfusions ( 2.4 ).

Use ESAs only for anemia from myelosuppressive chemotherapy ( 1.3 ).

ESAs are not indicated for patients receiving myelosuppressive chemotherapy when the anticipated outcome is cure ( 1.5 ).

Discontinue following the completion of a chemotherapy course ( 2.4 ).

Perisurgery: Due to increased risk of deep venous thrombosis (DVT), DVT prophylaxis is recommended ( 5.1 ).

Chronic Kidney Disease: In controlled trials, patients with chronic kidney disease (CKD) experienced greater risks for death, serious adverse cardiovascular reactions, and stroke when administered erythropoiesis-stimulating agents (ESAs) to target a hemoglobin level of greater than 11 g/dL [see Warnings and Precautions (5.1) ] .

No trial has identified a hemoglobin target level, ESA dose, or dosing strategy that does not increase these risks [see Dosage and Administration (2.2) ] .

Use the lowest PROCRIT dose sufficient to reduce the need for red blood cell (RBC) transfusions [see Warnings and Precautions (5.1) ] .

Cancer: ESAs shortened overall survival and/or increased the risk of tumor progression or recurrence in clinical studies of patients with breast, non-small cell lung, head and neck, lymphoid, and cervical cancers [see Warnings and Precautions (5.2) ] .

To decrease these risks, as well as the risk of serious cardiovascular and thromboembolic reactions, use the lowest dose needed to avoid RBC transfusions [see Dosage and Administration (2.4) ] .

Use ESAs only for anemia from myelosuppressive chemotherapy [see Indications and Usage (1.3) ] .

ESAs are not indicated for patients receiving myelosuppressive chemotherapy when the anticipated outcome is cure [see Indications and Usage (1.5) ] .

Discontinue following the completion of a chemotherapy course [see Dosage and Administration (2.4) ] .

Perisurgery: Due to increased risk of Deep Venous Thrombosis (DVT), DVT prophylaxis is recommended [see Dosage and Administration (2.5) , Warnings and Precautions (5.1) ] .

WARNING AND CAUTIONS

5 WARNINGS AND PRECAUTIONS Increased Mortality, Myocardial Infarction, Stroke, and Thromboembolism: Using ESAs to target a hemoglobin level of greater than 11 g/dL increases the risk of serious adverse cardiovascular reactions and has not been shown to provide additional benefit ( 5.1 and 14.1 ).

Use caution in patients with coexistent cardiovascular disease and stroke ( 5.1 ).

Increased Mortality and/or Increased Risk of Tumor Progression or Recurrence in Patients with Cancer ( 5.2 ).

Hypertension: Control hypertension prior to initiating and during treatment with PROCRIT ( 5.3 ).

Seizures: PROCRIT increases the risk for seizures in patients with CKD ( 5.4 ).

Increase monitoring of these patients for changes in seizure frequency or premonitory symptoms ( 5.4 ).

PRCA: If severe anemia and low reticulocyte count develop during PROCRIT treatment, withhold PROCRIT and evaluate for PRCA ( 5.6 ).

Serious Allergic Reactions: Discontinue PROCRIT and manage reactions ( 5.7 ).

Severe Cutaneous Reactions: Discontinue PROCRIT ( 5.8 ).

5.1 Increased Mortality, Myocardial Infarction, Stroke, and Thromboembolism In controlled clinical trials of patients with CKD comparing higher hemoglobin targets (13 – 14 g/dL) to lower targets (9 – 11.3 g/dL), PROCRIT and other ESAs increased the risk of death, myocardial infarction, stroke, congestive heart failure, thrombosis of hemodialysis vascular access, and other thromboembolic events in the higher target groups.

Using ESAs to target a hemoglobin level of greater than 11 g/dL increases the risk of serious adverse cardiovascular reactions and has not been shown to provide additional benefit [see Clinical Studies (14.1) ] .

Use caution in patients with coexistent cardiovascular disease and stroke [see Dosage and Administration (2.2) ] .

Patients with CKD and an insufficient hemoglobin response to ESA therapy may be at even greater risk for cardiovascular reactions and mortality than other patients.

A rate of hemoglobin rise of greater than 1 g/dL over 2 weeks may contribute to these risks.

In controlled clinical trials of patients with cancer, PROCRIT and other ESAs increased the risks for death and serious adverse cardiovascular reactions.

These adverse reactions included myocardial infarction and stroke.

In controlled clinical trials, ESAs increased the risk of death in patients undergoing coronary artery bypass graft surgery (CABG) and the risk of deep venous thrombosis (DVT) in patients undergoing orthopedic procedures.

The design and overall results of the 3 large trials comparing higher and lower hemoglobin targets are shown in Table 1.

Table 1: Randomized Controlled Trials Showing Adverse Cardiovascular Outcomes in Patients With CKD Normal Hematocrit Study (NHS) (N=1265) CHOIR (N=1432) TREAT (N=4038) Time Period of Trial 1993 to 1996 2003 to 2006 2004 to 2009 Population CKD patients on hemodialysis with coexisting CHF or CAD, hematocrit 30 ± 3% on epoetin alfa CKD patients not on dialysis with hemoglobin < 11 g/dL not previously administered epoetin alfa CKD patients not on dialysis with type II diabetes, hemoglobin ≤ 11 g/dL Hemoglobin Target; Higher vs.

Lower (g/dL) 14.0 vs.

10.0 13.5 vs.

11.3 13.0 vs.

≥ 9.0 Median (Q1, Q3) Achieved Hemoglobin level (g/dL) 12.6 (11.6, 13.3) vs.

10.3 (10.0, 10.7) 13.0 (12.2, 13.4) vs.

11.4 (11.1, 11.6) 12.5 (12.0, 12.8) vs.

10.6 (9.9, 11.3) Primary Endpoint All-cause mortality or non-fatal MI All-cause mortality, MI, hospitalization for CHF, or stroke All-cause mortality, MI, myocardial ischemia, heart failure, and stroke Hazard Ratio or Relative Risk (95% CI) 1.28 (1.06 – 1.56) 1.34 (1.03 – 1.74) 1.05 (0.94 – 1.17) Adverse Outcome for Higher Target Group All-cause mortality All-cause mortality Stroke Hazard Ratio or Relative Risk (95% CI) 1.27 (1.04 – 1.54) 1.48 (0.97 – 2.27) 1.92 (1.38 – 2.68) Patients with Chronic Kidney Disease Normal Hematocrit Study (NHS): A prospective, randomized, open-label study of 1265 patients with chronic kidney disease on dialysis with documented evidence of congestive heart failure or ischemic heart disease was designed to test the hypothesis that a higher target hematocrit (Hct) would result in improved outcomes compared with a lower target Hct.

In this study, patients were randomized to epoetin alfa treatment targeted to a maintenance hemoglobin of either 14 ± 1 g/dL or 10 ± 1 g/dL.

The trial was terminated early with adverse safety findings of higher mortality in the high hematocrit target group.

Higher mortality (35% vs.

29%) was observed for the patients randomized to a target hemoglobin of 14 g/dL than for the patients randomized to a target hemoglobin of 10 g/dL.

For all-cause mortality, the HR=1.27; 95% CI (1.04, 1.54); p=0.018.

The incidence of nonfatal myocardial infarction, vascular access thrombosis, and other thrombotic events was also higher in the group randomized to a target hemoglobin of 14 g/dL.

CHOIR: A randomized, prospective trial, 1432 patients with anemia due to CKD who were not undergoing dialysis and who had not previously received epoetin alfa therapy were randomized to epoetin alfa treatment targeting a maintenance hemoglobin concentration of either 13.5 g/dL or 11.3 g/dL.

The trial was terminated early with adverse safety findings.

A major cardiovascular event (death, myocardial infarction, stroke, or hospitalization for congestive heart failure) occurred in 125 of the 715 patients (18%) in the higher hemoglobin group compared to 97 of the 717 patients (14%) in the lower hemoglobin group [hazard ratio (HR) 1.34, 95% CI: 1.03, 1.74; p=0.03].

TREAT: A randomized, double-blind, placebo-controlled, prospective trial of 4038 patients with: CKD not on dialysis (eGFR of 20 – 60 mL/min), anemia (hemoglobin levels ≤ 11 g/dL), and type 2 diabetes mellitus, patients were randomized to receive either darbepoetin alfa treatment or a matching placebo.

Placebo group patients also received darbepoetin alfa when their hemoglobin levels were below 9 g/dL.

