throbber
(8-12-03)
`
`TABLETS
`CRESTOR®
`(rosuvastatin calcium)
`
`PCC 630100
`
`DESCRIPTION
`CRESTOR® (rosuvastatin calcium) is a synthetic lipid-lowering agent. Rosuvastatin is an
`inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase. This enzyme
`catalyzes the conversion of HMG-CoA to mevalonate, an early and rate-limiting step in
`cholesterol biosynthesis.
`
`bis[(E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-
`is
`calcium
`Rosuvastatin
`[methyl(methylsulfonyl)amino]
`pyrimidin-5-yl](3R,5S)-3,5-dihydroxyhept-6-enoic
`acid]
`calcium salt. The empirical formula for rosuvastatin calcium is (C22H27FN3O6S)2Ca. Its
`molecular weight is 1001.14. Its structural formula is:
`
`OH OH
`
`O
`
`O
`
`Ca2+
`
`2
`
`F
`
`N
`
`N S
`
`N
`O2Me
`
`Rosuvastatin calcium is a white amorphous powder that is sparingly soluble in water and
`methanol, and slightly soluble in ethanol. Rosuvastatin is a hydrophilic compound with a
`partition coefficient (octanol/water) of 0.13 at pH of 7.0.
`
`CRESTOR Tablets for oral administration contain 5, 10, 20, or 40 mg of rosuvastatin and the
`following inactive ingredients: microcrystalline cellulose NF, lactose monohydrate NF,
`tribasic calcium phosphate NF, crospovidone NF, magnesium stearate NF, hypromellose NF,
`triacetin NF, titanium dioxide USP, yellow ferric oxide, and red ferric oxide NF.
`
`CLINICAL PHARMACOLOGY
`General: In the bloodstream , cholesterol and triglycerides (TG) circulate as part of
`lipoprotein complexes.
` With ultracentrifugation,
`these complexes separate
`into
`very-low-density lipoprotein (VLDL), intermediate-density lipoprotein (IDL), and low-density
`lipoprotein (LDL) fractions that contain apolipoprotein B-100 (ApoB-100) and high-density
`lipoprotein (HDL) fractions.
`
`CFAD Exhibit 1046
`
`

`
`Cholesterol and TG synthesized in the liver are incorporated into VLDL and secreted into the
`circulation for delivery to peripheral tissues. TG are removed by the action of lipases, and in a
`series of steps, the modified VLDL is transformed first into IDL and then into cholesterol-rich
`LDL. IDL and LDL are removed from the circulation mainly by high affinity ApoB/E
`receptors, which are expressed to the greatest extent on liver cells. HDL is hypothesized to
`participate in the reverse transport of cholesterol from tissues back to the liver.
`
`Epidemiologic, experimental, and clinical studies have established that high LDL cholesterol
`(LDL-C), low HDL cholesterol (HDL-C), and high plasma TG promote human atherosclerosis
`and are risk factors for developing cardiovascular disease. In contrast, higher levels of HDL-
`C are associated with decreased cardiovascular risk.
`
`Like LDL, cholesterol-enriched triglyceride-rich lipoproteins, including VLDL, IDL, and
`remnants, can also promote atherosclerosis. Elevated plasma triglycerides are frequently
`found with low HDL-C levels and small LDL particles, as well as in association with
`non-lipid metabolic risk factors for coronary heart disease (CHD). As such, total plasma TG
`has not consistently been shown to be an independent risk factor for CHD. Furthermore, the
`independent effect of raising HDL or lowering TG on the risk of coronary and cardiovascular
`morbidity and mortality has not been determined.
`
`Mechanism of Action: Rosuvastatin is a selective and competitive inhibitor of HMG-CoA
`reductase, the rate-limiting enzyme that converts 3-hydroxy-3-methylglutaryl coenzyme A to
`mevalonate, a precursor of cholesterol. In vivo studies in animals, and in vitro studies in
`cultured animal and human cells have shown rosuvastatin to have a high uptake into, and
`selectivity for, action in the liver, the target organ for cholesterol lowering. In in vivo and
`in vitro studies, rosuvastatin produces its lipid-modifying effects in two ways. First, it
`increases the number of hepatic LDL receptors on the cell-surface to enhance uptake and
`catabolism of LDL. Second, rosuvastatin inhibits hepatic synthesis of VLDL, which reduces
`the total number of VLDL and LDL particles.
`
`Rosuvastatin reduces total cholesterol (total-C), LDL-C, ApoB, and nonHDL-C (total
`cholesterol minus HDL-C) in patients with homozygous and heterozygous familial
`hypercholesterolemia
`(FH), nonfamilial
`forms of hypercholesterolemia, and mixed
`dyslipidemia. Rosuvastatin also reduces TG and produces increases in HDL-C. Rosuvastatin
`reduces total-C, LDL-C, VLDL-cholesterol (VLDL-C), ApoB, nonHDL-C and TG, and
`increases HDL-C in patients with isolated hypertriglyceridemia. The effect of rosuvastatin on
`cardiovascular morbidity and mortality has not been determined.
`
`2
`
`