The trial objectives were to demonstrate the benefit of darbepoetin alfa treatment of the anemia to a target hemoglobin level of 13 g/dL, when compared to a “placebo” group, by reducing the occurrence of either of two primary endpoints: (1) a composite cardiovascular endpoint of all-cause mortality or a specified cardiovascular event (myocardial ischemia, CHF, MI, and CVA) or (2) a composite renal endpoint of all-cause mortality or progression to end stage renal disease.

The overall risks for each of the two primary endpoints (the cardiovascular composite and the renal composite) were not reduced with darbepoetin alfa treatment (see Table 1 ), but the risk of stroke was increased nearly two-fold in the darbepoetin alfa -treated group versus the placebo group: annualized stroke rate 2.1% vs.

1.1%, respectively, HR 1.92; 95% CI: 1.38, 2.68; p < 0.001.

The relative risk of stroke was particularly high in patients with a prior stroke: annualized stroke rate 5.2% in the darbepoetin alfa-treated group and 1.9% in the placebo group, HR 3.07; 95% CI: 1.44, 6.54.

Also, among darbepoetin alfa-treated subjects with a past history of cancer, there were more deaths due to all causes and more deaths adjudicated as due to cancer, in comparison with the control group.

Patients with Cancer An increased incidence of thromboembolic reactions, some serious and life-threatening, occurred in patients with cancer treated with ESAs.

In a randomized, placebo-controlled study (Study 2 in Table 2 [see Warnings and Precautions (5.2) ] ) of 939 women with metastatic breast cancer receiving chemotherapy, patients received either weekly epoetin alfa or placebo for up to a year.

This study was designed to show that survival was superior when epoetin alfa was administered to prevent anemia (maintain hemoglobin levels between 12 and 14 g/dL or hematocrit between 36% and 42%).

This study was terminated prematurely when interim results demonstrated a higher mortality at 4 months (8.7% vs.

3.4%) and a higher rate of fatal thrombotic reactions (1.1% vs.

0.2%) in the first 4 months of the study among patients treated with epoetin alfa.

Based on Kaplan-Meier estimates, at the time of study termination, the 12-month survival was lower in the epoetin alfa group than in the placebo group (70% vs.

76%; HR 1.37, 95% CI: 1.07, 1.75; p=0.012).

Patients Having Surgery An increased incidence of deep venous thrombosis (DVT) in patients receiving epoetin alfa undergoing surgical orthopedic procedures was demonstrated [see Adverse Reactions (6.1) ] .

In a randomized, controlled study, 680 adult patients, not receiving prophylactic anticoagulation and undergoing spinal surgery, were randomized to 4 doses of 600 Units/kg epoetin alfa (7, 14, and 21 days before surgery, and the day of surgery) and standard of care (SOC) treatment (n=340) or to SOC treatment alone (n=340).

A higher incidence of DVTs, determined by either color flow duplex imaging or by clinical symptoms, was observed in the epoetin alfa group (16 [4.7%] patients) compared with the SOC group (7 [2.1%] patients).

In addition to the 23 patients with DVTs included in the primary analysis, 19 [2.8%] patients (n=680) experienced 1 other thrombovascular event (TVE) each (12 [3.5%] in the epoetin alfa group and 7 [2.1%] in the SOC group).

Deep venous thrombosis prophylaxis is strongly recommended when ESAs are used for the reduction of allogeneic RBC transfusions in surgical patients [see Dosage and Administration (2.5) ] .

Increased mortality was observed in a randomized, placebo-controlled study of PROCRIT in adult patients who were undergoing CABG surgery (7 deaths in 126 patients randomized to PROCRIT versus no deaths among 56 patients receiving placebo).

Four of these deaths occurred during the period of study drug administration and all 4 deaths were associated with thrombotic events.

5.2 Increased Mortality and/or Increased Risk of Tumor Progression or Recurrence in Patients with Cancer ESAs resulted in decreased locoregional control/progression-free survival (PFS) and/or overall survival (OS) (see Table 2 ).

Adverse effects on PFS and/or OS were observed in studies of patients receiving chemotherapy for breast cancer (Studies 1, 2, and 4), lymphoid malignancy (Study 3), and cervical cancer (Study 5); in patients with advanced head and neck cancer receiving radiation therapy (Studies 6 and 7); and in patients with non-small cell lung cancer or various malignancies who were not receiving chemotherapy or radiotherapy (Studies 8 and 9).

Table 2: Randomized, Controlled Studies with Decreased Survival and/or Decreased Locoregional Control Study/Tumor/(n) Hemoglobin Target Achieved Hemoglobin (Median; Q1, Q3 Q1=25 th percentile, Q3=75 th percentile ) Primary Efficacy Outcome Adverse Outcome for ESA- containing Arm Chemotherapy Study 1 Metastatic breast cancer (n=2098) ≤12 g/dL This study did not include a defined hemoglobin target.

Doses were titrated to achieve and maintain the lowest hemoglobin level sufficient to avoid transfusion and not to exceed 12 g/dL.

11.6 g/dL 10.7, 12.1 g/dL Progression-free survival (PFS) Decreased progression-free and overall survival Study 2 Metastatic breast cancer (n=939) 12–14 g/dL 12.9 g/dL; 12.2, 13.3 g/dL 12-month overall survival Decreased 12-month survival Study 3 Lymphoid malignancy (n=344) 13–15 g/dL (M) 13–14 g/dL (F) 11 g/dL; 9.8, 12.1 g/dL Proportion of patients achieving a hemoglobin response Decreased overall survival Study 4 Early breast cancer (n=733) 12.5–13 g/dL 13.1 g/dL; 12.5, 13.7 g/dL Relapse-free and overall survival Decreased 3-year relapse-free and overall survival Study 5 Cervical cancer (n=114) 12–14 g/dL 12.7 g/dL; 12.1, 13.3 g/dL Progression-free and overall survival and locoregional control Decreased 3-year progression-free and overall survival and locoregional control Radiotherapy Alone Study 6 Head and neck cancer (n=351) ≥ 15 g/dL (M) ≥ 14 g/dL (F) Not available Locoregional progression-free survival Decreased 5-year locoregional progression-free and overall survival Study 7 Head and neck cancer (n=522) 14–15.5 g/dL Not available Locoregional disease control Decreased locoregional disease control No Chemotherapy or Radiotherapy Study 8 Non-small cell lung cancer (n=70) 12–14 g/dL Not available Quality of life Decreased overall survival Study 9 Non-myeloid malignancy (n=989) 12–13 g/dL 10.6 g/dL; 9.4, 11.8 g/dL RBC transfusions Decreased overall survival Decreased Overall Survival Study 2 was described in the previous section [see Warnings and Precautions (5.1) ] .

Mortality at 4 months (8.7% vs.

3.4%) was significantly higher in the epoetin alfa arm.

The most common investigator-attributed cause of death within the first 4 months was disease progression; 28 of 41 deaths in the epoetin alfa arm and 13 of 16 deaths in the placebo arm were attributed to disease progression.

Investigator-assessed time to tumor progression was not different between the 2 groups.

Survival at 12 months was significantly lower in the epoetin alfa arm (70% vs.

76%; HR 1.37, 95% CI: 1.07, 1.75; p=0.012).

Study 3 was a randomized, double-blind study (darbepoetin alfa vs.

placebo) conducted in 344 anemic patients with lymphoid malignancy receiving chemotherapy.

With a median follow-up of 29 months, overall mortality rates were significantly higher among patients randomized to darbepoetin alfa as compared to placebo (HR 1.36, 95% CI: 1.02, 1.82).

Study 8 was a multicenter, randomized, double-blind study (epoetin alfa vs.

placebo) in which patients with advanced non-small cell lung cancer receiving only palliative radiotherapy or no active therapy were treated with epoetin alfa to achieve and maintain hemoglobin levels between 12 and 14 g/dL.

Following an interim analysis of 70 patients (planned accrual 300 patients), a significant difference in survival in favor of the patients in the placebo arm of the study was observed (median survival 63 vs.

129 days; HR 1.84; p=0.04).

Study 9 was a randomized, double-blind study (darbepoetin alfa vs.

placebo) in 989 anemic patients with active malignant disease, neither receiving nor planning to receive chemotherapy or radiation therapy.

There was no evidence of a statistically significant reduction in proportion of patients receiving RBC transfusions.

The median survival was shorter in the darbepoetin alfa treatment group than in the placebo group (8 months vs.

10.8 months; HR 1.30, 95% CI: 1.07, 1.57).