`
`Pharmacokinetics and Drug Metabolism
`Absorption: In clinical pharmacology studies in man, peak plasma concentrations of
`rosuvastatin were reached 3 to 5 hours following oral dosing. Both peak concentration (Cmax)
`and area under the plasma concentration-time curve (AUC) increased in approximate
`proportion to rosuvastatin dose. The absolute bioavailability of rosuvastatin is approximately
`20%.
`
`Administration of rosuvastatin with food decreased the rate of drug absorption by 20% as
`assessed by Cmax, but there was no effect on the extent of absorption as assessed by AUC.
`
`Plasma concentrations of rosuvastatin do not differ following evening or morning drug
`administration.
`
`Significant LDL-C reductions are seen when rosuvastatin is given with or without food, and
`regardless of the time of day of drug administration.
`
`Distribution: Mean volume of distribution at steady-state of rosuvastatin is approximately
`134 liters. Rosuvastatin is 88% bound to plasma proteins, mostly albumin. This binding is
`reversible and independent of plasma concentrations.
`
`Metabolism: Rosuvastatin is not extensively metabolized; approximately 10% of a
`radiolabeled dose is recovered as metabolite. The major metabolite is N-desmethyl
`rosuvastatin, which is formed principally by cytochrome P450 2C9, and in vitro studies have
`demonstrated that N-desmethyl rosuvastatin has approximately one-sixth to one-half the
`HMG-CoA reductase inhibitory activity of rosuvastatin. Overall, greater than 90% of active
`plasma HMG-CoA reductase inhibitory activity is accounted for by rosuvastatin.
`
`Excretion: Following oral administration, rosuvastatin and its metabolites are primarily
`excreted in the feces (90%). The elimination half-life (t1/2) of rosuvastatin is approximately
`19 hours.
`
`After an intravenous dose, approximately 28% of total body clearance was via the renal route,
`and 72% by the hepatic route.
`
`Special Populations
`Race: A population pharmacokinetic analysis revealed no clinically relevant differences in
`pharmacokinetics among Caucasian, Hispanic, and Black or Afro-Caribbean groups.
`However, pharmacokinetic studies show an approximate 2-fold elevation in median exposure
`(AUC) in Japanese subjects residing in Japan and in Chinese subjects residing in Singapore
`when compared with Caucasians residing in North America and Europe. No studies directly
`examining Asian ethnic populations residing in the U.S. are available, so the contribution of
`environmental and genetic factors to the observed increases in rosuvastatin drug levels have
`not been determined. (See WARNINGS, Myopathy/Rhabdomyolysis, and PRECAUTIONS,
`General.)
`
`3
`
`

`
`Gender: There were no differences in plasma concentrations of rosuvastatin between men
`and women.
`
`Geriatric: There were no differences in plasma concentrations of rosuvastatin between the
`nonelderly and elderly populations (age ≥65 years).
`
`Pediatric: In a pharmacokinetic study, 18 patients (9 boys and 9 girls) 10 to 17 years of age
`with heterozygous FH received single and multiple oral doses of rosuvastatin. Both Cmax and
`AUC of rosuvastatin were similar to values observed in adult subjects administered the same
`doses.
`
`Insufficiency: Mild
`Renal
`(creatinine clearance
`impairment
`renal
`to moderate
`≥ 30 mL/min/1.73m2) had no influence on plasma concentrations of rosuvastatin when oral
`doses of 20 mg rosuvastatin were administered for 14 days. However, plasma concentrations
`of rosuvastatin increased to a clinically significant extent (about 3-fold) in patients with severe
`(CLcr < 30 mL/min/1.73m2)
`renal
`impairment
`compared with
`healthy
`subjects
`(CLcr > 80 mL/min/1.73m2) (see PRECAUTIONS, General).
`
`Hemodialysis: Steady-state plasma concentrations of rosuvastatin in patients on chronic
`hemodialysis were approximately 50% greater compared with healthy volunteer subjects with
`normal renal function.
`
`Hepatic Insufficiency: In patients with chronic alcohol liver disease, plasma concentrations
`of rosuvastatin were modestly increased. In patients with Child-Pugh A disease, Cmax and
`AUC were increased by 60% and 5%, respectively, as compared with patients with normal
`liver function. In patients with Child-Pugh B disease, Cmax and AUC were increased 100% and
`21%,
`respectively,
`compared with patients with normal
`liver
`function
`(see
`CONTRAINDICATIONS and WARNINGS, Liver Enzymes).
`
`Drug-Drug Interactions
`Cytochrome P450 3A4: In vitro and in vivo data indicate that rosuvastatin clearance is not
`dependent on metabolism by cytochrome P450 3A4 to a clinically significant extent. This has
`been confirmed in studies with known cytochrome P450 3A4 inhibitors (ketoconazole,
`erythromycin, itraconazole).
`
`Ketoconazole: Coadministration of ketoconazole (200 mg twice daily for 7 days) with
`rosuvastatin (80 mg) resulted in no change in plasma concentrations of rosuvastatin.
`
`Erythromycin: Coadministration of erythromycin (500 mg four times daily for 7 days) with
`rosuvastatin (80 mg) decreased AUC and Cmax of rosuvastatin by 20% and 31%, respectively.
`These reductions are not considered clinically significant.
`
`4
`
`

`
`Itraconazole: Itraconazole (200 mg once daily for 5 days) resulted in a 39% and 28%
`increase in AUC of rosuvastatin after 10 mg and 80 mg dosing, respectively. These increases
`are not considered clinically significant.
`
`Fluconazole: Coadministration of fluconazole (200 mg once daily for 11 days) with
`rosuvastatin (80 mg) resulted in a 14% increase in AUC of rosuvastatin. This increase is not
`considered clinically significant.
`
`Cyclosporine: Coadministration of cyclosporine with rosuvastatin resulted in no significant
`changes in cyclosporine plasma concentrations. However, Cmax and AUC of rosuvastatin
`increased 11- and 7-fold, respectively, compared with historical data in healthy subjects.
`These
`increases are considered
`to be clinically significant (see PRECAUTIONS,
`Drug Interactions, WARNINGS, Myopathy/Rhabdomyolysis,
`and DOSAGE AND
`ADMINISTRATION).
`
`Warfarin: Coadministration of warfarin (20 mg) with rosuvastatin (40 mg) did not change
`warfarin plasma concentrations but increased the International Normalized Ratio (INR) (see
`PRECAUTIONS, Drug Interactions).
`
`Digoxin: Coadministration of digoxin (0.5 mg) with rosuvastatin (40 mg) resulted in no
`change to digoxin plasma concentrations.
`
`Fenofibrate: Coadministration of fenofibrate (67 mg three times daily) with rosuvastatin
`(10 mg) resulted in no significant changes in plasma concentrations of rosuvastatin or
`fenofibrate
`(see
`PRECAUTIONS,
`Drug
`Interactions,
`and WARNINGS,
`Myopathy/Rhabdomyolysis).
`
`Gemfibrozil: Coadministration of gemfibrozil (600 mg twice daily for 7 days) with
`rosuvastatin (80 mg) resulted in a 90% and 120% increase for AUC and Cmax of rosuvastatin,
`respectively. This increase is considered to be clinically significant (see PRECAUTIONS,
`Drug
`Interactions, WARNINGS, Myopathy/Rhabdomyolysis, DOSAGE AND
`ADMINISTRATION).
`
`Antacid: Coadministration of an antacid (aluminum and magnesium hydroxide combination)
`with rosuvastatin (40 mg) resulted in a decrease in plasma concentrations of rosuvastatin by
`54%. However, when the antacid was given 2 hours after rosuvastatin, there were no
`clinically significant changes in plasma concentrations of rosuvastatin (see PRECAUTIONS,
`Information for Patients).
`
`Oral contraceptives: Coadministration of oral contraceptives (ethinyl estradiol and
`norgestrel) with rosuvastatin resulted in an increase in plasma concentrations of ethinyl
`estradiol and norgestrel by 26% and 34%, respectively.
`
`5
`
`