Decreased Progression-free Survival and Overall Survival Study 1 was a randomized, open-label, multicenter study in 2,098 anemic women with metastatic breast cancer, who received first line or second line chemotherapy.

This was a noninferiority study designed to rule out a 15% risk increase in tumor progression or death of epoetin alfa plus standard of care (SOC) as compared with SOC alone.

At the time of clinical data cutoff, the median progression free survival (PFS) per investigator assessment of disease progression was 7.4 months in each arm (HR 1.09, 95% CI: 0.99, 1.20), indicating the study objective was not met.

There were more deaths from disease progression in the epoetin alfa plus SOC arm (59% vs.

56%) and more thrombotic vascular events in the epoetin alfa plus SOC arm (3% vs.

1%).

At the final analysis, 1653 deaths were reported (79.8% subjects in the epoetin alfa plus SOC group and 77.8% subjects in the SOC group).

Median overall survival in the epoetin alfa plus SOC group was 17.8 months compared with 18.0 months in the SOC alone group (HR 1.07, 95% CI: 0.97, 1.18).

Study 4 was a randomized, open-label, controlled, factorial design study in which darbepoetin alfa was administered to prevent anemia in 733 women receiving neo-adjuvant breast cancer treatment.

A final analysis was performed after a median follow-up of approximately 3 years.

The 3-year survival rate was lower (86% vs.

90%; HR 1.42, 95% CI: 0.93, 2.18) and the 3-year relapse-free survival rate was lower (72% vs.

78%; HR 1.33, 95% CI: 0.99, 1.79) in the darbepoetin alfa-treated arm compared to the control arm.

Study 5 was a randomized, open-label, controlled study that enrolled 114 of a planned 460 cervical cancer patients receiving chemotherapy and radiotherapy.

Patients were randomized to receive epoetin alfa to maintain hemoglobin between 12 and 14 g/dL or to RBC transfusion support as needed.

The study was terminated prematurely due to an increase in thromboembolic adverse reactions in epoetin alfa-treated patients compared to control (19% vs.

9%).

Both local recurrence (21% vs.

20%) and distant recurrence (12% vs.

7%) were more frequent in epoetin alfa-treated patients compared to control.

Progression-free survival at 3 years was lower in the epoetin alfa-treated group compared to control (59% vs.

62%; HR 1.06, 95% CI: 0.58, 1.91).

Overall survival at 3 years was lower in the epoetin alfa-treated group compared to control (61% vs.

71%; HR 1.28, 95% CI: 0.68, 2.42).

Study 6 was a randomized, placebo-controlled study in 351 head and neck cancer patients where epoetin beta or placebo was administered to achieve target hemoglobins ≥ 14 and ≥ 15 g/dL for women and men, respectively.

Locoregional progression-free survival was significantly shorter in patients receiving epoetin beta (HR 1.62, 95% CI: 1.22, 2.14; p=0.0008) with medians of 406 days and 745 days in the epoetin beta and placebo arms, respectively.

Overall survival was significantly shorter in patients receiving epoetin beta (HR 1.39, 95% CI: 1.05, 1.84; p=0.02).

Decreased Locoregional Control Study 7 was a randomized, open-label, controlled study conducted in 522 patients with primary squamous cell carcinoma of the head and neck receiving radiation therapy alone (no chemotherapy) who were randomized to receive darbepoetin alfa to maintain hemoglobin levels of 14 to 15.5 g/dL or no darbepoetin alfa.

An interim analysis performed on 484 patients demonstrated that locoregional control at 5 years was significantly shorter in patients receiving darbepoetin alfa (RR 1.44, 95% CI: 1.06, 1.96; p=0.02).

Overall survival was shorter in patients receiving darbepoetin alfa (RR 1.28, 95% CI: 0.98, 1.68; p=0.08).

5.3 Hypertension PROCRIT is contraindicated in patients with uncontrolled hypertension.

Following initiation and titration of PROCRIT, approximately 25% of patients on dialysis required initiation of or increases in antihypertensive therapy; hypertensive encephalopathy and seizures have been reported in patients with CKD receiving PROCRIT.

Appropriately control hypertension prior to initiation of and during treatment with PROCRIT.

Reduce or withhold PROCRIT if blood pressure becomes difficult to control.

Advise patients of the importance of compliance with antihypertensive therapy and dietary restrictions [see Patient Counseling Information (17) ] .

5.4 Seizures PROCRIT increases the risk of seizures in patients with CKD.

During the first several months following initiation of PROCRIT, monitor patients closely for premonitory neurologic symptoms.

Advise patients to contact their healthcare practitioner for new-onset seizures, premonitory symptoms or change in seizure frequency.

5.5 Lack or Loss of Hemoglobin Response to PROCRIT For lack or loss of hemoglobin response to PROCRIT, initiate a search for causative factors (e.g., iron deficiency, infection, inflammation, bleeding).

If typical causes of lack or loss of hemoglobin response are excluded, evaluate for PRCA [see Warnings and Precautions (5.6) ] .

In the absence of PRCA, follow dosing recommendations for management of patients with an insufficient hemoglobin response to PROCRIT therapy [see Dosage and Administration (2.2) ] .

5.6 Pure Red Cell Aplasia Cases of PRCA and of severe anemia, with or without other cytopenias that arise following the development of neutralizing antibodies to erythropoietin have been reported in patients treated with PROCRIT.

This has been reported predominantly in patients with CKD receiving ESAs by subcutaneous administration.

PRCA has also been reported in patients receiving ESAs for anemia related to hepatitis C treatment (an indication for which PROCRIT is not approved).

If severe anemia and low reticulocyte count develop during treatment with PROCRIT, withhold PROCRIT and evaluate patients for neutralizing antibodies to erythropoietin.

Contact Janssen Products, LP at 1-800-JANSSEN (1-800-526-7736) to perform assays for binding and neutralizing antibodies.

Permanently discontinue PROCRIT in patients who develop PRCA following treatment with PROCRIT or other erythropoietin protein drugs.

Do not switch patients to other ESAs.

5.7 Serious Allergic Reactions Serious allergic reactions, including anaphylactic reactions, angioedema, bronchospasm, skin rash, and urticaria may occur with PROCRIT.

Immediately and permanently discontinue PROCRIT and administer appropriate therapy if a serious allergic or anaphylactic reaction occurs.

5.8 Severe Cutaneous Reactions Blistering and skin exfoliation reactions including Erythema multiforme and Stevens-Johnson Syndrome (SJS)/Toxic Epidermal Necrolysis (TEN), have been reported in patients treated with ESAs (including PROCRIT) in the postmarketing setting.

Discontinue PROCRIT therapy immediately if a severe cutaneous reaction, such as SJS/TEN, is suspected.

5.9 Risk of Serious Adverse Reactions Due to Benzyl Alcohol Preservative PROCRIT from multiple-dose vials contains benzyl alcohol and is contraindicated for use in neonates, infants, pregnant women, and lactating women [see Contraindications (4) ] .

In addition, do not mix PROCRIT with bacteriostatic saline (which also contains benzyl alcohol) when administering PROCRIT to these patient populations [see Dosage and Administration (2) ].

Serious and fatal reactions including “gasping syndrome” can occur in neonates and infants treated with benzyl alcohol-preserved drugs, including PROCRIT multiple-dose vials.

The “gasping syndrome” is characterized by central nervous system (CNS) depression, metabolic acidosis, and gasping respirations.

There is a potential for similar risks to fetuses and infants exposed to benzyl alcohol in utero or in breast-fed milk, respectively.

PROCRIT multiple-dose vials contain 11 mg of benzyl alcohol per mL.

The minimum amount of benzyl alcohol at which serious adverse reactions may occur is not known [see Use in Specific Populations (8.1 , 8.2 , and 8.4) ] .

5.10 Risk of Infectious Diseases Due to Albumin (Human) Content PROCRIT contains albumin, a derivative of human blood [see Description (11) ] .

Based on effective donor screening and product manufacturing processes, it carries an extremely remote risk for transmission of viral diseases.

A theoretical risk for transmission of Creutzfeldt-Jakob disease (CJD) also is considered extremely remote.

No cases of transmission of viral diseases or CJD have ever been identified for albumin.

5.11 Dialysis Management Patients may require adjustments in their dialysis prescriptions after initiation of PROCRIT.

Patients receiving PROCRIT may require increased anticoagulation with heparin to prevent clotting of the extracorporeal circuit during hemodialysis.

INFORMATION FOR PATIENTS

17 PATIENT COUNSELING INFORMATION Advise the patient to read the FDA-approved patient labeling (Medication Guide and Instructions for Use).