`
`Clinical Studies
`Hypercholesterolemia (Heterozygous Familial and Nonfamilial)
`and Mixed Dyslipidemia (Fredrickson Type IIa and IIb)
`CRESTOR reduces total-C, LDL-C, ApoB, nonHDL-C, and TG, and increases HDL-C, in
`patients with hypercholesterolemia and mixed dyslipidemia. Therapeutic response is seen
`within 1 week, and maximum response is usually achieved within 4 weeks and maintained
`during long-term therapy.
`
`in a wide variety of adult patient populations with
`is effective
`CRESTOR
`hypercholesterolemia, with and without hypertriglyceridemia, regardless of race, gender, or
`age and in special populations such as diabetics or patients with heterozygous FH. Experience
`in pediatric patients has been limited to patients with homozygous FH.
`
`Dose-Ranging Study: In a multicenter, double-blind, placebo-controlled, dose-ranging study
`in patients with hypercholesterolemia, CRESTOR given as a single daily dose for 6 weeks
`significantly reduced total-C, LDL-C, nonHDL-C, and ApoB, across the dose range (Table 1).
`
`Table 1. Dose-Response in Patients With Primary Hypercholesterolemia
`(Adjusted Mean % Change From Baseline at Week 6)
`
`Dose
`Placebo
`5
`10
`20
`40
`
`N
`13
`17
`17
`17
`18
`
`Total-C
`-5
`-33
`-36
`-40
`-46
`
`LDL-C
`-7
`-45
`-52
`-55
`-63
`
`NonHDL-C
`-7
`-44
`-48
`-51
`-60
`
`ApoB
`-3
`-38
`-42
`-46
`-54
`
`TG
`-3
`-35
`-10
`-23
`-28
`
`HDL-C
`3
`13
`14
`8
`10
`
`Active-Controlled Study: CRESTOR was compared with the HMG-CoA reductase inhibitors
`atorvastatin, simvastatin, and pravastatin in a multicenter, open-label, dose-ranging study of
`2,240 patients with Type IIa and IIb hypercholesterolemia. After randomization, patients
`were treated for 6 weeks with a single daily dose of either CRESTOR, atorvastatin,
`simvastatin, or pravastatin (Figure 1 and Table 2).
`
`6
`
`

`
`Figure 1. Percent LDL-C Change by Dose of CRESTOR,
` Atorvastatin, Simvastatin, and Pravastatin
` at Week 6 in Patients With Type IIa/IIb Dyslipidemia
`
`Table 2. Percent Change in LDL-C From Baseline to Week 6 (LS means §) by
`Treatment Group (sample sizes ranging from 156-167 patients per group)
`
`Treatment
`
`CRESTOR
`
`Atorvastatin
`
`Pravastatin
`
`Simvastatin
`
`10 mg
`
`-46*
`
`-37
`
`-20
`
`-28
`
`Treatment Daily Dose
`20 mg
`40 mg
`
`80 mg
`
`-52†
`
`-43
`
`-24
`
`-35
`
`-55‡
`
`-48
`
`-30
`
`-39
`
`---
`
`-51
`
`---
`
`-46
`
`* CRESTOR 10 mg reduced LDL-C significantly more than atorvastatin 10 mg; pravastatin 10 mg, 20 mg, and
`40 mg; simvastatin 10 mg, 20 mg, and 40 mg. (p<0.002)
`† CRESTOR 20 mg reduced LDL-C significantly more than atorvastatin 20 mg and 40 mg; pravastatin 20 mg,
`and 40 mg; simvastatin 20 mg, 40 mg, and 80 mg. (p<0.002)
`‡ CRESTOR 40 mg reduced LDL-C significantly more than atorvastatin 40 mg; pravastatin 40 mg; simvastatin
`40 mg, and 80 mg (p<0.002)
`§ Corresponding standard errors are approximately 1.00
`
`7
`
`

`
`Heterozygous Familial Hypercholesterolemia
`In a study of patients with heterozygous FH (baseline mean LDL of 291), patients were
`randomized to CRESTOR 20 mg or atorvastatin 20 mg. The dose was increased by 6-week
`intervals. Significant LDL-C reductions from baseline were seen at each dose in both
`treatment groups (Table 3).
`
`Table 3. Mean LDL-C Percentage Change from Baseline
`
`CRESTOR
`(n=435)
`LS Mean* (95% CI)
`-47% (-49%, -46%)
`Week 6 20 mg
`-55% (-57%, -54%)
`Week 12 40 mg
`NA
`Week 18 80 mg
`* LS Means are least square means adjusted for baseline LDL.
`
`Atorvastatin
`(n=187)
`LS Mean (95% CI)
`-38% (-40%, -36%)
`-47% (-49%, -45%)
`-52% (-54%, -50%)
`
`Hypertriglyceridemia
`(Fredrickson Type IIb & IV)
`In a double-blind, placebo-controlled dose-response study in patients with baseline TG levels
`from 273 to 817 mg/dL, CRESTOR given as a single daily dose (5 to 40 mg) over 6 weeks
`significantly reduced serum TG levels (Table 4).
`
`Table 4. Dose-Response in Patients With Primary Hypertriglyceridemia Over 6 Weeks Dosing
`Median (Min, Max) Percent Change From Baseline
`
`CRESTOR
`20 mg
`N=27
`-37 (-72, 11)
`-43 (-74, -12)
`-49 (-83, 20)
`-34 (-61, -11)
`-31 (-66, 34)
`22 (-5, 50)
`
`CRESTOR
`40 mg
`N=25
`-43 (-80, -7)
`-51 (-62, -6)
`-56 (-83, 10)
`-40 (-51, -4)
`-43 (-61, -3)
`17 (-14, 63)
`
`CRESTOR
`CRESTOR
`Placebo
`5 mg
`10 mg
`N=26
`N=25
`N=23
`Triglycerides
`1 (-40, 72)
`-21 (-58, 38)
`-37 (-65, 5)
`NonHDL-C
`2 (-13, 19)
`-29 (-43, -8)
`-49 (-59, -20)
`VLDL-C
`2 (-36, 53)
`-25 (-62, 49)
`-48 (-72, 14)
`Total-C
`1 (-13, 17)
`-24 (-40, -4)
`-40 (-51, -14)
`LDL-C
`5 (-30, 52)
`-28 (-71, 2)
`-45 (-59, 7)
`HDL-C
`-3 (-25, 18)
`3 (-38, 33)
`8 (-8, 24)
`Homozygous Familial Hypercholesterolemia
`In an open-label, forced-titration study, homozygous FH patients (n=40, 8-63 years) were
`evaluated for their response to CRESTOR 20 to 40 mg titrated at a 6-week interval. In the
`overall population, the mean LDL-C reduction from baseline was 22%. About one-third of the
`patients benefited from increasing their dose from 20 mg to 40 mg with further LDL lowering
`of greater than 6%. In the 27 patients with at least a 15% reduction in LDL-C, the mean
`LDL-C reduction was 30% (median 28% reduction). Among 13 patients with an LDL-C
`reduction of <15%, 3 had no change or an increase in LDL-C. Reductions in LDL-C of 15%
`or greater were observed in 3 of 5 patients with known receptor negative status.
`
`Dose
`
`8
`
`

`
`INDICATIONS AND USAGE
`CRESTOR is indicated:
`
`1. as an adjunct to diet to reduce elevated total-C, LDL-C, ApoB, nonHDL-C, and TG levels
`and to increase HDL-C in patients with primary hypercholesterolemia (heterozygous
`familial and nonfamilial) and mixed dyslipidemia (Fredrickson Type IIa and IIb);
`
`2. as an adjunct to diet for the treatment of patients with elevated serum TG levels
`(Fredrickson Type IV);
`
`3.
`
`in patients with homozygous familial
`total-C, and ApoB
`to reduce LDL-C,
`hypercholesterolemia as an adjunct to other lipid-lowering treatments (e.g., LDL
`apheresis) or if such treatments are unavailable.
`
`According to NCEP-ATPIII guidelines, therapy with lipid-altering agents should be a
`component of multiple-risk-factor intervention in individuals at increased risk for coronary
`heart disease due to hypercholesterolemia. The two major modalities of LDL-lowering
`therapy are therapeutic lifestyle changes (TLC) and drug therapy. The TLC Diet stresses
`reductions in saturated fat and cholesterol intake. Table 5 defines LDL-C goals and cutpoints
`for initiation of TLC and for drug consideration.
`
`9
`
`