Inform patients: Of the increased risks of mortality, serious cardiovascular reactions, thromboembolic reactions, stroke, and tumor progression [see Warnings and Precautions (5.1 , 5.2) ] .

To undergo regular blood pressure monitoring, adhere to prescribed anti-hypertensive regimen and follow recommended dietary restrictions.

To contact their healthcare provider for new-onset neurologic symptoms or change in seizure frequency.

Of the need to have regular laboratory tests for hemoglobin.

Risks are associated with benzyl alcohol in neonates, infants, pregnant women, and lactating women [see Use in Specific Populations (8.1 , 8.2 , 8.4) ].

Instruct patients who self-administer PROCRIT of the: Importance of following the Instructions for Use.

Dangers of reusing needles, syringes, or unused portions of single-dose vials.

Proper disposal of used syringes, needles, and unused vials, and of the full container.

DOSAGE AND ADMINISTRATION

2 Evaluate iron status before and during treatment and maintain iron repletion.

Correct or exclude other causes of anemia before initiating treatment ( 2.1 ).

In pregnant women, lactating women, neonates, infants: Use only single-dose vials ( 2.1 ).

Patients with CKD: Initial dose: 50 to 100 Units/kg 3 times weekly (adults) and 50 Units/kg 3 times weekly (pediatric patients).

Individualize maintenance dose.

Intravenous route recommended for patients on hemodialysis ( 2.2 ).

Patients on Zidovudine due to HIV Infection: 100 Units/kg 3 times weekly ( 2.3 ).

Patients with Cancer on Chemotherapy: 40,000 Units weekly or 150 Units/kg 3 times weekly (adults); 600 Units/kg intravenously weekly (pediatric patients ≥ 5 years) ( 2.4 ).

Surgery Patients: 300 Units/kg per day daily for 15 days or 600 Units/kg weekly ( 2.5 ).

2.1 Important Dosing Information Evaluation of Iron Stores and Nutritional Factors Evaluate the iron status in all patients before and during treatment.

Administer supplemental iron therapy when serum ferritin is less than 100 mcg/L or when serum transferrin saturation is less than 20%.

The majority of patients with CKD will require supplemental iron during the course of ESA therapy.

Monitoring of Response to Therapy Correct or exclude other causes of anemia (e.g., vitamin deficiency, metabolic or chronic inflammatory conditions, bleeding, etc.) before initiating PROCRIT.

Following initiation of therapy and after each dose adjustment, monitor hemoglobin weekly until the hemoglobin level is stable and sufficient to minimize the need for RBC transfusion.

Selection of Formulation In pregnant women, lactating women, neonates, and infants use only single-dose vials (the benzyl alcohol-free formulation) [see Contraindications (4) and Use in Specific Populations (8.1 , 8.2 , and 8.4) ].

2.2 Patients with Chronic Kidney Disease In controlled trials, patients experienced greater risks for death, serious adverse cardiovascular reactions, and stroke when administered ESAs to target a hemoglobin level of greater than 11 g/dL.

No trial has identified a hemoglobin target level, ESA dose, or dosing strategy that does not increase these risks.

Individualize dosing and use the lowest dose of PROCRIT sufficient to reduce the need for RBC transfusions [see Warnings and Precautions (5.1) ] .

Physicians and patients should weigh the possible benefits of decreasing transfusions against the increased risks of death and other serious cardiovascular adverse reactions [see Boxed Warning and Clinical Studies (14) ].

For all patients with CKD: When initiating or adjusting therapy, monitor hemoglobin levels at least weekly until stable, then monitor at least monthly.

When adjusting therapy consider hemoglobin rate of rise, rate of decline, ESA responsiveness and hemoglobin variability.

A single hemoglobin excursion may not require a dosing change.

Do not increase the dose more frequently than once every 4 weeks.

Decreases in dose can occur more frequently.

Avoid frequent dose adjustments.

If the hemoglobin rises rapidly (e.g., more than 1 g/dL in any 2-week period), reduce the dose of PROCRIT by 25% or more as needed to reduce rapid responses.

For patients who do not respond adequately, if the hemoglobin has not increased by more than 1 g/dL after 4 weeks of therapy, increase the dose by 25%.

For patients who do not respond adequately over a 12-week escalation period, increasing the PROCRIT dose further is unlikely to improve response and may increase risks.

Use the lowest dose that will maintain a hemoglobin level sufficient to reduce the need for RBC transfusions.

Evaluate other causes of anemia.

Discontinue PROCRIT if responsiveness does not improve.

For adult patients with CKD on dialysis: Initiate PROCRIT treatment when the hemoglobin level is less than 10 g/dL.

If the hemoglobin level approaches or exceeds 11 g/dL, reduce or interrupt the dose of PROCRIT.

The recommended starting dose for adult patients is 50 to 100 Units/kg 3 times weekly intravenously or subcutaneously.

The intravenous route is recommended for patients on hemodialysis.

For adult patients with CKD not on dialysis: Consider initiating PROCRIT treatment only when the hemoglobin level is less than 10 g/dL and the following considerations apply: The rate of hemoglobin decline indicates the likelihood of requiring a RBC transfusion and , Reducing the risk of alloimmunization and/or other RBC transfusion-related risks is a goal If the hemoglobin level exceeds 10 g/dL, reduce or interrupt the dose of PROCRIT, and use the lowest dose of PROCRIT sufficient to reduce the need for RBC transfusions.

The recommended starting dose for adult patients is 50 to 100 Units/kg 3 times weekly intravenously or subcutaneously.

For pediatric patients with CKD: Initiate PROCRIT treatment only when the hemoglobin level is less than 10 g/dL.

If the hemoglobin level approaches or exceeds 12 g/dL, reduce or interrupt the dose of PROCRIT.

The recommended starting dose for pediatric patients (ages 1 month or older) is 50 Units/kg 3 times weekly intravenously or subcutaneously.

When treating patients who have chronic kidney disease and cancer, physicians should refer to Warnings and Precautions (5.1 and 5.2) .

2.3 Zidovudine-treated Patients with HIV Infection Starting Dose The recommended starting dose in adults is 100 Units/kg as an intravenous or subcutaneous injection 3 times per week.

Dose Adjustment If hemoglobin does not increase after 8 weeks of therapy, increase PROCRIT dose by approximately 50 to 100 Units/kg at 4- to 8-week intervals until hemoglobin reaches a level needed to avoid RBC transfusions or 300 Units/kg.

Withhold PROCRIT if hemoglobin exceeds 12 g/dL.

Resume therapy at a dose 25% below the previous dose when hemoglobin declines to less than 11 g/dL.

Discontinue PROCRIT if an increase in hemoglobin is not achieved at a dose of 300 Units/kg for 8 weeks.

2.4 Patients on Cancer Chemotherapy Initiate PROCRIT in patients on cancer chemotherapy only if the hemoglobin is less than 10 g/dL, and if there is a minimum of two additional months of planned chemotherapy.

Use the lowest dose of PROCRIT necessary to avoid RBC transfusions.

Recommended Starting Dose Adults: 150 Units/kg subcutaneously 3 times per week until completion of a chemotherapy course or 40,000 Units subcutaneously weekly until completion of a chemotherapy course.

Pediatric Patients (5 to 18 years): 600 Units/kg intravenously weekly until completion of a chemotherapy course.

Dose Reduction Reduce dose by 25% if: Hemoglobin increases greater than 1 g/dL in any 2-week period or Hemoglobin reaches a level needed to avoid RBC transfusion.

Withhold dose if hemoglobin exceeds a level needed to avoid RBC transfusion.

Reinitiate at a dose 25% below the previous dose when hemoglobin approaches a level where RBC transfusions may be required.

Dose Increase After the initial 4 weeks of PROCRIT therapy, if hemoglobin increases by less than 1 g/dL and remains below 10 g/dL, increase dose to: 300 Units/kg three times per week in adults or 60,000 Units weekly in adults 900 Units/kg (maximum 60,000 Units) weekly in pediatric patients After 8 weeks of therapy, if there is no response as measured by hemoglobin levels or if RBC transfusions are still required, discontinue PROCRIT.

2.5 Surgery Patients The recommended PROCRIT regimens are: 300 Units/kg per day subcutaneously for 15 days total: administered daily for 10 days before surgery, on the day of surgery, and for 4 days after surgery.

600 Units/kg subcutaneously in 4 doses administered 21, 14, and 7 days before surgery and on the day of surgery.

Deep venous thrombosis prophylaxis is recommended during PROCRIT therapy [see Warnings and Precautions (5.1) ] .