`
`Table 5. NCEP Treatment Guidelines: LDL-C Goals and Cutpoints for Therapeutic Lifestyle Changes
`and Drug Therapy in Different Risk Categories
`
`Risk Category
`
`LDL Goal
`
`CHDa or CHD Risk Equivalent
`(10 year risk > 20%)
`
`<100 mg/dL
`
`LDL level at which
`to initiate TLC
`≥100 mg/dL
`
`LDL level at which to consider
`drug therapy
`≥130 mg/dL
`(100-129 mg/dL:drug optional)b
`
`2+ Risk Factors
`(10-year risk ≤ 20%)
`
`<130 mg/dL
`
`≥130 mg/dL
`
`0-1 Risk Factorc
`
`<160 mg/dL
`
`≥160 mg/dL
`
`≥130 mg/dL
`10-year risk 10-20%
`≥160 mg/dL
`10-year risk <10%
`
`≥190 mg/dL (160-189 mg/dL
`(LDL-lowering drug optional)
`
`a CHD = coronary heart disease.
`b Some authorities recommend use of LDL-lowering drugs in this category if an LDL-C <100 mg/dL cannot be
`achieved by TLC. Others prefer use of drugs that primarily modify triglycerides and HDL-C, e.g., nicotinic
`acid or fibrate. Clinical judgment also may call for deferring drug therapy in this subcategory.
`c Almost all people with 0-1 risk factor have 10-year risk<10%; thus, 10-year risk assessment in people with
`0-1 risk factor is not necessary.
`
`After the LDL-C goal has been achieved, if the TG is still ≥ 200 mg/dL, nonHDL-C (total-C
`minus HDL-C) becomes a secondary target of therapy. NonHDL-C goals are set 30 mg/dL
`higher than LDL-C goals for each risk category.
`
`At the time of hospitalization for a coronary event, consideration can be given to initiating
`drug therapy at discharge if the LDL-C is ≥ 130 mg/dL (see NCEP Treatment Guidelines,
`above).
`
`Patients >20 years of age should be screened for elevated cholesterol levels every 5 years.
`
`Prior to initiating therapy with CRESTOR, secondary causes for hypercholesterolemia (e.g.,
`poorly-controlled
`diabetes
`mellitus,
`hypothyroidism,
`nephrotic
`syndrome,
`dyslipoproteinemias, obstructive liver disease, other drug therapy, and alcoholism) should be
`excluded, and a lipid profile performed to measure total-C, LDL-C, HDL-C, and TG. For
`patients with TG <400 mg/dL (<4.5 mmol/L), LDL-C can be estimated using the following
`equation: LDL-C = total-C - (0.20 x [TG] + HDL-C). For TG levels >400 mg/dL
`(>4.5 mmol/L), this equation is less accurate and LDL-C concentrations should be determined
`by ultracentrifugation.
`
`CRESTOR has not been studied in Fredrickson Type I, III, and V dyslipidemias.
`
`10
`
`

`
`CONTRAINDICATIONS
`CRESTOR is contraindicated in patients with a known hypersensitivity to any component of
`this product.
`
`Rosuvastatin is contraindicated in patients with active liver disease or with unexplained
`persistent elevations of serum transaminases (see WARNINGS, Liver Enzymes).
`
`Pregnancy and Lactation
`Atherosclerosis is a chronic process and discontinuation of lipid-lowering drugs during
`pregnancy should have little impact on the outcome of long-term therapy of primary
`hypercholesterolemia. Cholesterol and other products of cholesterol biosynthesis are essential
`components for fetal development (including synthesis of steroids and cell membranes). Since
`HMG-CoA reductase inhibitors decrease cholesterol synthesis and possibly the synthesis of
`other biologically active substances derived from cholesterol, they may cause fetal harm when
`administered
`to pregnant women. Therefore, HMG-CoA
`reductase
`inhibitors are
`contraindicated during pregnancy and in nursing mothers. ROSUVASTATIN SHOULD BE
`ADMINISTERED TO WOMEN OF CHILDBEARING AGE ONLY WHEN SUCH
`PATIENTS ARE HIGHLY UNLIKELY TO CONCEIVE AND HAVE BEEN INFORMED
`OF THE POTENTIAL HAZARDS. If the patient becomes pregnant while taking this drug,
`therapy should be discontinued immediately and the patient apprised of the potential hazard to
`the fetus.
`
`WARNINGS
`Liver Enzymes
`HMG-CoA reductase inhibitors, like some other lipid-lowering therapies, have been
`associated with biochemical abnormalities of liver function. The incidence of persistent
`elevations (>3 times the upper limit of normal [ULN] occurring on 2 or more consecutive
`occasions) in serum transaminases in fixed dose studies was 0.4, 0, 0, and 0.1% in patients
`who received rosuvastatin 5, 10, 20, and 40 mg, respectively. In most cases, the elevations
`were transient and resolved or improved on continued therapy or after a brief interruption in
`therapy. There were two cases of jaundice, for which a relationship to rosuvastatin therapy
`could not be determined, which resolved after discontinuation of therapy. There were no cases
`of liver failure or irreversible liver disease in these trials.
`
`It is recommended that liver function tests be performed before and at 12 weeks
`following both the initiation of therapy and any elevation of dose, and periodically (e.g.,
`semiannually) thereafter. Liver enzyme changes generally occur in the first 3 months of
`treatment with rosuvastatin. Patients who develop increased transaminase levels should be
`monitored until the abnormalities have resolved. Should an increase in ALT or AST of
`>3 times ULN persist, reduction of dose or withdrawal of rosuvastatin is recommended.
`
`Rosuvastatin should be used with caution in patients who consume substantial quantities of
`alcohol and/or have a history of liver disease (see CLINICAL PHARMACOLOGY, Special
`Populations, Hepatic Insufficiency).
` Active liver disease or unexplained persistent
`transaminase elevations are contraindications
`to
`the use of
`rosuvastatin
`(see
`CONTRAINDICATIONS).
`
`11
`
`

`
`Myopathy/Rhabdomyolysis
`Rare cases of rhabdomyolysis with acute renal failure secondary to myoglobinuria have
`been reported with rosuvastatin and with other drugs in this class.
`
`Uncomplicated myalgia has been reported in rosuvastatin-treated patients (see ADVERSE
`REACTIONS). Creatine kinase (CK) elevations (>10 times upper limit of normal) occurred in
`0.2% to 0.4% of patients taking rosuvastatin at doses up to 40 mg in clinical studies.
`Treatment-related myopathy, defined as muscle aches or muscle weakness in conjunction with
`increases in CK values >10 times upper limit of normal, was reported in up to 0.1% of patients
`taking rosuvastatin doses of up to 40 mg in clinical studies. Rare cases of rhabdomyolysis
`were seen with higher than recommended doses (80 mg) of rosuvastatin in clinical trials.
`Factors that may predispose patients to myopathy with HMG-CoA reductase inhibitors
`include advanced age (≥65 years), hypothyroidism, and renal insufficiency. The incidence of
`myopathy increased at doses of rosuvastatin above the recommended dosage range.
`
`Consequently:
`
`1. Rosuvastatin should be prescribed with caution in patients with predisposing factors for
`myopathy, such as, renal impairment (see DOSAGE AND ADMINISTRATION),
`advanced age, and hypothyroidism.
`
`2. Patients should be advised to promptly report unexplained muscle pain, tenderness, or
`weakness, particularly if accompanied by malaise or fever. Rosuvastatin therapy should be
`discontinued if markedly elevated CK levels occur or myopathy is diagnosed or suspected.
`
`3. The risk of myopathy during treatment with rosuvastatin may be increased with concurrent
`administration of other lipid-lowering therapies or cyclosporine, (see CLINICAL
`PHARMACOLOGY, Drug Interactions, PRECAUTIONS, Drug Interactions, and
`DOSAGE AND ADMINISTRATION). The benefit of further alterations in lipid
`levels by the combined use of rosuvastatin with fibrates or niacin should be carefully
`weighed against the potential risks of this combination. Combination therapy with
`rosuvastatin and gemfibrozil should generally be avoided. (See DOSAGE AND
`ADMINISTRATION and PRECAUTIONS, Drug Interactions).
`
`4. The risk of myopathy during treatment with rosuvastatin may be increased in
`circumstances which
`increase
`rosuvastatin drug
`levels
`(see CLINICAL
`PHARMACOLOGY, Special Populations, Race and Renal Insufficiency, and
`PRECAUTIONS, General).
`
`5. Rosuvastatin therapy should also be temporarily withheld in any patient with an
`acute, serious condition suggestive of myopathy or predisposing to the development
`of renal failure secondary to rhabdomyolysis (e.g., sepsis, hypotension, major
`
`12
`
`

`
`surgery, trauma, severe metabolic, endocrine, and electrolyte disorders, or
`uncontrolled seizures).
`
`PRECAUTIONS
`General
`Before instituting therapy with rosuvastatin, an attempt should be made to control
`hypercholesterolemia with appropriate diet and exercise, weight reduction in obese patients,
`and treatment of underlying medical problems (see INDICATIONS AND USAGE).
`
`impairment
`to patients with severe renal
`Administration of rosuvastatin 20 mg
`(CLcr <30 mL/min/1.73 m2) resulted in a 3-fold increase in plasma concentrations of
`rosuvastatin compared with healthy volunteers (see WARNINGS, Myopathy/Rhabdomyolysis
`and DOSAGE AND ADMINISTRATION).
`
`Pharmacokinetic studies show an approximate 2-fold elevation in median exposure in
`Japanese subjects residing in Japan and in Chinese subjects residing in Singapore compared
`with Caucasians residing in North America and Europe. The contribution of environmental
`and genetic factors to the difference observed has not been determined. However, these
`increases should be considered when making rosuvastatin dosing decisions for patients of
`Japanese and Chinese ancestry. (See WARNINGS Myopathy/Rhabdomyolysis; CLINICAL
`PHARMACOLOGY, Special Populations, Race.)
`
`Information for Patients
`Patients should be advised to report promptly unexplained muscle pain, tenderness, or
`weakness, particularly if accompanied by malaise or fever.
`
`When taking rosuvastatin with an aluminum and magnesium hydroxide combination antacid,
`the antacid should be taken at least 2 hours after rosuvastatin administration (see CLINICAL
`PHARMACOLOGY, Drug Interactions).
`
`Laboratory Tests
`In the rosuvastatin clinical trial program, dipstick-positive proteinuria and microscopic
`hematuria were observed among rosuvastatin treated patients, predominantly in patients dosed
`above the recommended dose range (i.e., 80 mg). However, this finding was more frequent in
`patients taking rosuvastatin 40 mg, when compared to lower doses of rosuvastatin or
`comparator statins, though it was generally transient and was not associated with worsening
`renal function. Although the clinical significance of this finding is unknown, a dose reduction
`should be considered for patients on rosuvastatin 40 mg therapy with unexplained persistent
`proteinuria during routine urinalysis testing.
`
`13
`
`