2.6 Preparation and Administration Do not shake.

Do not use PROCRIT that has been shaken or frozen.

Protect vials from light.

Parenteral drug products should be inspected visually for particulate matter and discoloration prior to administration.

Do not use any vials exhibiting particulate matter or discoloration.

Discard unused portions of PROCRIT in preservative-free vials.

Do not re-enter preservative-free vials.

Store unused portions of PROCRIT in multiple-dose vials at 36°F to 46°F (2°C to 8°C).

Discard 21 days after initial entry.

Do not dilute.

Do not mix with other drug solutions except for admixing as described below: Preservative-free PROCRIT from single-dose vials may be admixed in a syringe with bacteriostatic 0.9% sodium chloride injection, USP, with benzyl alcohol 0.9% (bacteriostatic saline) in a 1:1 ratio using aseptic technique at the time of administration.

Do not mix PROCRIT with bacteriostatic saline when administering to pregnant women, lactating women, neonates, and infants [see Use in Specific Populations (8.1 , 8.2 , 8.4) ].

Synthroid 0.05 MG Oral Tablet

WARNINGS

Boxed Warning WARNING: Thyroid hormones, including SYNTHROID, either alone or with other therapeutic agents, should not be used for the treatment of obesity or for weight loss.

In euthyroid patients, doses within the range of daily hormonal requirements are ineffective for weight reduction.

Larger doses may produce serious or even life threatening manifestations of toxicity, particularly when given in association with sympathomimetic amines such as those used for their anorectic effects.

Levothyroxine sodium should not be used in the treatment of male or female infertility unless this condition is associated with hypothyroidism.

In patients with nontoxic diffuse goiter or nodular thyroid disease, particularly the elderly or those with underlying cardiovascular disease, levothyroxine sodium therapy is contraindicated if the serum TSH level is already suppressed due to the risk of precipitating overt thyrotoxicosis (see CONTRAINDICATIONS ).

If the serum TSH level is not suppressed, SYNTHROID should be used with caution in conjunction with careful monitoring of thyroid function for evidence of hyperthyroidism and clinical monitoring for potential associated adverse cardiovascular signs and symptoms of hyperthyroidism.

DRUG INTERACTIONS

Drug Interactions Many drugs affect thyroid hormone pharmacokinetics and metabolism (e.g., absorption, synthesis, secretion, catabolism, protein binding, and target tissue response) and may alter the therapeutic response to SYNTHROID.

In addition, thyroid hormones and thyroid status have varied effects on the pharmacokinetics and actions of other drugs.

A listing of drug-thyroidal axis interactions is contained in Table 2 .

The list of drug-thyroidal axis interactions in Table 2 may not be comprehensive due to the introduction of new drugs that interact with the thyroidal axis or the discovery of previously unknown interactions.

The prescriber should be aware of this fact and should consult appropriate reference sources (e.g., package inserts of newly approved drugs, medical literature) for additional information if a drug-drug interaction with levothyroxine is suspected.

Table 2.

Drug-Thyroidal Axis Interactions Drug or Drug Class Effect Drugs that may reduce TSH secretion – the reduction is not sustained; therefore, hypothyroidism does not occur Dopamine/Dopamine Agonists Glucocorticoids Octreotide Use of these agents may result in a transient reduction in TSH secretion when administered at the following doses: Dopamine (≥ 1 mcg/kg/min); Glucocorticoids (hydrocortisone ≥ 100 mg/day or equivalent); Octreotide (> 100 mcg/day).

Drugs that alter thyroid hormone secretion Drugs that may decrease thyroid hormone secretion, which may result in hypothyroidism Aminoglutethimide Amiodarone Iodide (including iodine-containing radiographic contrast agents) Lithium Methimazole Propylthiouracil (PTU) Sulfonamides Tolbutamide Long-term lithium therapy can result in goiter in up to 50% of patients, and either subclinical or overt hypothyroidism, each in up to 20% of patients.

The fetus, neonate, elderly and euthyroid patients with underlying thyroid disease (e.g., Hashimoto’s thyroiditis or with Grave’s disease previously treated with radioiodine or surgery) are among those individuals who are particularly susceptible to iodine-induced hypothyroidism.

Oral cholecystographic agents and amiodarone are slowly excreted, producing more prolonged hypothyroidism than parenterally administered iodinated contrast agents.

Long-term aminoglutethimide therapy may minimally decrease T 4 and T 3 levels and increase TSH, although all values remain within normal limits in most patients.

Drugs that may increase thyroid hormone secretion, which may result in hyperthyroidism Amiodarone Iodide (including iodine-containing radiographic contrast agents) Iodide and drugs that contain pharmacologic amounts of iodide may cause hyperthyroidism in euthyroid patients with Grave’s disease previously treated with antithyroid drugs or in euthyroid patients with thyroid autonomy (e.g., multinodular goiter or hyperfunctioning thyroid adenoma).

Hyperthyroidism may develop over several weeks and may persist for several months after therapy discontinuation.

Amiodarone may induce hyperthyroidism by causing thyroiditis.

Drugs that may decrease T 4 absorption, which may result in hypothyroidism Antacids – Aluminum & Magnesium Hydroxides – Simethicone Bile Acid Sequestrants – Cholestyramine – Colestipol Calcium Carbonate Cation Exchange Resins – Kayexalate Ferrous Sulfate Orlistat Sucralfate Concurrent use may reduce the efficacy of levothyroxine by binding and delaying or preventing absorption, potentially resulting in hypothyroidism.

Calcium carbonate may form an insoluble chelate with levothyroxine, and ferrous sulfate likely forms a ferric-thyroxine complex.

Administer levothyroxine at least 4 hours apart from these agents.

Patients treated concomitantly with orlistat and levothyroxine should be monitored for changes in thyroid function.

Drugs that may alter T 4 and T 3 serum transport – but FT 4 concentration remains normal; and therefore, the patient remains euthyroid Drugs that may increase serum TBG concentration Drugs that may decrease serum TBG concentration Clofibrate Estrogen-containing oral contraceptives Estrogens (oral) Heroin / Methadone 5-Fluorouracil Mitotane Tamoxifen Androgens / Anabolic Steroids Asparaginase Glucocorticoids Slow-Release Nicotinic Acid Drugs that may cause protein-binding site displacement Furosemide (> 80 mg IV) Heparin Hydantoins Non Steroidal Anti-Inflammatory Drugs – Fenamates – Phenylbutazone Salicylates (> 2 g/day) Administration of these agents with levothyroxine results in an initial transient increase in FT 4 .

Continued administration results in a decrease in serum T 4 and normal FT 4 and TSH concentrations and, therefore, patients are clinically euthyroid.

Salicylates inhibit binding of T 4 and T 3 to TBG and transthyretin.

An initial increase in serum FT 4 is followed by return of FT 4 to normal levels with sustained therapeutic serum salicylate concentrations, although total-T 4 levels may decrease by as much as 30%.

Drugs that may alter T 4 and T 3 metabolism Drugs that may increase hepatic metabolism, which may result in hypothyroidism Carbamazepine Hydantoins Phenobarbital Rifampin Stimulation of hepatic microsomal drug-metabolizing enzyme activity may cause increased hepatic degradation of levothyroxine, resulting in increased levothyroxine requirements.

Phenytoin and carbamazepine reduce serum protein binding of levothyroxine, and total- and free- T 4 may be reduced by 20% to 40%, but most patients have normal serum TSH levels and are clinically euthyroid.

Drugs that may decrease T 4 5′-deiodinase activity Amiodarone Beta-adrenergic antagonists – (e.g., Propranolol > 160 mg/day) Glucocorticoids – (e.g., Dexamethasone ≥ 4 mg/day) Propylthiouracil (PTU) Administration of these enzyme inhibitors decreases the peripheral conversion of T 4 to T 3 , leading to decreased T 3 levels.

However, serum T 4 levels are usually normal but may occasionally be slightly increased.

In patients treated with large doses of propranolol (> 160 mg/day), T 3 and T 4 levels change slightly, TSH levels remain normal, and patients are clinically euthyroid.

It should be noted that actions of particular beta-adrenergic antagonists may be impaired when the hypothyroid patient is converted to the euthyroid state.

Short-term administration of large doses of glucocorticoids may decrease serum T 3 concentrations by 30% with minimal change in serum T 4 levels.

However, long-term glucocorticoid therapy may result in slightly decreased T 3 and T 4 levels due to decreased TBG production (see above).

Miscellaneous Anticoagulants (oral) – Coumarin Derivatives – Indandione Derivatives Thyroid hormones appear to increase the catabolism of vitamin K-dependent clotting factors, thereby increasing the anticoagulant activity of oral anticoagulants.

Concomitant use of these agents impairs the compensatory increases in clotting factor synthesis.

Prothrombin time should be carefully monitored in patients taking levothyroxine and oral anticoagulants and the dose of anticoagulant therapy adjusted accordingly.

Antidepressants – Tricyclics (e.g., Amitriptyline) – Tetracyclics (e.g., Maprotiline) – Selective Serotonin Reuptake Inhibitors (SSRIs; e.g., Sertraline) Concurrent use of tri/tetracyclic antidepressants and levothyroxine may increase the therapeutic and toxic effects of both drugs, possibly due to increased receptor sensitivity to catecholamines.

Toxic effects may include increased risk of cardiac arrhythmias and CNS stimulation; onset of action of tricyclics may be accelerated.

Administration of sertraline in patients stabilized on levothyroxine may result in increased levothyroxine requirements.

Antidiabetic Agents – Biguanides – Meglitinides – Sulfonylureas – Thiazolidinediones – Insulin Addition of levothyroxine to antidiabetic or insulin therapy may result in increased antidiabetic agent or insulin requirements.

Careful monitoring of diabetic control is recommended, especially when thyroid therapy is started, changed, or discontinued.

Cardiac Glycosides Serum digitalis glycoside levels may be reduced in hyperthyroidism or when the hypothyroid patient is converted to the euthyroid state.

Therapeutic effect of digitalis glycosides may be reduced.

Cytokines – Interferon-α – Interleukin-2 Therapy with interferon-α has been associated with the development of antithyroid microsomal antibodies in 20% of patients and some have transient hypothyroidism, hyperthyroidism, or both.

Patients who have antithyroid antibodies before treatment are at higher risk for thyroid dysfunction during treatment.

Interleukin-2 has been associated with transient painless thyroiditis in 20% of patients.

Interferon-β and -γ have not been reported to cause thyroid dysfunction.

Growth Hormones – Somatrem – Somatropin Excessive use of thyroid hormones with growth hormones may accelerate epiphyseal closure.

However, untreated hypothyroidism may interfere with growth response to growth hormone.

Ketamine Concurrent use may produce marked hypertension and tachycardia; cautious administration to patients receiving thyroid hormone therapy is recommended.

Methylxanthine Bronchodilators – (e.g., Theophylline) Decreased theophylline clearance may occur in hypothyroid patients; clearance returns to normal when the euthyroid state is achieved.

Radiographic Agents Thyroid hormones may reduce the uptake of 123 I, 131 I, and 99m Tc.

Sympathomimetics Concurrent use may increase the effects of sympathomimetics or thyroid hormone.

Thyroid hormones may increase the risk of coronary insufficiency when sympathomimetic agents are administered to patients with coronary artery disease.

Chloral Hydrate Diazepam Ethionamide Lovastatin Metoclopramide 6-Mercaptopurine Nitroprusside Para-aminosalicylate sodium Perphenazine Resorcinol (excessive topical use) Thiazide Diuretics These agents have been associated with thyroid hormone and/or TSH level alterations by various mechanisms.

Oral anticoagulants Levothyroxine increases the response to oral anticoagulant therapy.

Therefore, a decrease in the dose of anticoagulant may be warranted with correction of the hypothyroid state or when the SYNTHROID dose is increased.

Prothrombin time should be closely monitored to permit appropriate and timely dosage adjustments (see Table 2 ).

Digitalis glycosides The therapeutic effects of digitalis glycosides may be reduced by levothyroxine.

Serum digitalis glycoside levels may be decreased when a hypothyroid patient becomes euthyroid, necessitating an increase in the dose of digitalis glycosides (see Table 2 ).

OVERDOSAGE

Overdosage The signs and symptoms of overdosage are those of hyperthyroidism (see PRECAUTIONS and ADVERSE REACTIONS ).

In addition, confusion and disorientation may occur.

Cerebral embolism, shock, coma, and death have been reported.

Seizures have occurred in a child ingesting 18 mg of levothyroxine.

Symptoms may not necessarily be evident or may not appear until several days after ingestion of levothyroxine sodium.

Treatment of Overdosage Levothyroxine sodium should be reduced in dose or temporarily discontinued if signs or symptoms of overdosage occur.

Acute Massive Overdosage This may be a life-threatening emergency, therefore, symptomatic and supportive therapy should be instituted immediately.

If not contraindicated (e.g., by seizures, coma, or loss of the gag reflex), the stomach should be emptied by emesis or gastric lavage to decrease gastrointestinal absorption.

Activated charcoal or cholestyramine may also be used to decrease absorption.

Central and peripheral increased sympathetic activity may be treated by administering β-receptor antagonists, e.g., propranolol, provided there are no medical contraindications to their use.

Provide respiratory support as needed; control congestive heart failure and arrhythmia; control fever, hypoglycemia, and fluid loss as necessary.

Large doses of antithyroid drugs (e.g., methimazole or propylthiouracil) followed in one to two hours by large doses of iodine may be given to inhibit synthesis and release of thyroid hormones.

Glucocorticoids may be given to inhibit the conversion of T 4 to T 3 .

Plasmapheresis, charcoal hemoperfusion and exchange transfusion have been reserved for cases in which continued clinical deterioration occurs despite conventional therapy.

Because T 4 is highly protein bound, very little drug will be removed by dialysis.

DESCRIPTION

SYNTHROID (levothyroxine sodium tablets, USP) contain synthetic crystalline L-3,3′,5,5′-tetraiodothyronine sodium salt [levothyroxine (T 4 ) sodium].

Synthetic T 4 is identical to that produced in the human thyroid gland.

Levothyroxine (T 4 ) sodium has an empirical formula of C 15 H 10 I 4 N NaO 4 • H 2 O, molecular weight of 798.86 g/mol (anhydrous), and structural formula as shown: Inactive Ingredients Acacia, confectioner’s sugar (contains corn starch), lactose monohydrate, magnesium stearate, povidone, and talc.

The following are the color additives by tablet strength: Strength (mcg) Color additive(s) 25 FD&C Yellow No.

6 Aluminum Lake* 50 None 75 FD&C Red No.

40 Aluminum Lake, FD&C Blue No.

2 Aluminum Lake 88 FD&C Blue No.

1 Aluminum Lake, FD&C Yellow No.

6 Aluminum Lake*, D&C Yellow No.

10 Aluminum Lake 100 D&C Yellow No.

10 Aluminum Lake, FD&C Yellow No.

6 Aluminum Lake* 112 D&C Red No.

27 & 30 Aluminum Lake 125 FD&C Yellow No.

6 Aluminum Lake*, FD&C Red No.

40 Aluminum Lake, FD&C Blue No.

1 Aluminum Lake 137 FD&C Blue No.

1 Aluminum Lake 150 FD&C Blue No.

2 Aluminum Lake 175 FD&C Blue No.

1 Aluminum Lake, D&C Red No.

27 & 30 Aluminum Lake 200 FD&C Red No.

40 Aluminum Lake 300 D&C Yellow No.

10 Aluminum Lake, FD&C Yellow No.

6 Aluminum Lake*, FD&C Blue No.

1 Aluminum Lake *Note – FD&C Yellow No.

6 is orange in color.

Meets USP Dissolution Test 3 Chemical structure for Levothyroxine.

HOW SUPPLIED

SYNTHROID ( levothyroxine sodium tablets, USP ) are round, color coded, scored and debossed with “SYNTHROID” on one side and potency on the other side.

They are supplied as follows: Strength (mcg) Color NDC# for bottles of 90 NDC # for bottles of 100 NDC # for bottles of 1000 NDC # for unit dose cartons of 100 25 orange 0074-4341-90 0074-4341-13 0074-4341-19 — 50 white 0074-4552-90 0074-4552-13 0074-4552-19 0074-4552-11 75 violet 0074-5182-90 0074-5182-13 0074-5182-19 0074-5182-11 88 olive 0074-6594-90 0074-6594-13 0074-6594-19 — 100 yellow 0074-6624-90 0074-6624-13 0074-6624-19 0074-6624-11 112 rose 0074-9296-90 0074-9296-13 0074-9296-19 — 125 brown 0074-7068-90 0074-7068-13 0074-7068-19 0074-7068-11 137 turquoise 0074-3727-90 0074-3727-13 0074-3727-19 — 150 blue 0074-7069-90 0074-7069-13 0074-7069-19 0074-7069-11 175 lilac 0074-7070-90 0074-7070-13 0074-7070-19 — 200 pink 0074-7148-90 0074-7148-13 0074-7148-19 0074-7148-11 300 green 0074-7149-90 0074-7149-13 0074-7149-19 — Storage Conditions Store at 25°C (77°F); excursions permitted to 15°-30°C (59°-86°F) [see USP Controlled Room Temperature].

SYNTHROID tablets should be protected from light and moisture.

Abbott Laboratories North Chicago, IL 60064, U.S.A.

03–A500 June 2011

GERIATRIC USE

Geriatric Use Because of the increased prevalence of cardiovascular disease among the elderly, levothyroxine therapy should not be initiated at the full replacement dose (see WARNINGS , PRECAUTIONS , and DOSAGE AND ADMINISTRATION ).

INDICATIONS AND USAGE

Levothyroxine sodium is used for the following indications: Hypothyroidism As replacement or supplemental therapy in congenital or acquired hypothyroidism of any etiology, except transient hypothyroidism during the recovery phase of subacute thyroiditis.

Specific indications include: primary (thyroidal), secondary (pituitary), and tertiary (hypothalamic) hypothyroidism and subclinical hypothyroidism.

Primary hypothyroidism may result from functional deficiency, primary atrophy, partial or total congenital absence of the thyroid gland, or from the effects of surgery, radiation, or drugs, with or without the presence of goiter.

Pituitary TSH Suppression In the treatment or prevention of various types of euthyroid goiters (see WARNINGS and PRECAUTIONS ), including thyroid nodules (see WARNINGS and PRECAUTIONS ), subacute or chronic lymphocytic thyroiditis (Hashimoto’s thyroiditis), multinodular goiter (see WARNINGS and PRECAUTIONS ) and, as an adjunct to surgery and radioiodine therapy in the management of thyrotropin-dependent well-differentiated thyroid cancer.

PEDIATRIC USE

Pediatric Use General The goal of treatment in pediatric patients with hypothyroidism is to achieve and maintain normal intellectual and physical growth and development.

The initial dose of levothyroxine varies with age and body weight (see DOSAGE AND ADMINISTRATION – Table 3 ).

Dosing adjustments are based on an assessment of the individual patient’s clinical and laboratory parameters (see PRECAUTIONS – Laboratory Tests ).

In children in whom a diagnosis of permanent hypothyroidism has not been established, it is recommended that levothyroxine administration be discontinued for a 30-day trial period, but only after the child is at least 3 years of age.

Serum T 4 and TSH levels should then be obtained.

If the T 4 is low and the TSH high, the diagnosis of permanent hypothyroidism is established, and levothyroxine therapy should be reinstituted.

If the T 4 and TSH levels are normal, euthyroidism may be assumed and, therefore, the hypothyroidism can be considered to have been transient.

In this instance, however, the physician should carefully monitor the child and repeat the thyroid function tests if any signs or symptoms of hypothyroidism develop.

In this setting, the clinician should have a high index of suspicion of relapse.

If the results of the levothyroxine withdrawal test are inconclusive, careful follow-up and subsequent testing will be necessary.

Since some more severely affected children may become clinically hypothyroid when treatment is discontinued for 30 days, an alternate approach is to reduce the replacement dose of levothyroxine by half during the 30-day trial period.

If, after 30 days, the serum TSH is elevated above 20 mU/L, the diagnosis of permanent hypothyroidism is confirmed, and full replacement therapy should be resumed.

However, if the serum TSH has not risen to greater than 20 mU/L, levothyroxine treatment should be discontinued for another 30-day trial period followed by repeat serum T 4 and TSH testing.

The presence of concomitant medical conditions should be considered in certain clinical circumstances and, if present, appropriately treated (see PRECAUTIONS ).

Congenital Hypothyroidism (see PRECAUTIONS – Laboratory Tests and DOSAGE AND ADMINISTRATION ) Rapid restoration of normal serum T 4 concentrations is essential for preventing the adverse effects of congenital hypothyroidism on intellectual development as well as on overall physical growth and maturation.

Therefore, SYNTHROID therapy should be initiated immediately upon diagnosis and is generally continued for life.

During the first 2 weeks of SYNTHROID therapy, infants should be closely monitored for cardiac overload, arrhythmias, and aspiration from avid suckling.

The patient should be monitored closely to avoid undertreatment or overtreatment.

Undertreatment may have deleterious effects on intellectual development and linear growth.

Overtreatment has been associated with craniosynostosis in infants, and may adversely affect the tempo of brain maturation and accelerate the bone age with resultant premature closure of the epiphyses and compromised adult stature.

Acquired Hypothyroidism in Pediatric Patients The patient should be monitored closely to avoid undertreatment and overtreatment.

Undertreatment may result in poor school performance due to impaired concentration and slowed mentation and in reduced adult height.

Overtreatment may accelerate the bone age and result in premature epiphyseal closure and compromised adult stature.

Treated children may manifest a period of catch-up growth, which may be adequate in some cases to normalize adult height.

In children with severe or prolonged hypothyroidism, catch-up growth may not be adequate to normalize adult height.

PREGNANCY

Pregnancy Category A Studies in women taking levothyroxine sodium during pregnancy have not shown an increased risk of congenital abnormalities.

Therefore, the possibility of fetal harm appears remote.

SYNTHROID should not be discontinued during pregnancy and hypothyroidism diagnosed during pregnancy should be promptly treated.

Hypothyroidism during pregnancy is associated with a higher rate of complications, including spontaneous abortion, pre-eclampsia, stillbirth and premature delivery.

Maternal hypothyroidism may have an adverse effect on fetal and childhood growth and development.

During pregnancy, serum T 4 levels may decrease and serum TSH levels increase to values outside the normal range.

Since elevations in serum TSH may occur as early as 4 weeks gestation, pregnant women taking SYNTHROID should have their TSH measured during each trimester.

An elevated serum TSH level should be corrected by an increase in the dose of SYNTHROID.

Since postpartum TSH levels are similar to preconception values, the SYNTHROID dosage should return to the pre-pregnancy dose immediately after delivery.

A serum TSH level should be obtained 6-8 weeks postpartum.

Thyroid hormones cross the placental barrier to some extent as evidenced by levels in cord blood of athyreotic fetuses being approximately one-third maternal levels.

Transfer of thyroid hormone from the mother to the fetus, however, may not be adequate to prevent in utero hypothyroidism.

NUSRING MOTHERS

Nursing Mothers Although thyroid hormones are excreted only minimally in human milk, caution should be exercised when SYNTHROID is administered to a nursing woman.

However, adequate replacement doses of levothyroxine are generally needed to maintain normal lactation.

BOXED WARNING

Boxed Warning WARNING: Thyroid hormones, including SYNTHROID, either alone or with other therapeutic agents, should not be used for the treatment of obesity or for weight loss.

In euthyroid patients, doses within the range of daily hormonal requirements are ineffective for weight reduction.

Larger doses may produce serious or even life threatening manifestations of toxicity, particularly when given in association with sympathomimetic amines such as those used for their anorectic effects.

INFORMATION FOR PATIENTS

Information for Patients Patients should be informed of the following information to aid in the safe and effective use of SYNTHROID: Notify your physician if you are allergic to any foods or medicines, are pregnant or intend to become pregnant, are breast-feeding or are taking any other medications, including prescription and over-the-counter preparations.

Notify your physician of any other medical conditions you may have, particularly heart disease, diabetes, clotting disorders, and adrenal or pituitary gland problems.

Your dose of medications used to control these other conditions may need to be adjusted while you are taking SYNTHROID.

If you have diabetes, monitor your blood and/or urinary glucose levels as directed by your physician and immediately report any changes to your physician.

If you are taking anticoagulants (blood thinners), your clotting status should be checked frequently.

Use SYNTHROID only as prescribed by your physician.

Do not discontinue or change the amount you take or how often you take it, unless directed to do so by your physician.

The levothyroxine in SYNTHROID is intended to replace a hormone that is normally produced by your thyroid gland.

Generally, replacement therapy is to be taken for life, except in cases of transient hypothyroidism, which is usually associated with an inflammation of the thyroid gland (thyroiditis).

Take SYNTHROID as a single dose, preferably on an empty stomach, one-half to one hour before breakfast.

Levothyroxine absorption is increased on an empty stomach.

It may take several weeks before you notice an improvement in your symptoms.

Notify your physician if you experience any of the following symptoms: rapid or irregular heartbeat, chest pain, shortness of breath, leg cramps, headache, nervousness, irritability, sleeplessness, tremors, change in appetite, weight gain or loss, vomiting, diarrhea, excessive sweating, heat intolerance, fever, changes in menstrual periods, hives or skin rash, or any other unusual medical event.

Notify your physician if you become pregnant while taking SYNTHROID.

It is likely that your dose of SYNTHROID will need to be increased while you are pregnant.

Notify your physician or dentist that you are taking SYNTHROID prior to any surgery.

Partial hair loss may occur rarely during the first few months of SYNTHROID therapy, but this is usually temporary.

SYNTHROID should not be used as a primary or adjunctive therapy in a weight control program.

Keep SYNTHROID out of the reach of children.

Store SYNTHROID away from heat, moisture, and light.

Agents such as iron and calcium supplements and antacids can decrease the absorption of levothyroxine sodium tablets.

Therefore, levothyroxine sodium tablets should not be administered within 4 hours of these agents.

DOSAGE AND ADMINISTRATION

General Principles The goal of replacement therapy is to achieve and maintain a clinical and biochemical euthyroid state.

The goal of suppressive therapy is to inhibit growth and/or function of abnormal thyroid tissue.

The dose of SYNTHROID that is adequate to achieve these goals depends on a variety of factors including the patient’s age, body weight, cardiovascular status, concomitant medical conditions, including pregnancy, concomitant medications, and the specific nature of the condition being treated (see WARNINGS and PRECAUTIONS ).

Hence, the following recommendations serve only as dosing guidelines.

Dosing must be individualized and adjustments made based on periodic assessment of the patient’s clinical response and laboratory parameters (see PRECAUTIONS – Laboratory Tests ).

SYNTHROID is administered as a single daily dose, preferably one-half to one-hour before breakfast.

SYNTHROID should be taken at least 4 hours apart from drugs that are known to interfere with its absorption (see PRECAUTIONS – Drug Interactions ).

Due to the long half-life of levothyroxine, the peak therapeutic effect at a given dose of levothyroxine sodium may not be attained for 4-6 weeks.

Caution should be exercised when administering SYNTHROID to patients with underlying cardiovascular disease, to the elderly, and to those with concomitant adrenal insufficiency (see PRECAUTIONS ).

Specific Patient Populations Hypothyroidism in Adults and in Children in Whom Growth and Puberty are Complete (see WARNINGS and PRECAUTIONS – Laboratory Tests ) Therapy may begin at full replacement doses in otherwise healthy individuals less than 50 years old and in those older than 50 years who have been recently treated for hyperthyroidism or who have been hypothyroid for only a short time (such as a few months).

The average full replacement dose of levothyroxine sodium is approximately 1.7 mcg/kg/day (e.g., 100-125 mcg/day for a 70 kg adult).

Older patients may require less than 1 mcg/kg/day.

Levothyroxine sodium doses greater than 200 mcg/day are seldom required.

An inadequate response to daily doses ≥ 300 mcg/day is rare and may indicate poor compliance, malabsorption, and/or drug interactions.

For most patients older than 50 years or for patients under 50 years of age with underlying cardiac disease, an initial starting dose of 25-50 mcg/day of levothyroxine sodium is recommended, with gradual increments in dose at 6-8 week intervals, as needed.

The recommended starting dose of levothyroxine sodium in elderly patients with cardiac disease is 12.5-25 mcg/day , with gradual dose increments at 4-6 week intervals.

The levothyroxine sodium dose is generally adjusted in 12.5-25 mcg increments until the patient with primary hypothyroidism is clinically euthyroid and the serum TSH has normalized.

In patients with severe hypothyroidism, the recommended initial levothyroxine sodium dose is 12.5-25 mcg/day with increases of 25 mcg/day every 2-4 weeks, accompanied by clinical and laboratory assessment, until the TSH level is normalized.

In patients with secondary (pituitary) or tertiary (hypothalamic) hypothyroidism, the levothyroxine sodium dose should be titrated until the patient is clinically euthyroid and the serum free- T 4 level is restored to the upper half of the normal range.

Pediatric Dosage – Congenital or Acquired Hypothyroidism (see PRECAUTIONS – Laboratory Tests ) General Principles In general, levothyroxine therapy should be instituted at full replacement doses as soon as possible.

Delays in diagnosis and institution of therapy may have deleterious effects on the child’s intellectual and physical growth and development.

Undertreatment and overtreatment should be avoided (see PRECAUTIONS – Pediatric Use ).

SYNTHROID may be administered to infants and children who cannot swallow intact tablets by crushing the tablet and suspending the freshly crushed tablet in a small amount (5-10 mL or 1-2 teaspoons) of water.

This suspension can be administered by spoon or by dropper.

DO NOT STORE THE SUSPENSION .

Foods that decrease absorption of levothyroxine, such as soybean infant formula, should not be used for administering levothyroxine sodium tablets (see PRECAUTIONS – Drug-Food Interactions ).

Newborns The recommended starting dose of levothyroxine sodium in newborn infants is 10-15 mcg/kg/day .

A lower starting dose (e.g., 25 mcg/day) should be considered in infants at risk for cardiac failure, and the dose should be increased in 4-6 weeks as needed based on clinical and laboratory response to treatment.

In infants with very low (< 5 mcg/dL) or undetectable serum T 4 concentrations, the recommended initial starting dose is 50 mcg/day of levothyroxine sodium.

Infants and Children Levothyroxine therapy is usually initiated at full replacement doses, with the recommended dose per body weight decreasing with age (see Table 3 ).

However, in children with chronic or severe hypothyroidism, an initial dose of 25 mcg/day of levothyroxine sodium is recommended with increments of 25 mcg every 2-4 weeks until the desired effect is achieved.

Hyperactivity in an older child can be minimized if the starting dose is one-fourth of the recommended full replacement dose, and the dose is then increased on a weekly basis by an amount equal to one-fourth the full-recommended replacement dose until the full recommended replacement dose is reached.

Table 3.

Levothyroxine Sodium Dosing Guidelines for Pediatric Hypothyroidism AGE Daily Dose Per Kg Body Weight a 0-3 months 10-15 mcg/kg/day 3-6 months 8-10 mcg/kg/day 6-12 months 6-8 mcg/kg/day 1-5 years 5-6 mcg/kg/day 6-12 years 4-5 mcg/kg/day > 12 years but growth and puberty incomplete 2-3 mcg/kg/day Growth and puberty complete 1.7 mcg/kg/day a The dose should be adjusted based on clinical response and laboratory parameters (see PRECAUTIONS – Laboratory Tests and Pediatric Use ).

Pregnancy Pregnancy may increase levothyroxine requirements (see PREGNANCY ).

Subclinical Hypothyroidism If this condition is treated, a lower levothyroxine sodium dose (e.g., 1 mcg/kg/day ) than that used for full replacement may be adequate to normalize the serum TSH level.

Patients who are not treated should be monitored yearly for changes in clinical status and thyroid laboratory parameters.

TSH Suppression in Well-differentiated Thyroid Cancer and Thyroid Nodules The target level for TSH suppression in these conditions has not been established with controlled studies.

In addition, the efficacy of TSH suppression for benign nodular disease is controversial.

Therefore, the dose of SYNTHROID used for TSH suppression should be individualized based on the specific disease and the patient being treated.

In the treatment of well-differentiated (papillary and follicular) thyroid cancer, levothyroxine is used as an adjunct to surgery and radioiodine therapy.

Generally, TSH is suppressed to < 0.1 mU/L, and this usually requires a levothyroxine sodium dose of greater than 2 mcg/kg/day .

However, in patients with high-risk tumors, the target level for TSH suppression may be < 0.01 mU/L.

In the treatment of benign nodules and nontoxic multinodular goiter, TSH is generally suppressed to a higher target (e.g., 0.1 to either 0.5 or 1.0 mU/L) than that used for the treatment of thyroid cancer.

Levothyroxine sodium is contraindicated if the serum TSH is already suppressed due to the risk of precipitating overt thyrotoxicosis (see CONTRAINDICATIONS , WARNINGS and PRECAUTIONS ).

Myxedema Coma Myxedema coma is a life-threatening emergency characterized by poor circulation and hypometabolism, and may result in unpredictable absorption of levothyroxine sodium from the gastrointestinal tract.

Therefore, oral thyroid hormone drug products are not recommended to treat this condition.

Thyroid hormone products formulated for intravenous administration should be administered.