`
`Drug Interactions
`Cyclosporine: When rosuvastatin 10 mg was co-administered with cyclosporine in cardiac
`transplant patients, rosuvastatin mean Cmax and mean AUC were increased 11-fold and 7-fold,
`respectively, compared with healthy volunteers. These increases are considered to be
`clinically significant and require special consideration in the dosing of rosuvastatin to patients
`taking concomitant cyclosporine (see WARNINGS, Myopathy/Rhabdomyolysis, and
`DOSAGE AND ADMINISTRATION).
`
`Warfarin: Coadministration of rosuvastatin to patients on stable warfarin therapy resulted in
`clinically significant rises in INR (>4, baseline 2-3). In patients taking coumarin
`anticoagulants and rosuvastatin concomitantly, INR should be determined before starting
`rosuvastatin and frequently enough during early therapy to ensure that no significant alteration
`of INR occurs. Once a stable INR time has been documented, INR can be monitored at the
`intervals usually recommended for patients on coumarin anticoagulants. If the dose of
`rosuvastatin is changed, the same procedure should be repeated. Rosuvastatin therapy has not
`been associated with bleeding or with changes in INR in patients not taking anticoagulants.
`
`Gemfibrozil: Coadministration of a single rosuvastatin dose to healthy volunteers on
`gemfibrozil (600 mg twice daily) resulted in 2.2- and 1.9-fold, respectively, increase in mean
`Cmax and mean AUC of rosuvastatin (see DOSAGE AND ADMINISTRATION).
`
`Endocrine Function
`Although clinical studies have shown that rosuvastatin alone does not reduce basal plasma
`cortisol concentration or impair adrenal reserve, caution should be exercised if any HMG-CoA
`reductase inhibitor or other agent used to lower cholesterol levels is administered
`concomitantly with drugs that may decrease the levels or activity of endogenous steroid
`hormones such as ketoconazole, spironolactone, and cimetidine.
`
`CNS Toxicity
`CNS vascular lesions, characterized by perivascular hemorrhages, edema, and mononuclear
`cell infiltration of perivascular spaces, have been observed in dogs treated with several other
`members of this drug class. A chemically similar drug in this class produced dose-dependent
`optic nerve degeneration (Wallerian degeneration of retinogeniculate fibers) in dogs, at a dose
`that produced plasma drug levels about 30 times higher than the mean drug level in humans
`taking the highest recommended dose. Edema, hemorrhage, and partial necrosis in the
`interstitium of the choroid plexus was observed in a female dog sacrificed moribund at day 24
`at 90 mg/kg/day by oral gavage (systemic exposures 100 times the human exposure at
`40 mg/day based on AUC comparisons). Corneal opacity was seen in dogs treated for 52
`weeks at 6 mg/kg/day by oral gavage (systemic exposures 20 times the human exposure at
`40 mg/day based on AUC comparisons). Cataracts were seen in dogs treated for 12 weeks by
`oral gavage at 30 mg/kg/day (systemic exposures 60 times the human exposure at 40 mg/day
`based on AUC comparisons). Retinal dysplasia and retinal loss were seen in dogs treated for
`4 weeks by oral gavage at 90 mg/kg/day (systemic exposures 100 times the human exposure at
`40 mg/day based on AUC). Doses ≤30 mg/kg/day (systemic exposures ≤60 times the human
`exposure at 40 mg/day based on AUC comparisons) following treatment up to one year, did
`not reveal retinal findings.
`
`14
`
`

`
`Carcinogenesis, Mutagenesis, Impairment of Fertility
`In a 104-week carcinogenicity study in rats at dose levels of 2, 20, 60, or 80 mg/kg/day by oral
`gavage, the incidence of uterine stromal polyps was significantly increased in females at
`80 mg/kg/day at systemic exposure 20 times the human exposure at 40 mg/day based on
`AUC. Increased incidence of polyps was not seen at lower doses.
`
`In a 107-week carcinogenicity study in mice given 10, 60, 200 mg/kg/day by oral gavage, an
`increased incidence of hepatocellular adenoma/carcinoma was observed at 200 mg/kg/day at
`systemic exposures 20 times human exposure at 40 mg/day based on AUC. An increased
`incidence of hepatocellular tumors was

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket