WO2018009842A1 - Methods for rosuvastatin dosing by auc - Google Patents

Methods for rosuvastatin dosing by auc Download PDF

Info

Publication number
WO2018009842A1
WO2018009842A1 PCT/US2017/041162 US2017041162W WO2018009842A1 WO 2018009842 A1 WO2018009842 A1 WO 2018009842A1 US 2017041162 W US2017041162 W US 2017041162W WO 2018009842 A1 WO2018009842 A1 WO 2018009842A1
Authority
WO
WIPO (PCT)
Prior art keywords
rosuvastatin
auc
dose
dyslipidemia
drug
Prior art date
Application number
PCT/US2017/041162
Other languages
French (fr)
Inventor
Vuong Trieu
Original Assignee
Autotelic Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Autotelic Llc filed Critical Autotelic Llc
Publication of WO2018009842A1 publication Critical patent/WO2018009842A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to method for dosing rosuvastatin by AUC and related methods for treating dyslipidemia.
  • Dyslipidemia (hyperlipidemia) is a condition that is regarded as a risk factor that progress heart diseases and ultimately cause adverse cardiac symptoms. Individual therapeutic agents for individually treating dyslipidemia are well known.
  • HMG-CoA reductase inhibitors are potent therapeutic agents for the reduction of plasma cholesterol and therefore the reduction of risk for atherosclerosis/ cardiovascular events.
  • statins are associated with myalgia and other side effects which limited its usage.
  • Rosuvastatin an HMG-CoA reductase inhibitor, is useful for the treatment of hypercholesterolemia, hyperlipoproteinemia, and atherosclerosis; and rosuvastatin' s calcium salt is commercially available under the designation CrestorTM.
  • statins such as rosuvastatin
  • statins that suffer from race effect such that a lower dose of statin is required for Asian in order to achieve comparable efficacy as non-Asian.
  • the primary responsible factor being higher drug exposure - as AUC - in Asian versus non-Asian including Caucasian populations.
  • PK-guided rosuvastatin dosing e.g., area-under-the-curve, AUC.
  • target rosuvastatin exposure level e.g., target AUC level
  • the inventor performed clinical PK studies to define the target AUC based on the median AUC of Caucasians treated at optimal dose of 20 mg/day.
  • PK for the Asian population is not dose proportional and therefore PK-guided dosing will require determining the AUC; adjusting the rosuvastatin dose, confirming the rosuvastatin AUC, and re-dosing rosuvastatin. These steps are repeated (i.e., iterated) until the target rosuvastatin AUC is achieved.
  • the invention defines a target AUC for patients treated with rosuvastatin and provides a method for rosuvastatin dosing guided by AUC.
  • AUC for patients treated with rosuvastatin was determined to be 80 ng*hr/mL ⁇ 15% (i.e., from 68 to 92 ng*hr/mL).
  • the method comprises:
  • the invention provides a method for anti-dyslipidemia drug dosing by one or more pharmacokinetic parameters.
  • the method comprises:
  • the one or more pharmacokinetic parameters can be one or more of concentration time course, peak concentration (C ⁇ x), and time after administration to peak concentration, terminal half-life, area-under-the-curve (AUC), bioavailability, absorption, distribution, metabolism, excretion, biotransformation, and combinations thereof.
  • the pharmacokinetic parameter is area-under-the-curve (AUC).
  • the target optimal value (rosuvastatin AUC) is ⁇ 15% of the target optimal value. In other embodiments, the target optimal value (rosuvastatin AUC) is ⁇ 20% of the target optimal value. In further embodiments, the target optimal value (rosuvastatin AUC) is ⁇ 30% of the target optimal value.
  • the second dose is the same or substantially the same as the first dose; in other embodiments, the second dose is greater than the first dose; and in further embodiments, the second dose is less than the first dose.
  • the above methods further including repeating steps (a)-(d) until dyslipidemia control is achieved.
  • the subject is in need of treatment for dyslipidemia, and the method comprises administration of an anti-dyslipidemia drug.
  • FIGURE 1 compares rosuvastatin AUC from three BE studies conducted in Asian and non-Asian populations.
  • FIGURE 2 compares mean single dose AUC of rosuvastatin at different dose strengths in Asian and non- Asian healthy subjects (AUC, area under the concentration- time curve; PK, pharmacokinetics; RC, rosuvastatin).
  • AUC area under the concentration- time curve
  • PK pharmacokinetics
  • RC rosuvastatin
  • FIGURE 3 compares dose-normalized AUC of rosuvastatin at different dose strengths in Asian and non- Asian healthy subjects (AUC, area under the concentration- time curve; AUC/dose, dose-normalized area under the concentration-time curve; PK, pharmacokinetics; RC, rosuvastatin).
  • AUC area under the concentration- time curve
  • AUC/dose dose-normalized area under the concentration-time curve
  • PK pharmacokinetics
  • RC rosuvastatin
  • FIGURES 4A and 4B compare dose-normalized AUC of rosuvastatin (4A) and body weight (4B) among non- Asian, Asian and Korean (ST-101) subjects (AUC, area under the concentration-time curve; AUC/dose, dose-normalized area under the concentration-time curve; PK, pharmacokinetics; RC, rosuvastatin).
  • AUC area under the concentration-time curve
  • AUC/dose dose-normalized area under the concentration-time curve
  • PK pharmacokinetics
  • RC rosuvastatin
  • FIGURE 5 compares plots of AUC/dose of rosuvastatin against body weight with regression lines for Asian and non-Asian groups (AUC/dose, dose-normalized area under the concentration-time curve; solid dot, PK study in non- Asian subjects; circle, PK study in Asian subjects; triangle, ST101 PK study in Korean subjects).
  • FIGURE 6 illustrates mean rate of LDL-C reduction from clinical studies at different dose of rosuvastatin calcium (LDL-C, low-density lipoprotein cholesterol; LS, least squares; solid dot, PD study in non- Asian subjects; circle, PD study in Asian subjects; shaded circle, PD study in Korean subjects; triangle, ST101 PD study in Korean subjects).
  • LDL-C low-density lipoprotein cholesterol
  • LS least squares
  • solid dot PD study in non- Asian subjects
  • circle PD study in Asian subjects
  • shaded circle PD study in Korean subjects
  • triangle ST101 PD study in Korean subjects
  • FIGURE 7 compares plots of LDL-C reduction rate against dose with regression lines for Asian and non-Asian groups (LDL-C, low-density lipoprotein cholesterol; LS, least squares; solid dot, PD study in non- Asian subjects; circle, PD study in Asian subjects; shaded circle, PD study in Korean subjects; red triangle, ST101 PD study in Korean subjects).
  • LDL-C low-density lipoprotein cholesterol
  • LS least squares
  • solid dot PD study in non- Asian subjects
  • circle PD study in Asian subjects
  • shaded circle PD study in Korean subjects
  • red triangle ST101 PD study in Korean subjects
  • the invention provides methods for treating dyslipidemia by administering rosuvastatin.
  • a dosing regimen targeting specific rosuvastatin AUC is provided in which AUC determined from first dosing with rosuvastatin is used to adjust subsequent rosuvastatin dosing to achieve the targeted AUC.
  • the targeted AUC dosing regimen for rosuvastatin was made taking into account the targeted AUC value derived from rosuvastatin pharmacokinetic studies.
  • the pharmacokinetic parameter used in the method is area-under-the-curve (AUC).
  • the methods of the invention are effective in treating dyslipidemia.
  • the high PK variability and high drug exposure and dose non-proportionality of rosuvastatin require repeated drug monitoring following dosing to achieve the desired AUC (i.e., target AUC).
  • Anti-dyslipidemia drug pharmacokinetic variability was determined for rosuvastatin, a representative anti-dyslipidemia drug.
  • PK pharmacodynamics
  • rosuvastatin anti-dyslipidemia drug
  • pharmacokinetic parameters e.g., AUC.
  • the complexity of pharmacokinetics precludes any predictive methods for the accurate prediction of human rosuvastatin pharmacokinetics.
  • the only means of predicting the pharmacokinetic parameters is to actually perform the pharmacokinetic study, which is shown herein to be predictive of subsequent pharmacokinetic parameter determination.
  • the method of the invention provides a method that improves the delivery of anti-dyslipidemia drugs in general and rosuvastatin in particular.
  • rosuvastatin is administered to a subject to achieve a target AUC, which is defined as the median of conventional rosuvastatin PK (AUC) at conventional mg/day dosing (e.g., 80 ng*hr/mL ⁇ 15%). If dosing is below or above the target AUC, dose adjustment is adjusted up (increased) or down (decreased), respectively.
  • AUC the median of conventional rosuvastatin PK
  • the target AUC dosing can be varied to higher or lower target AUC when the patient is demonstrating resistance or sensitivity, respectively, to the anti-dyslipidemia drug. Regardless, once the targeted AUC for the patient has been defined, the target AUC needed to be maintained despite changes in physical condition (weight) and physiological condition (kidney, liver functional status etc.). Variables such as weight and organ function factors are known to affect rosuvastatin pharmacokinetics. However, with frequent monitoring, it is possible to keep dosing at constant AUC by appropriate adjustment of dosing when changes are noted.
  • the methods of the invention effectively administer anti-dyslipidemia drugs (e.g., rosuvastatin) by pharmacokinetic parameter (e.g., AUC) dosing to avoid the problems associated with pharmacokinetic variability described above.
  • Pharmacokinetic variability is avoided in the method by adjusting drug dose to achieve a targeted AUC.
  • the invention provides a method for rosuvastatin dosing by AUC.
  • the method comprises:
  • rosuvastatin at a first dose (e.g., under a first regimen, such as once a day) to a subject in need of therapy;
  • rosuvastatin concentration/time data points to provide area-under-the-curve (AUC) (i.e., the AUC resulting from the first dose); and (d) administering rosuvastatin at subsequent doses (e.g., a second dose and subsequent doses) to achieve a target rosuvastatin AUC (i.e., 80 ng*hr/mL ⁇ 15%).
  • AUC area-under-the-curve
  • the target rosuvastatin AUC is ⁇ 15% of the target. In other embodiments, the target rosuvastatin AUC is ⁇ 20% of the target. In further embodiments, the target rosuvastatin AUC is ⁇ 30% of the target.
  • the dose adjustment e.g., reduction
  • the dose adjustment cannot be accurately determined from the curve.
  • Area-under-the-curve is a pharmacokinetic parameter that is used in the method of the invention to dose olmesartan.
  • AUC Area-under-the-curve
  • AUC is the area under the curve in a plot of concentration of drug in blood plasma as a function of time. Typically, the area is calculated starting at the time the drug is administered and ending when the concentration in plasma is negligible. AUC represents the total drug exposure over time.
  • the phrase "transforming the concentration/time data points" refers to the application of mathematical operations, formulas, theories, and/or principles (i.e., a formula for calculating AUC) to the concentrations/time data points of the individual subject to provide AUC.
  • the target rosuvastatin AUC was determined from statistical analysis of a subject population receiving rosuvastatin.
  • the target AUC is the median AUC value determined from a population of subjects receiving rosuvastatin at a conventional dose (20 mg/day, daily administration) among non- Asian/Caucasian patients.
  • the nature of the device or method for determining the concentrations/time data points for calculating AUC is not critical. Some methods and devices for determining therapeutic drug (e.g., rosuvastatin) concentrations are known in the art and can be used.
  • a point-of-care device can be used to determine the concentrations and create the concentration/time data, transmit the data to a central location, and/or transmitting instructions to the patient to alter the administration.
  • the device and method for determining the concentrations/time data points for calculating AUC is an immunoassay assay device and method that utilizes one or more rosuvastatin antibodies (e.g., monoclonal antibodies) or functional fragments thereof.
  • the device is a lateral flow device.
  • the method of the invention is therapeutically effective for delivery of rosuvastatin and therefore is effective for treating dyslipidemia.
  • the above method is also effective for treating subjects in need of combined hypertension and dyslipidemia therapy.
  • the subject treatable by the method is a subject that is in need of treatment for hypertension and dyslipidemia.
  • olmesartan and an anti-dyslipidemia drug are individually administered.
  • a single dosage form that comprises olmesartan and an anti-dyslipidemia drug e.g., rosuvastatin or a salt thereof
  • the single dosage form comprises olmesartan and rosuvastatin calcium.
  • the invention provides a method for anti-dyslipidemia drug dosing by AUC.
  • the method comprises:
  • the target pharmacokinetic parameter is the pre-determined optimal value. In certain embodiments, the target pharmacokinetic parameter is the pre-determined optimal value ⁇ 15%. In other embodiments, the target pharmacokinetic parameter is the pre-determined optimal value ⁇ 20%. In further embodiments, the target pharmacokinetic parameter is the pre-determined optimal value ⁇ 30%.
  • the method further comprising repeating steps (a)-(d) until the target pharmacokinetic parameter value(s) is achieved.
  • PK pharmacokinetic parameter or parameters
  • concentration concentration time course, peak concentration, and time after administration to peak concentration, terminal half-life, area-under-the-curve (AUC), bioavailability, absorption, distribution, metabolism, excretion, biotransformation, or a combination thereof.
  • AUC area-under-the-curve
  • the phrase "transforming the concentration/time data points" refers to the application of mathematical operations, formulas, theories, and/or principles (e.g., a formula for calculating AUC) to the concentrations/time data points of the individual subject to provide the pharmacokinetic value (e.g., AUC).
  • the target pharmacokinetic value is pre-determined by statistical analysis from a population of subjects receiving the anti-dyslipidemia drug at its optimal dose.
  • the term "optimal dose” refers to a dose (e.g., mg/day) associated with desirable drug efficacy at lower risk doses of a drug (e.g., the Cmax range corresponding to patients experiencing high drug efficacy at a low dose) and is determined from a statistical analysis of a subject population receiving doses of the anti-dyslipidemia drug for whom there was therapeutic improvement without significant adverse drug reactions or significant side effects.
  • Significant adverse drug reactions refer to ADRs that the subject finds intolerable, impair physiologic functions, and put the subject at risk for immobility and/or death or combinations thereof.
  • Significant side effects refer to side effects that the subject finds intolerable, impair physiologic functions, and put the patient at risk for immobility and/or death or combinations thereof.
  • the nature of the device or method for determining the concentrations/time data points for calculating the pharmacokinetic parameter is not critical.
  • Methods and devices for determining therapeutic drug (e.g., anti-dyslipidemia drugs) concentrations are known in the art and can be used.
  • a point-of-care device can be used.
  • the device and method for determining the concentrations/time data points for calculating the pharmacokinetic parameter is an immunoassay assay device and method that utilizes one or more dyslipidemia antibodies (e.g., monoclonal antibodies) or functional fragments thereof.
  • the device is a lateral flow device.
  • the above method is also effective for treating subjects in need of combined hypertension and dyslipidemia therapy.
  • the subject treatable by the method is a subject that is in need of treatment for hypertension and dyslipidemia.
  • an antihypertensive drug and an anti-dyslipidemia drug are individually administered.
  • a single dosage form that comprises an antihypertensive drug and an anti-dyslipidemia drug e.g., rosuvastatin or a salt thereof
  • the single dosage form comprises olmesartan and rosuvastatin.
  • Rosuvastatin an HMG-CoA reductase inhibitor, is useful for the treatment of hypercholesterolemia, hyperlipoproteinemia, and atherosclerosis; and rosuvastatin's calcium salt is commercially available under the designation CrestorTM. Olmesartan medoxomil is useful for the treatment of essential hypertension and is commercially available under the designation BenicarTM.
  • a drug-drug interaction between rosuvastatin and olmesartan medoxomil occurs that results in delaying the in vivo release (i.e., dissolution) of rosuvastatin calcium to the gastrointestinal fluid and thus delaying the translocation thereof to the gastrointestinal membrane, inhibiting the absorption of rosuvastatin.
  • Olmesartan medoxomil formulations should be designed so as to exhibit high dissolution rate of olmesartan medoxomil in an in vitro comparative dissolution test, in order to obtain a bioequivalent formulation to other single formulations containing olmesartan medoxomil.
  • olmesartan medoxomil tablet comprises a preferred disintegrant, which may be one or more selected from the group consisting of low substituted hydroxypropyl cellulose, carboxymethylcellulose calcium, croscarmellose sodium, crospovidone, sodium starch glycolate, and pregelatinized starch.
  • olmesartan medoxomil tablet comprises 7.5 or more % by weight of low substituted hydroxypropyl cellulose, 5 or more % by weight of carboxymethylcellulose calcium, 15 or more % by weight of croscarmellose sodium, 10 or more % by weight of crospovidone, 5 or more % by weight of sodium starch glycolate, or 5 or more % by weight of pregelatinized starch, based on the total weight of the tablet comprising olmesartan medoxomil.
  • the compartment comprising olmesartan medoxomil comprises 7.5 to 65 % by weight of low substituted hydroxypropyl cellulose, 5 to 60 % by weight of carboxymethylcellulose calcium, 15 to 30 % by weight of croscarmellose sodium, 10 to 40 % by weight of crospovidone, 5 to 40 % by weight of sodium starch glycolate, or 5 to 60 % by weight of pregelatinized starch, based on the total weight of the tablet comprising olmesartan medoxomil.
  • the tablet comprising olmesartan medoxomil comprises 7.5 to 65 % by weight, preferably 10 to 60 % by weight, more preferably about 20 ⁇ 1 % by weight of low substituted hydroxypropyl cellulose, based on the total weight of the tablet comprising olmesartan medoxomil.
  • WO 2013/147462 An improved pharmaceutical composition that is a single dosage form of olmesartan medoxomil and rosuvastatin or its salts is described in WO 2013/147462.
  • This single dosage form comprises separate compartments for each drug in which each drug is separately and independently formulated.
  • the single dosage form is administered the interaction to in vivo absorption is minimized and the combination formulation is bioequivalent to the single formulation of each of drugs.
  • the subject treatable by the methods of the invention is a subject that is in need of treatment for hypertension and dyslipidemia.
  • an antihypertensive drug and an anti-dyslipidemia drug are individually administered.
  • a single dosage form that comprises an antihypertensive drug and an anti-dyslipidemia drug e.g., rosuvastatin or a salt thereof
  • the single dosage form comprises olmesartan and rosuvastatin.
  • the pharmaceutical composition useful in the methods of the invention which includes olmesartan medoxomil and rosuvastatin or its salt (e.g., rosuvastatin calcium), are formulated into a combination dosage form having separate compartments. That is, the pharmaceutical composition has a single dosage form comprising a compartment comprising olmesartan medoxomil; and a compartment comprising rosuvastatin or its salt, wherein the compartments are formulated in a separate form.
  • olmesartan medoxomil and rosuvastatin or its salt e.g., rosuvastatin calcium
  • the active ingredients i.e., olmesartan medoxomil and rosuvastatin or its salt
  • olmesartan medoxomil may be used in an amount of about 5 mg to about 80 mg, preferably about 10 mg to about 40 mg, in a unit formulation (i.e., unit dosage form).
  • rosuvastatin or its salt may be used in an amount of about 2 mg to about 40 mg, preferably about 5 mg to about 20 mg, in a unit formulation (i.e., unit dosage form).
  • the salt of rosuvastatin may be a conventional pharmaceutically acceptable salt, such as calcium salt, hydrochloride, hydrobromide, sulfate, phosphate, acetate, maleate, fumarate, lactate, tartrate, citrate, gluconate, besylate, and camsylate.
  • rosuvastatin calcium may be used in the present invention.
  • the pharmaceutical composition may be administered once a day, but not limited thereto.
  • the pharmaceutical composition has a combination dosage form having separate compartments (i.e., a double-layered tablet form), comprising or consisting essentially of a layer comprising rosuvastatin or its salt and a layer comprising olmesartan medoxomil.
  • the compartment comprising rosuvastatin or its salt includes a certain disintegrant (i.e., cellulose-type and/or povidone-type disintegrants), in a certain amount, rapid disintegration and high initial dissolution rate of rosuvastatin or its salt can be accomplished, thereby being able to obtain a combination formulation bioequivalent to the single formulation of rosuvastatin or its salt.
  • the disintegrant may be one or more selected from the group consisting of povidone (for example, KolidoneTM), crospovidone (for example, PolyplasdoneTM), low substituted hydroxypropyl cellulose, croscarmellose sodium, and carboxymethylcellulose calcium.
  • the disintegrant may be a mixture of crospovidone and croscarmellose sodium; or croscarmellose sodium.
  • the disintegrant may be present in an amount ranging from 2 to 20 % by weight, preferably from 3 to 15 % by weight, based on the total weight of the compartment comprising rosuvastatin or its salt.
  • the dissolution rate of rosuvastatin or its salt is decreased; and/or the amount used is increased, which may cause insufficient compression force during the compressing step, thereby leading to high friability of the resulting formulation (e.g., tablet).
  • the use of other disintegrants brings about insufficient hardness, which may cause unwanted problems in packaging or delivery.
  • a combination formulation comprising rosuvastatin and olmesartan medoxomil should be designed so as to exhibit high dissolution rate of olmesartan medoxomil in an in vitro comparative dissolution test, in order to obtain a bioequivalent formulation to the single formulation containing olmesartan medoxomil.
  • the compartment comprising olmesartan medoxomil comprises a preferred disintegrant, which may be one or more selected from the group consisting of low substituted hydroxypropyl cellulose, carboxymethylcellulose calcium, croscarmellose sodium, crospovidone, sodium starch glycolate, and pregelatinized starch.
  • the compartment comprising olmesartan medoxomil comprises 7.5 or more % by weight of low substituted hydroxypropyl cellulose, 5 or more % by weight of carboxymethylcellulose calcium, 15 or more % by weight of croscarmellose sodium, 10 or more % by weight of crospovidone, 5 or more % by weight of sodium starch glycolate, or 5 or more % by weight of pregelatinized starch, based on the total weight of the compartment comprising olmesartan medoxomil.
  • the compartment comprising olmesartan medoxomil comprises 7.5 to 65 % by weight of low substituted hydroxypropyl cellulose, 5 to 60 % by weight of carboxymethylcellulose calcium, 15 to 30 % by weight of croscarmellose sodium, 10 to 40 % by weight of crospovidone, 5 to 40 % by weight of sodium starch glycolate, or 5 to 60 % by weight of pregelatinized starch, based on the total weight of the compartment comprising olmesartan medoxomil.
  • the compartment comprising olmesartan medoxomil comprises 7.5 to 65 % by weight, preferably 10 to 60 % by weight, more preferably about 20 ⁇ 1 % by weight of low substituted hydroxypropyl cellulose, based on the total weight of the compartment comprising olmesartan medoxomil.
  • the pharmaceutical composition may further comprise one or more excipients conventionally used in the field of pharmaceutics, for example a diluent (or additive), a binder, a lubricant, in addition to said disintegrant.
  • excipients conventionally used in the field of pharmaceutics
  • a diluent or additive
  • a binder or a lubricant
  • the pharmaceutical composition may be also coated with an appropriate coating agent, such as a film-coating agent.
  • the diluent includes lactose (including its hydrate), dextrin, mannitol, sorbitol, starch, microcrystalline cellulose (for example, CelphereTM), silicified microcrystalline cellulose (for example, ProsolvTM), calcium hydrogen phosphate (including its hydrate), anhydrous calcium hydrogen phosphate, calcium carbonate, saccharides, and a mixture thereof.
  • the binder includes polyvinylpyrrolidone, copovidone, gelatin, starch, sucrose, methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl alkylcellulose (for example, hydroxypropyl methylcellulose), and a mixture thereof.
  • the lubricant includes stearic acid, stearates (for example, magnesium stearate), talc, corn starch, carnauba wax, light anhydrous silicic acid, magnesium silicate, synthetic aluminum silicate, hydrogenated oil, hydrogenated oil, titanium oxide, microcrystalline cellulose, macrogol 4000 or 6000, isopropyl myristate, calcium hydrogen phosphate, and a mixture thereof.
  • the coating agent for example a film-coating agent, includes a conventional polymer such as Opadry . The film-coating agent may be used in a minimum amount for providing an appropriate size of the formulation, but not limited thereto.
  • the pharmaceutical composition having a double-layered tablet form may be prepared by preparing granules containing rosuvastatin and granules containing olmesartan medoxomil, respectively; and then compressing the mixture thereof with a double-layer tablet-press machine. If necessary, the resulting double-layered tablet may be coated with a film-coating agent such as OpadryTM.
  • the granules containing rosuvastatin and the granules containing olmesartan medoxomil may be prepared according to dry granulation methods or wet granulation methods. For example, the granules containing rosuvastatin may be prepared according to a dry granulation method.
  • the granules containing rosuvastatin may be prepared by mixing rosuvastatin calcium, an additive (diluent), a disintegrant, and a lubricant according to a conventional method; and then granulating the mixture with e.g., a roller compactor (TF mini, Vector). And also, the granules containing olmesartan medoxomil may be prepared according to a wet granulation method.
  • the granules containing olmesartan medoxomil may be prepared by mixing olmesartan medoxomil, a binder, an additive (diluent), a disintegrant; granulating the mixture with a high speed mixer (MIC Developer-5, COMASA); and then drying and sieving the resulting granules.
  • Representative double-layer tablets can be prepared as described below.
  • Step 1 Preparation of granules containing rosuvastatin.
  • Rosuvastatin calcium, lactose monohydrate, ProsolvTM, dibasic calcium phosphate dihydrate, crospovidone, croscarmellose sodium, light anhydrous silicic acid, and magnesium stearate (85% of the total amount used in the rosuvastatin-layer) were sieved through a 24 mesh and then mixed. The resulting mixture was granulated using a roller compactor (TF mini, Vector). The obtained granules were sieved through a 24 mesh and then mixed with magnesium stearate pre-sieved though a 35 mesh (15% of the total amount used in the rosuvastatin-layer) to prepare a rosuvastatin-containing granule mixture.
  • TF mini roller compactor
  • Step 2 Preparation of granules containing olmesartan medoxomil.
  • Olmesartan medoxomil, hydroxypropyl cellulose, lactose monohydrate, microcrystalline cellulose, and low substituted hydroxypropyl cellulose were sieved through a 24 mesh and then mixed.
  • the resulting mixture was granulated using a high speed mixer (MIC Developer-5, COMASA).
  • the resulting dry granules were sieved through a 24 mesh and then mixed with magnesium stearate pre-sieved though a 35 mesh and yellow iron oxide pre-sieved through an 80 mesh to prepare a olmesartan medoxomil-containing granule mixture.
  • the rosuvastatin-containing granule mixture prepared in Step 1 and the olmesartan medoxomil-containing granule mixture prepared in Step 2 were compressed with a double-layer tablet-press machine (BB-11, RIVA) to obtain double-layered tablets.
  • the resulting tablets were film-coated with OpadryTM in a pan coating machine (LDCS, VECTOR).
  • PK rosuvastatin pharmacokinetics
  • PD pharmacodynamics
  • Hyperlipidemia Hypertriglyceridemia, Dysbetalipoproteinemia, and
  • Hyperlipidemia is a condition in which blood lipid levels increase due to abnormal lipid metabolism of triglycerides, cholesterol, and other fatty acids. Hyperlipidemia can be caused by genetic factors or by secondary factors such as hypothyroidism, hepatic disease, nephrotic syndrome, or diabetes mellitus. Serum lipids primarily comprise cholesterol, triglycerides, phospholipids, and free fatty acids transported in a lipoprotein form. Lipoproteins are categorized into 4 types as follows: chylomicrons, very low-density lipoprotein (VLDL), LDL, and HDL.
  • VLDL very low-density lipoprotein
  • VLDLs are synthesized by the liver in the fasting state and are responsible for transporting triglycerides to peripheral tissues where some are converted to LDLs.
  • LDLs are the major carriers of cholesterol to peripheral tissues; however, they are also a major risk factor for coronary arteriosclerosis.
  • Treatment for hyperlipidemia includes lifestyle changes such as dietary modification, weight management, and physical activity to decrease LDL levels.
  • Cholesterol-lowering agents e.g., statins, bile acid sequestrants, nicotinic acid, fibrates, and ezetimibe
  • Rosuvastatin calcium is a statin drug (HMG-CoA reductase inhibitor) that decreases LDL cholesterol by inhibiting HMG-CoA reductase, which is an enzyme in the metabolic pathway for cholesterol production (Olsson et al. 2002b).
  • the geometric mean ratios of test drug and the reference drug in connection of AUCi ast and Cma X for rosuvastatin were 91.31% and 90.37%), respectively.
  • the 90% confidence intervals of the geometric mean ratios of test drug and the reference drug were within the bioequivalence acceptance range (80.00-125.00%).
  • the second BE study was conducted in Indian population (Indian BE study). This was an open label, balanced, randomized, two-treatment, two-sequence, two-period, crossover, single-dose oral BE study of ST-101 (Test drug, fixed dose combination of RC/OM 10/20 mg) and co-administration of RC 10 mg and OM 20 mg (Reference drug) in healthy, adult, male, human subjects under fasting conditions. 26 subjects had completed the study.
  • the geometric mean ratios of test drug and the reference drug in connection of AUCi ast and C max for rosuvastatin were 99.80%) and 99.40%), respectively.
  • the 90% confidence intervals of the geometric mean ratios of test drug and the reference drug were within the bioequivalence acceptance range.
  • the third BE study was conducted in non-Asian population in Czech Republic (EU BE study). This was a single-dose, randomized, two-period, two-treatment, two-sequence, crossover BE study on ST-101 (test drug, fixed dose combination of OM/RC 40 mg/20 mg) versus two co-administered reference products OM 40 mg and RC 20 mg in healthy volunteers under fasting conditions. 56 subjects had completed the study.
  • the geometric mean ratios of test drug and the reference drug in connection of AUCi ast and C max for rosuvastatin were 100.16 % and 98.54 %, respectively.
  • the 90% confidence intervals of the geometric mean ratios of test drug and the reference drug were within the bioequivalence acceptance range.
  • FIGURE 1 A first figure.
  • the target AUC was defined as Caucasian median AUC at 20 mg/day. This was rounded up to 80 mg*hr/mL.
  • PubMed database was searched to identify relevant scientific and review articles from October 2002 to March 2015 using the search term rosuvastatin PK. Reference lists of included articles were reviewed to locate additional references of interest.
  • PK studies with single dose AUC of rosuvastatin calcium in healthy subjects were selected. Rosuvastatin AUC from co-administration of rosuvastatin calcium with other drugs were also included if there was no known DDI. More than 50% of the studies reported AUC in geometric mean. Thus, to make comparable analysis, only studies with AUC calculated by geometric mean were included in the analysis.
  • PK and demographic data from a total of 745 healthy subjects in 11 clinical studies were included in the final analysis: CSR Synopsis DW_DWJ 1276002; Cooper et al. 2002; Martin et al.
  • Dose proportional analysis was performed to confirm reported dose proportional nature of rosuvastatin PK.
  • the AUC/dose was then used in multivariate correlation to define the variable that has significant impact on PK.
  • the analysis yielded significant correlation between weight and race to AUC/dose, but not age.
  • ANCOVA analysis was then performed to show that race is a significant factor even after weight was taken in consideration.
  • the mean AUCi ast of rosuvastatin from each study were plotted versus dose (FIGURE 2).
  • the plot showed that the mean AUCs of rosuvastatin in Asian subjects are higher than the mean AUCs in non- Asian subjects at the same dose.
  • Rosuvastatin AUC from subjects treated with ST-101 overlapped with AUC from 20 mg of rosuvastatin in Asian subjects.
  • rosuvastatin PK validated the use of dose-normalized AUC for ANCOVA.
  • non -Asian subjects have a significantly lower dose-normalized AUC of rosuvastatin (p ⁇ 0.0001)
  • ANCOVA was conducted to determine whether there was a significant difference between Asian and non- Asian subjects on dose-normalized AUC after controlling for BW, using Fit Model Platform (JMP version 11.2.1 clinical software). The number of subjects in each study was used to assign a weight to each study in the analysis.
  • rosuvastatin AUC from Korean subjects treated with ST-101 is the same as rosuvastatin AUC from Korean and other Asian subjects treated with rosuvastatin (FIGURE 4A), which is well understood.
  • PubMed database was searched to identify relevant scientific and review articles from September 2001 to June 2015 using the search terms rosuvastatin efficacy, response, effect/clinical trial. Reference lists of included articles were reviewed to locate additional references of interest.
  • PD studies with rosuvastatin calcium dose range from 1 to 80 mg monotherapy were selected.
  • the mean reduction rate in LDL-C in patients from each study was plotted versus dose (FIGURE 6).
  • the LDL-C reduction rate by ST-101 is similar to the effects by 20 mg of rosuvastatin calcium in Korean, other Asian, and non-Asian patients.
  • Regression analysis showed a significant correlation between LDL-C reduction and dose (p ⁇ 0.0001) (FIGURE 6). This dose-related response for rosuvastatin calcium has been reported previously (CRESTOR ® Prescribing Information 2014).
  • the interaction between dose and race on the LDL-C reduction was evaluated.
  • the number of subjects in each study was used to assign a weight to each study in the analysis.
  • the slope for dose-response in Asian patients is much steeper than that in non-Asian patients, therefore, race is an important determinant of PD response for rosuvastatin.
  • Li Y, Jiang X, Lan K, Zhang R, Li X, Jiang Q Pharmacokinetic properties of rosuvastatin after single-dose, oral administration in Chinese volunteers: a randomized, open-label, three-way crossover study. Clin Ther. 2007 Oct; 29(10):2194-2203.
  • Lu TM Ding YA, Leu HB, Yin WH, Sheu WH, Chu KM. Effect of rosuvastatin on plasma levels of asymmetric dimethylarginine in patients with hypercholesterolemia. Am J Cardiol. 2004 Jul 15; 94(2): 157-161.
  • Martin PD Warwick MJ, Dane AL, Brindley C, Short T. Absolute oral bioavailability of rosuvastatin in healthy white adult male volunteers. Clin Ther. 2003a Oct; 25(10):2553-2563.
  • Mazza F Stefanutti C, Di Giacomo S, Vivenzio A, Fraone N, Mazzarella B, et al. Effects of low-dose atorvastatin and rosuvastatin on plasma lipid profiles: a long-term, randomized, open-label study in patients with primary hypercholesterolemia. Am J Cardiovasc Drugs. 2008; 8(4):265-270.
  • Pirro M Schillaci G, Romagno PF, Mannarino MR, Bagaglia F, Razzi R, et al. Influence of short-term rosuvastatin therapy on endothelial progenitor cells and endothelial function. J Cardiovasc Pharmacol Ther. 2009 Mar; 14(1): 14-21.

Abstract

Methods for anti-dyslipidemia drug dosing by pharmacokinetic parameter determination. In one embodiment, the anti-dyslipidemia drug is rosuvastatin and the pharmacokinetic parameter is AUC. The methods are effective for treating dyslipidemia.

Description

METHODS FOR ROSUVASTATIN DOSING BY AUC
CROSS-REFERENCE TO RELATED APPLICATION This application claims the benefit of US Application No. 62/360,262, filed
July 8, 2016, expressly incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The present invention relates to method for dosing rosuvastatin by AUC and related methods for treating dyslipidemia.
BACKGROUND OF THE INVENTION
Dyslipidemia (hyperlipidemia) is a condition that is regarded as a risk factor that progress heart diseases and ultimately cause adverse cardiac symptoms. Individual therapeutic agents for individually treating dyslipidemia are well known.
HMG-CoA reductase inhibitors (e.g., statins) are potent therapeutic agents for the reduction of plasma cholesterol and therefore the reduction of risk for atherosclerosis/ cardiovascular events. However, statins are associated with myalgia and other side effects which limited its usage. Rosuvastatin, an HMG-CoA reductase inhibitor, is useful for the treatment of hypercholesterolemia, hyperlipoproteinemia, and atherosclerosis; and rosuvastatin' s calcium salt is commercially available under the designation Crestor™.
There is high variability of the pharmacokinetics for statins when conventionally dosed at mg/day, which in turn results in highly variable outcomes. Additionally, there are statins, such as rosuvastatin, that suffer from race effect such that a lower dose of statin is required for Asian in order to achieve comparable efficacy as non-Asian. The primary responsible factor being higher drug exposure - as AUC - in Asian versus non-Asian including Caucasian populations.
Therefore, despite advances in the development of dyslipidemia treatments and related therapeutic drugs, their formulations, and methods for their administration, a need exists to further improve the clinical effectiveness of the administration of statins. The present invention seeks to fulfill this need and provides further related advantages.
SUMMARY OF THE INVENTION
Pharmacokinetic (PK) variability and differences can be mitigated with PK-guided rosuvastatin dosing (e.g., area-under-the-curve, AUC). Because lower rosuvastatin dose is recommended for Asian population, and that no target rosuvastatin exposure level (e.g., target AUC level) has been suggested by the art, the inventor performed clinical PK studies to define the target AUC based on the median AUC of Caucasians treated at optimal dose of 20 mg/day. Furthermore, the inventor demonstrated that surprisingly, PK for the Asian population is not dose proportional and therefore PK-guided dosing will require determining the AUC; adjusting the rosuvastatin dose, confirming the rosuvastatin AUC, and re-dosing rosuvastatin. These steps are repeated (i.e., iterated) until the target rosuvastatin AUC is achieved.
In one aspect, the invention defines a target AUC for patients treated with rosuvastatin and provides a method for rosuvastatin dosing guided by AUC.
In accordance with the invention and as described herein, a target rosuvastatin
AUC for patients treated with rosuvastatin was determined to be 80 ng*hr/mL ± 15% (i.e., from 68 to 92 ng*hr/mL).
In one embodiment, the method comprises:
(a) administering rosuvastatin at a first dose to a subject (i.e., patient) in need of therapy;
(b) determining the concentration of rosuvastatin the subject's blood at one or more time points after administration of rosuvastatin to provide a set of rosuvastatin concentration/time data points;
(c) transforming the set of rosuvastatin concentration/time data points to provide rosuvastatin area-under-the-curve (AUC); and
(d) administering rosuvastatin to the subject at subsequent doses (e.g., second and subsequent doses) to achieve a target rosuvastatin AUC of 80 ng*hr/mL.
In another aspect, the invention provides a method for anti-dyslipidemia drug dosing by one or more pharmacokinetic parameters. In one embodiment, the method comprises:
(a) administering an anti-dyslipidemia drug at a first dose to a subject in need of therapy;
(b) determining the concentration of the anti-dyslipidemia drug in the subject's blood at one or more time points after administration of the anti-dyslipidemia drug to provide a set of anti-dyslipidemia drug concentration/time data points;
(c) transforming the set of anti-dyslipidemia drug concentration/time data points to provide one or more pharmacokinetic parameters; and (d) administering the anti-dyslipidemia drug to the subject at subsequent doses
(e.g., second and subsequent doses) to achieve a target optimal value for the one or more pharmacokinetic parameters.
The one or more pharmacokinetic parameters can be one or more of concentration time course, peak concentration (C^x), and time after administration to peak concentration, terminal half-life, area-under-the-curve (AUC), bioavailability, absorption, distribution, metabolism, excretion, biotransformation, and combinations thereof. In one embodiment, the pharmacokinetic parameter is area-under-the-curve (AUC).
In certain embodiments, the target optimal value (rosuvastatin AUC) is ± 15% of the target optimal value. In other embodiments, the target optimal value (rosuvastatin AUC) is ± 20% of the target optimal value. In further embodiments, the target optimal value (rosuvastatin AUC) is ± 30% of the target optimal value.
In certain embodiments of the above methods, the second dose is the same or substantially the same as the first dose; in other embodiments, the second dose is greater than the first dose; and in further embodiments, the second dose is less than the first dose.
In certain embodiments, the above methods further including repeating steps (a)-(d) until dyslipidemia control is achieved.
In certain embodiments of the above methods, the subject is in need of treatment for dyslipidemia, and the method comprises administration of an anti-dyslipidemia drug.
BRIEF DESCRIPTION OF THE DRAWINGS
The foregoing aspects and many of the attendant advantages of this invention will become more readily appreciated as the same become better understood by reference to the following detailed description, when taken in conjunction with the accompanying drawings.
FIGURE 1 compares rosuvastatin AUC from three BE studies conducted in Asian and non-Asian populations.
FIGURE 2 compares mean single dose AUC of rosuvastatin at different dose strengths in Asian and non- Asian healthy subjects (AUC, area under the concentration- time curve; PK, pharmacokinetics; RC, rosuvastatin). The large solid circle indicates the rosuvastatin PK study in Korean subjects.
FIGURE 3 compares dose-normalized AUC of rosuvastatin at different dose strengths in Asian and non- Asian healthy subjects (AUC, area under the concentration- time curve; AUC/dose, dose-normalized area under the concentration-time curve; PK, pharmacokinetics; RC, rosuvastatin). The large solid circle indicates the rosuvastatin PK study in Korean subjects.
FIGURES 4A and 4B compare dose-normalized AUC of rosuvastatin (4A) and body weight (4B) among non- Asian, Asian and Korean (ST-101) subjects (AUC, area under the concentration-time curve; AUC/dose, dose-normalized area under the concentration-time curve; PK, pharmacokinetics; RC, rosuvastatin). The large solid circles indicate the rosuvastatin PK study in Korean subjects.
FIGURE 5 compares plots of AUC/dose of rosuvastatin against body weight with regression lines for Asian and non-Asian groups (AUC/dose, dose-normalized area under the concentration-time curve; solid dot, PK study in non- Asian subjects; circle, PK study in Asian subjects; triangle, ST101 PK study in Korean subjects).
FIGURE 6 illustrates mean rate of LDL-C reduction from clinical studies at different dose of rosuvastatin calcium (LDL-C, low-density lipoprotein cholesterol; LS, least squares; solid dot, PD study in non- Asian subjects; circle, PD study in Asian subjects; shaded circle, PD study in Korean subjects; triangle, ST101 PD study in Korean subjects). The regression line indicates the correlation between LDL-C reduction and dose.
FIGURE 7 compares plots of LDL-C reduction rate against dose with regression lines for Asian and non-Asian groups (LDL-C, low-density lipoprotein cholesterol; LS, least squares; solid dot, PD study in non- Asian subjects; circle, PD study in Asian subjects; shaded circle, PD study in Korean subjects; red triangle, ST101 PD study in Korean subjects).
DETAILED DESCRIPTION OF THE INVENTION
In one aspect, the invention provides methods for treating dyslipidemia by administering rosuvastatin. In the method, a dosing regimen targeting specific rosuvastatin AUC is provided in which AUC determined from first dosing with rosuvastatin is used to adjust subsequent rosuvastatin dosing to achieve the targeted AUC. The targeted AUC dosing regimen for rosuvastatin was made taking into account the targeted AUC value derived from rosuvastatin pharmacokinetic studies.
In certain embodiments, the pharmacokinetic parameter used in the method is area-under-the-curve (AUC).
The methods of the invention are effective in treating dyslipidemia. As noted above, the high PK variability and high drug exposure and dose non-proportionality of rosuvastatin require repeated drug monitoring following dosing to achieve the desired AUC (i.e., target AUC).
Pharmacokinetic Variability
Anti-dyslipidemia drug pharmacokinetic variability was determined for rosuvastatin, a representative anti-dyslipidemia drug.
Anti-Dyslipidemia Drug Dose Response Correlation
The high variability of PK and the resulting pharmacodynamics (PD) of rosuvastatin can result in weak correlation between dose and response. Furthermore, as described herein, differences between Asian and non- Asian subjects was observed.
Determination of First Pharmacokinetic Parameter and Prediction of Subsequent Pharmacokinetic Parameter
There is no known natural law to predict anti-dyslipidemia drug (e.g., rosuvastatin) pharmacokinetic parameters (e.g., AUC). The complexity of pharmacokinetics precludes any predictive methods for the accurate prediction of human rosuvastatin pharmacokinetics. The only means of predicting the pharmacokinetic parameters is to actually perform the pharmacokinetic study, which is shown herein to be predictive of subsequent pharmacokinetic parameter determination.
Anti-dyslipidemia Drug Dose Non-Proportionality in Asian Populations
A demonstration of anti-dyslipidemia drug dose proportionality allows for AUC dosing. However, as described herein, this was not observed for the Asian population.
Anti-dyslipidemia Drug Dosing by AUC
Given that subsequent pharmacokinetic parameters (e.g., AUC) can only be estimated by first pharmacokinetic parameter quantitation, the method of the invention provides a method that improves the delivery of anti-dyslipidemia drugs in general and rosuvastatin in particular.
In a representative method, rosuvastatin is administered to a subject to achieve a target AUC, which is defined as the median of conventional rosuvastatin PK (AUC) at conventional mg/day dosing (e.g., 80 ng*hr/mL ± 15%). If dosing is below or above the target AUC, dose adjustment is adjusted up (increased) or down (decreased), respectively.
The target AUC dosing can be varied to higher or lower target AUC when the patient is demonstrating resistance or sensitivity, respectively, to the anti-dyslipidemia drug. Regardless, once the targeted AUC for the patient has been defined, the target AUC needed to be maintained despite changes in physical condition (weight) and physiological condition (kidney, liver functional status etc.). Variables such as weight and organ function factors are known to affect rosuvastatin pharmacokinetics. However, with frequent monitoring, it is possible to keep dosing at constant AUC by appropriate adjustment of dosing when changes are noted.
The methods of the invention effectively administer anti-dyslipidemia drugs (e.g., rosuvastatin) by pharmacokinetic parameter (e.g., AUC) dosing to avoid the problems associated with pharmacokinetic variability described above. Pharmacokinetic variability is avoided in the method by adjusting drug dose to achieve a targeted AUC.
Rosuvastatin Dosing by AUC
In one aspect, the invention provides a method for rosuvastatin dosing by AUC. In certain embodiments, the method comprises:
(a) administering rosuvastatin at a first dose (e.g., under a first regimen, such as once a day) to a subject in need of therapy;
(b) determining the concentration of rosuvastatin in the subject's blood at one or more time points (e.g., a series of time points, such as 0.5, 1, 2, 4, 8, 12, 16, hour) after rosuvastatin administration to provide a set of rosuvastatin concentration/time data points;
(c) transforming the set of rosuvastatin concentration/time data points to provide area-under-the-curve (AUC) (i.e., the AUC resulting from the first dose); and (d) administering rosuvastatin at subsequent doses (e.g., a second dose and subsequent doses) to achieve a target rosuvastatin AUC (i.e., 80 ng*hr/mL ± 15%).
In certain embodiments, the target rosuvastatin AUC is ± 15% of the target. In other embodiments, the target rosuvastatin AUC is ± 20% of the target. In further embodiments, the target rosuvastatin AUC is ± 30% of the target.
Because of dose non-proportionality, determination of the AUC following the second dose (and subsequent does, as necessary) is required. Although the dose adjustment (e.g., reduction) can be estimated from a dose response curve, the dose adjustment cannot be accurately determined from the curve.
Area-under-the-curve (AUC) is a pharmacokinetic parameter that is used in the method of the invention to dose olmesartan. As used herein, the term "area under the curve (AUC)" is the area under the curve in a plot of concentration of drug in blood plasma as a function of time. Typically, the area is calculated starting at the time the drug is administered and ending when the concentration in plasma is negligible. AUC represents the total drug exposure over time.
As used herein, the phrase "transforming the concentration/time data points" refers to the application of mathematical operations, formulas, theories, and/or principles (i.e., a formula for calculating AUC) to the concentrations/time data points of the individual subject to provide AUC.
The target rosuvastatin AUC was determined from statistical analysis of a subject population receiving rosuvastatin. The target AUC is the median AUC value determined from a population of subjects receiving rosuvastatin at a conventional dose (20 mg/day, daily administration) among non- Asian/Caucasian patients.
In the methods of the invention, the nature of the device or method for determining the concentrations/time data points for calculating AUC is not critical. Some methods and devices for determining therapeutic drug (e.g., rosuvastatin) concentrations are known in the art and can be used. In certain embodiments, a point-of-care device can be used to determine the concentrations and create the concentration/time data, transmit the data to a central location, and/or transmitting instructions to the patient to alter the administration.
In certain embodiments, the device and method for determining the concentrations/time data points for calculating AUC is an immunoassay assay device and method that utilizes one or more rosuvastatin antibodies (e.g., monoclonal antibodies) or functional fragments thereof. In certain of these embodiments, the device is a lateral flow device.
The method of the invention is therapeutically effective for delivery of rosuvastatin and therefore is effective for treating dyslipidemia.
The above method is also effective for treating subjects in need of combined hypertension and dyslipidemia therapy. In certain embodiments, the subject treatable by the method is a subject that is in need of treatment for hypertension and dyslipidemia. In certain embodiments of this method, olmesartan and an anti-dyslipidemia drug are individually administered. In other embodiments of this method, a single dosage form that comprises olmesartan and an anti-dyslipidemia drug (e.g., rosuvastatin or a salt thereof) is administered. In certain embodiments of this method, the single dosage form comprises olmesartan and rosuvastatin calcium.
Anti-dyslipidemia Drug Dosing by AUC In another aspect, the invention provides a method for anti-dyslipidemia drug dosing by AUC. In certain embodiments, the method comprises:
(a) administering an anti-dyslipidemia drug at a first dose to a subject in need of therapy;
(b) determining the concentration of the anti-dyslipidemia drug the subject's blood at one or more time points after anti-dyslipidemia drug administration to provide a set of anti-dyslipidemia drug concentration/time data points;
(c) transforming the set of anti-dyslipidemia drug concentration/time data points to provide one or more pharmacokinetic parameters; and
(d) administering the anti-dyslipidemia drug at subsequent doses (e.g., a second dose and subsequent doses) to the subject to achieve a target optimal value for the one or more pharmacokinetic parameters.
As noted above, the target pharmacokinetic parameter is the pre-determined optimal value. In certain embodiments, the target pharmacokinetic parameter is the pre-determined optimal value ±15%. In other embodiments, the target pharmacokinetic parameter is the pre-determined optimal value ±20%. In further embodiments, the target pharmacokinetic parameter is the pre-determined optimal value ±30%.
In certain embodiments, the method further comprising repeating steps (a)-(d) until the target pharmacokinetic parameter value(s) is achieved.
Any suitable pharmacokinetic (PK) parameter or parameters can be used in accordance with this aspect of the invention, including without limiting concentration, concentration time course, peak concentration, and time after administration to peak concentration, terminal half-life, area-under-the-curve (AUC), bioavailability, absorption, distribution, metabolism, excretion, biotransformation, or a combination thereof.
As used herein, the phrase "transforming the concentration/time data points" refers to the application of mathematical operations, formulas, theories, and/or principles (e.g., a formula for calculating AUC) to the concentrations/time data points of the individual subject to provide the pharmacokinetic value (e.g., AUC).
The target pharmacokinetic value is pre-determined by statistical analysis from a population of subjects receiving the anti-dyslipidemia drug at its optimal dose. The term "optimal dose" refers to a dose (e.g., mg/day) associated with desirable drug efficacy at lower risk doses of a drug (e.g., the Cmax range corresponding to patients experiencing high drug efficacy at a low dose) and is determined from a statistical analysis of a subject population receiving doses of the anti-dyslipidemia drug for whom there was therapeutic improvement without significant adverse drug reactions or significant side effects. Significant adverse drug reactions refer to ADRs that the subject finds intolerable, impair physiologic functions, and put the subject at risk for immobility and/or death or combinations thereof. Significant side effects refer to side effects that the subject finds intolerable, impair physiologic functions, and put the patient at risk for immobility and/or death or combinations thereof.
As noted above, in the methods of the invention, the nature of the device or method for determining the concentrations/time data points for calculating the pharmacokinetic parameter is not critical. Methods and devices for determining therapeutic drug (e.g., anti-dyslipidemia drugs) concentrations are known in the art and can be used. In certain embodiments, a point-of-care device can be used.
In certain embodiments, the device and method for determining the concentrations/time data points for calculating the pharmacokinetic parameter is an immunoassay assay device and method that utilizes one or more dyslipidemia antibodies (e.g., monoclonal antibodies) or functional fragments thereof. In certain of these embodiments, the device is a lateral flow device.
The above method is also effective for treating subjects in need of combined hypertension and dyslipidemia therapy. In certain embodiments, the subject treatable by the method is a subject that is in need of treatment for hypertension and dyslipidemia. In certain embodiments of this method, an antihypertensive drug and an anti-dyslipidemia drug are individually administered. In other embodiments of this method, a single dosage form that comprises an antihypertensive drug and an anti-dyslipidemia drug (e.g., rosuvastatin or a salt thereof) is administered. In certain embodiments of this method, the single dosage form comprises olmesartan and rosuvastatin.
Hypertension and Dyslipidemia
An estimated 40 to 45 percent of hypertensive patients also suffer from dyslipidemia. Because it is considered advantageous to treat patients suffering from hypertension and dyslipidemia with a single therapeutic agent, combinations of therapeutic agents in single dose form have been developed for concomitantly treating both diseases. Combination formulations of antihypertensive and antihyperlipidemic agents are described in WO 95/26188, WO 97/37688, WO 99/11260, WO 00/45818, WO 04/062729, and WO 06/040085. One such single dose form is Caduet , which is a clinically useful combination formulation of atorvastatin and amlodipine.
Rosuvastatin, an HMG-CoA reductase inhibitor, is useful for the treatment of hypercholesterolemia, hyperlipoproteinemia, and atherosclerosis; and rosuvastatin's calcium salt is commercially available under the designation Crestor™. Olmesartan medoxomil is useful for the treatment of essential hypertension and is commercially available under the designation Benicar™. When a single matrix formulation of rosuvastatin and olmesartan medoxomil is administered, a drug-drug interaction (DDI) between rosuvastatin and olmesartan medoxomil occurs that results in delaying the in vivo release (i.e., dissolution) of rosuvastatin calcium to the gastrointestinal fluid and thus delaying the translocation thereof to the gastrointestinal membrane, inhibiting the absorption of rosuvastatin.
Olmesartan
Olmesartan medoxomil formulations should be designed so as to exhibit high dissolution rate of olmesartan medoxomil in an in vitro comparative dissolution test, in order to obtain a bioequivalent formulation to other single formulations containing olmesartan medoxomil. In order to obtain the high in vitro dissolution rate, olmesartan medoxomil tablet comprises a preferred disintegrant, which may be one or more selected from the group consisting of low substituted hydroxypropyl cellulose, carboxymethylcellulose calcium, croscarmellose sodium, crospovidone, sodium starch glycolate, and pregelatinized starch. In one embodiment, olmesartan medoxomil tablet comprises 7.5 or more % by weight of low substituted hydroxypropyl cellulose, 5 or more % by weight of carboxymethylcellulose calcium, 15 or more % by weight of croscarmellose sodium, 10 or more % by weight of crospovidone, 5 or more % by weight of sodium starch glycolate, or 5 or more % by weight of pregelatinized starch, based on the total weight of the tablet comprising olmesartan medoxomil. In another embodiment, the compartment comprising olmesartan medoxomil comprises 7.5 to 65 % by weight of low substituted hydroxypropyl cellulose, 5 to 60 % by weight of carboxymethylcellulose calcium, 15 to 30 % by weight of croscarmellose sodium, 10 to 40 % by weight of crospovidone, 5 to 40 % by weight of sodium starch glycolate, or 5 to 60 % by weight of pregelatinized starch, based on the total weight of the tablet comprising olmesartan medoxomil. In a further embodiment, the tablet comprising olmesartan medoxomil comprises 7.5 to 65 % by weight, preferably 10 to 60 % by weight, more preferably about 20 ± 1 % by weight of low substituted hydroxypropyl cellulose, based on the total weight of the tablet comprising olmesartan medoxomil.
Olmesartan/rosuvastatin FDC
An improved pharmaceutical composition that is a single dosage form of olmesartan medoxomil and rosuvastatin or its salts is described in WO 2013/147462. This single dosage form comprises separate compartments for each drug in which each drug is separately and independently formulated. When the single dosage form is administered the interaction to in vivo absorption is minimized and the combination formulation is bioequivalent to the single formulation of each of drugs.
In certain embodiments, the subject treatable by the methods of the invention is a subject that is in need of treatment for hypertension and dyslipidemia. In certain embodiments of this method, an antihypertensive drug and an anti-dyslipidemia drug are individually administered. In other embodiments of this method, a single dosage form that comprises an antihypertensive drug and an anti-dyslipidemia drug (e.g., rosuvastatin or a salt thereof) is administered. In certain embodiments of this method, the single dosage form comprises olmesartan and rosuvastatin.
A description of representative single dosage forms useful in the methods of the invention and methods for making the single dose forms are described in WO 2013/147462, expressly incorporated herein by reference in its entirety. Representative single dose forms useful in the method of the inventions and a method for making them are described below.
The pharmaceutical composition useful in the methods of the invention, which includes olmesartan medoxomil and rosuvastatin or its salt (e.g., rosuvastatin calcium), are formulated into a combination dosage form having separate compartments. That is, the pharmaceutical composition has a single dosage form comprising a compartment comprising olmesartan medoxomil; and a compartment comprising rosuvastatin or its salt, wherein the compartments are formulated in a separate form.
In the pharmaceutical composition, the active ingredients (i.e., olmesartan medoxomil and rosuvastatin or its salt) may be used in a therapeutically effect amount. For example, olmesartan medoxomil may be used in an amount of about 5 mg to about 80 mg, preferably about 10 mg to about 40 mg, in a unit formulation (i.e., unit dosage form).; and rosuvastatin or its salt may be used in an amount of about 2 mg to about 40 mg, preferably about 5 mg to about 20 mg, in a unit formulation (i.e., unit dosage form). The salt of rosuvastatin may be a conventional pharmaceutically acceptable salt, such as calcium salt, hydrochloride, hydrobromide, sulfate, phosphate, acetate, maleate, fumarate, lactate, tartrate, citrate, gluconate, besylate, and camsylate. Preferably, rosuvastatin calcium may be used in the present invention. The pharmaceutical composition may be administered once a day, but not limited thereto.
The pharmaceutical composition has a combination dosage form having separate compartments (i.e., a double-layered tablet form), comprising or consisting essentially of a layer comprising rosuvastatin or its salt and a layer comprising olmesartan medoxomil.
When the compartment comprising rosuvastatin or its salt includes a certain disintegrant (i.e., cellulose-type and/or povidone-type disintegrants), in a certain amount, rapid disintegration and high initial dissolution rate of rosuvastatin or its salt can be accomplished, thereby being able to obtain a combination formulation bioequivalent to the single formulation of rosuvastatin or its salt. The disintegrant may be one or more selected from the group consisting of povidone (for example, Kolidone™), crospovidone (for example, Polyplasdone™), low substituted hydroxypropyl cellulose, croscarmellose sodium, and carboxymethylcellulose calcium. Preferably, the disintegrant may be a mixture of crospovidone and croscarmellose sodium; or croscarmellose sodium. The disintegrant may be present in an amount ranging from 2 to 20 % by weight, preferably from 3 to 15 % by weight, based on the total weight of the compartment comprising rosuvastatin or its salt. When other disintegrants are used, the dissolution rate of rosuvastatin or its salt is decreased; and/or the amount used is increased, which may cause insufficient compression force during the compressing step, thereby leading to high friability of the resulting formulation (e.g., tablet). In addition, the use of other disintegrants brings about insufficient hardness, which may cause unwanted problems in packaging or delivery.
For olmesartan medoxomil, a combination formulation comprising rosuvastatin and olmesartan medoxomil should be designed so as to exhibit high dissolution rate of olmesartan medoxomil in an in vitro comparative dissolution test, in order to obtain a bioequivalent formulation to the single formulation containing olmesartan medoxomil. In order to obtain the high in vitro dissolution rate, the compartment comprising olmesartan medoxomil comprises a preferred disintegrant, which may be one or more selected from the group consisting of low substituted hydroxypropyl cellulose, carboxymethylcellulose calcium, croscarmellose sodium, crospovidone, sodium starch glycolate, and pregelatinized starch. In an embodiment, the compartment comprising olmesartan medoxomil comprises 7.5 or more % by weight of low substituted hydroxypropyl cellulose, 5 or more % by weight of carboxymethylcellulose calcium, 15 or more % by weight of croscarmellose sodium, 10 or more % by weight of crospovidone, 5 or more % by weight of sodium starch glycolate, or 5 or more % by weight of pregelatinized starch, based on the total weight of the compartment comprising olmesartan medoxomil. In another embodiment, the compartment comprising olmesartan medoxomil comprises 7.5 to 65 % by weight of low substituted hydroxypropyl cellulose, 5 to 60 % by weight of carboxymethylcellulose calcium, 15 to 30 % by weight of croscarmellose sodium, 10 to 40 % by weight of crospovidone, 5 to 40 % by weight of sodium starch glycolate, or 5 to 60 % by weight of pregelatinized starch, based on the total weight of the compartment comprising olmesartan medoxomil. In a further embodiment, the compartment comprising olmesartan medoxomil comprises 7.5 to 65 % by weight, preferably 10 to 60 % by weight, more preferably about 20 ± 1 % by weight of low substituted hydroxypropyl cellulose, based on the total weight of the compartment comprising olmesartan medoxomil.
The pharmaceutical composition may further comprise one or more excipients conventionally used in the field of pharmaceutics, for example a diluent (or additive), a binder, a lubricant, in addition to said disintegrant. The pharmaceutical composition may be also coated with an appropriate coating agent, such as a film-coating agent.
The diluent (or additive) includes lactose (including its hydrate), dextrin, mannitol, sorbitol, starch, microcrystalline cellulose (for example, Celphere™), silicified microcrystalline cellulose (for example, Prosolv™), calcium hydrogen phosphate (including its hydrate), anhydrous calcium hydrogen phosphate, calcium carbonate, saccharides, and a mixture thereof. The binder includes polyvinylpyrrolidone, copovidone, gelatin, starch, sucrose, methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl alkylcellulose (for example, hydroxypropyl methylcellulose), and a mixture thereof. The lubricant includes stearic acid, stearates (for example, magnesium stearate), talc, corn starch, carnauba wax, light anhydrous silicic acid, magnesium silicate, synthetic aluminum silicate, hydrogenated oil, hydrogenated oil, titanium oxide, microcrystalline cellulose, macrogol 4000 or 6000, isopropyl myristate, calcium hydrogen phosphate, and a mixture thereof. The coating agent, for example a film-coating agent, includes a conventional polymer such as Opadry . The film-coating agent may be used in a minimum amount for providing an appropriate size of the formulation, but not limited thereto.
The pharmaceutical composition having a double-layered tablet form may be prepared by preparing granules containing rosuvastatin and granules containing olmesartan medoxomil, respectively; and then compressing the mixture thereof with a double-layer tablet-press machine. If necessary, the resulting double-layered tablet may be coated with a film-coating agent such as Opadry™. The granules containing rosuvastatin and the granules containing olmesartan medoxomil may be prepared according to dry granulation methods or wet granulation methods. For example, the granules containing rosuvastatin may be prepared according to a dry granulation method. That is, the granules containing rosuvastatin may be prepared by mixing rosuvastatin calcium, an additive (diluent), a disintegrant, and a lubricant according to a conventional method; and then granulating the mixture with e.g., a roller compactor (TF mini, Vector). And also, the granules containing olmesartan medoxomil may be prepared according to a wet granulation method. That is, the granules containing olmesartan medoxomil may be prepared by mixing olmesartan medoxomil, a binder, an additive (diluent), a disintegrant; granulating the mixture with a high speed mixer (MIC Developer-5, COMASA); and then drying and sieving the resulting granules.
Representative double-layer tablets can be prepared as described below.
Step 1. Preparation of granules containing rosuvastatin.
Rosuvastatin calcium, lactose monohydrate, Prosolv™, dibasic calcium phosphate dihydrate, crospovidone, croscarmellose sodium, light anhydrous silicic acid, and magnesium stearate (85% of the total amount used in the rosuvastatin-layer) were sieved through a 24 mesh and then mixed. The resulting mixture was granulated using a roller compactor (TF mini, Vector). The obtained granules were sieved through a 24 mesh and then mixed with magnesium stearate pre-sieved though a 35 mesh (15% of the total amount used in the rosuvastatin-layer) to prepare a rosuvastatin-containing granule mixture.
Step 2. Preparation of granules containing olmesartan medoxomil.
Olmesartan medoxomil, hydroxypropyl cellulose, lactose monohydrate, microcrystalline cellulose, and low substituted hydroxypropyl cellulose were sieved through a 24 mesh and then mixed. The resulting mixture was granulated using a high speed mixer (MIC Developer-5, COMASA). The resulting dry granules were sieved through a 24 mesh and then mixed with magnesium stearate pre-sieved though a 35 mesh and yellow iron oxide pre-sieved through an 80 mesh to prepare a olmesartan medoxomil-containing granule mixture.
Step 3. Preparation of double-layered tablets.
The rosuvastatin-containing granule mixture prepared in Step 1 and the olmesartan medoxomil-containing granule mixture prepared in Step 2 were compressed with a double-layer tablet-press machine (BB-11, RIVA) to obtain double-layered tablets. The resulting tablets were film-coated with Opadry™ in a pan coating machine (LDCS, VECTOR).
The following is a description of a rosuvastatin clinical trials that demonstrates race (non-Asi an/Caucasian) is an important factor in rosuvastatin pharmacokinetics (PK) and pharmacodynamics (PD).
Hyperlipidemia, Hypertriglyceridemia, Dysbetalipoproteinemia,
Hypercholesterolemia, and Atherosclerosis
Hyperlipidemia is a condition in which blood lipid levels increase due to abnormal lipid metabolism of triglycerides, cholesterol, and other fatty acids. Hyperlipidemia can be caused by genetic factors or by secondary factors such as hypothyroidism, hepatic disease, nephrotic syndrome, or diabetes mellitus. Serum lipids primarily comprise cholesterol, triglycerides, phospholipids, and free fatty acids transported in a lipoprotein form. Lipoproteins are categorized into 4 types as follows: chylomicrons, very low-density lipoprotein (VLDL), LDL, and HDL. VLDLs are synthesized by the liver in the fasting state and are responsible for transporting triglycerides to peripheral tissues where some are converted to LDLs. LDLs are the major carriers of cholesterol to peripheral tissues; however, they are also a major risk factor for coronary arteriosclerosis.
In the US, approximately 73.5 million adults (31.7%) have high LDL, and only 29.5%) of affected adults have the condition under control (Mozaffarian et al. 2015; CDC MMWR 2014). An estimated 48.1% of US adults with high LDL cholesterol receive treatment to decrease their levels (Mozaffarian et al. 2015). Individuals with elevated cholesterol have an approximately 2-times greater risk of developing heart disease compared with individuals with normal cholesterol levels (CDC 201 la).
Treatment for hyperlipidemia includes lifestyle changes such as dietary modification, weight management, and physical activity to decrease LDL levels. Cholesterol-lowering agents (e.g., statins, bile acid sequestrants, nicotinic acid, fibrates, and ezetimibe) can be prescribed in addition to enacting lifestyle changes. Rosuvastatin calcium is a statin drug (HMG-CoA reductase inhibitor) that decreases LDL cholesterol by inhibiting HMG-CoA reductase, which is an enzyme in the metabolic pathway for cholesterol production (Olsson et al. 2002b).
PK Studies
A series of PK studies were performed to characterized the PK of rosuvastatin in Asian and non-Asian populations. The first clinical bioequivalent (BE) study was conducted in Korean population (Korean BE study). This was a randomized, open Label, single-dose, 2-way cross-over clinical trial to compare the safety and pharmacokinetic characteristics of combination therapy of rosuvastatin calcium (RC) 20 mg and olmesartan medoxomil (OM) 40 mg (Reference drug, RC+OM) to monotherapy of ST-101 (as used herein "ST-101 " refers to Test drug, fixed dose combination of OM 40 mg and RC 20 mg) in healthy male volunteers. 54 subjects in total had completed the study. In terms of pharmacokinetic assessment, the geometric mean ratios of test drug and the reference drug in connection of AUCiast and CmaX for rosuvastatin were 91.31% and 90.37%), respectively. The 90% confidence intervals of the geometric mean ratios of test drug and the reference drug were within the bioequivalence acceptance range (80.00-125.00%).
The second BE study was conducted in Indian population (Indian BE study). This was an open label, balanced, randomized, two-treatment, two-sequence, two-period, crossover, single-dose oral BE study of ST-101 (Test drug, fixed dose combination of RC/OM 10/20 mg) and co-administration of RC 10 mg and OM 20 mg (Reference drug) in healthy, adult, male, human subjects under fasting conditions. 26 subjects had completed the study. The geometric mean ratios of test drug and the reference drug in connection of AUCiast and Cmax for rosuvastatin were 99.80%) and 99.40%), respectively. The 90% confidence intervals of the geometric mean ratios of test drug and the reference drug were within the bioequivalence acceptance range.
The third BE study was conducted in non-Asian population in Czech Republic (EU BE study). This was a single-dose, randomized, two-period, two-treatment, two-sequence, crossover BE study on ST-101 (test drug, fixed dose combination of OM/RC 40 mg/20 mg) versus two co-administered reference products OM 40 mg and RC 20 mg in healthy volunteers under fasting conditions. 56 subjects had completed the study. The geometric mean ratios of test drug and the reference drug in connection of AUCiast and C max for rosuvastatin were 100.16 % and 98.54 %, respectively. The 90% confidence intervals of the geometric mean ratios of test drug and the reference drug were within the bioequivalence acceptance range.
The PK data from the three BE studies are summarized in the Table 1 and
FIGURE 1.
The median and mean AUC of rosuvastatin from the Korean, Indian and EU BE
Figure imgf000018_0001
The target AUC was defined as Caucasian median AUC at 20 mg/day. This was rounded up to 80 mg*hr/mL.
Analysis of rosuvastatin PK-demonstration of higher drug exposure and dose non-proportional PK for Asian population
PubMed database was searched to identify relevant scientific and review articles from October 2002 to March 2015 using the search term rosuvastatin PK. Reference lists of included articles were reviewed to locate additional references of interest. PK studies with single dose AUC of rosuvastatin calcium in healthy subjects were selected. Rosuvastatin AUC from co-administration of rosuvastatin calcium with other drugs were also included if there was no known DDI. More than 50% of the studies reported AUC in geometric mean. Thus, to make comparable analysis, only studies with AUC calculated by geometric mean were included in the analysis. PK and demographic data from a total of 745 healthy subjects in 11 clinical studies were included in the final analysis: CSR Synopsis DW_DWJ 1276002; Cooper et al. 2002; Martin et al. 2003a; Martin et al. 2003b; Schneck et al. 2004; Lee et al. 2005; Li et al. 2007; Li et al. 2010b; Trabelsi et al. 2012; Birmingham et al. 2015b). Birmingham et al 2015b reported rosuvastatin PK in Korean subjects.
Dose proportional analysis was performed to confirm reported dose proportional nature of rosuvastatin PK. The AUC/dose was then used in multivariate correlation to define the variable that has significant impact on PK. The analysis yielded significant correlation between weight and race to AUC/dose, but not age. ANCOVA analysis was then performed to show that race is a significant factor even after weight was taken in consideration. However, there was no observed difference between rosuvastatin PK in ST-101 population versus Asian population. This allowed us to bridge ST-101 PK data to Asian PK data and to Caucasian PK data.
The mean AUCiast of rosuvastatin from each study were plotted versus dose (FIGURE 2). The plot showed that the mean AUCs of rosuvastatin in Asian subjects are higher than the mean AUCs in non- Asian subjects at the same dose. Rosuvastatin AUC from subjects treated with ST-101 overlapped with AUC from 20 mg of rosuvastatin in Asian subjects.
There is a linear dose-dependent increase in AUC from 5 to 80 mg of rosuvastatin in non- Asian and from 5 to 40 mg in Asian subjects. Dose proportional analysis was performed by demonstrating constant AUC/dose across doses. The dose-normalized AUC was plotted versus dose (FIGURE 3). The linear regression line of AUC/dose against dose was horizontal with 95% CIs of slopes close to 0 for both Asian (-0.24 to 0.05; p=0.20) and non-Asian groups (-0.02 to 0.02; p = 0.92). The observed dose proportionality is similar to previous reports (Martin et al. 2003a, CRESTOR® Prescribing Information 2014). The dose proportionality nature of rosuvastatin PK validated the use of dose-normalized AUC for ANCOVA. Compared to Asian subjects, non -Asian subjects have a significantly lower dose-normalized AUC of rosuvastatin (p < 0.0001), while dose-normalized AUCs of rosuvastatin from ST-101 administered or from olmesartan medoxomil/rosuvastatin calcium co-administered in Korean subjects are comparable to reported AUCs of rosuvastatin of Asian subjects (p = 0.50) (FIGURE 4A). Additionally, Asian and non-Asian subjects were significantly different in BW (p < 0.0001), with BWs of Korean subjects in the ST-101 studies being similar to other Asian subjects (p = 0.34) (FIGURE 4B).
After confirming no significant interaction between BW and race, ANCOVA was conducted to determine whether there was a significant difference between Asian and non- Asian subjects on dose-normalized AUC after controlling for BW, using Fit Model Platform (JMP version 11.2.1 clinical software). The number of subjects in each study was used to assign a weight to each study in the analysis. The slopes of the regression lines of dose-normalized AUC of rosuvastatin against BW are significantly different between Asian and non-Asian groups (p = 0.02) (FIGURE 5). This implies that dose- normalized AUC of rosuvastatin are different between Asian and non-Asian groups even after taking BW into account, indicating that race is an important determinant of drug exposure. This finding is well described in a population PK model (Tzeng et al. 2008). In this model, Asians were assigned 2 times greater drug exposure than Caucasians. Similar differences have been observed between Asian and white subjects living in Singapore, suggesting that these ethnic differences are independent of geographic residence (Lee et al. 2005). Birmingham et al. (2015a) affirmed a higher exposure to rosuvastatin in Asian subjects (Chinese, Filipinos, Asian Indians, Koreans, Vietnamese, and Japanese) residing in the US than in white subjects. Even though the differences in rosuvastatin exposure between Asian and non-Asian existed, rosuvastatin AUC from Korean subjects treated with ST-101 is the same as rosuvastatin AUC from Korean and other Asian subjects treated with rosuvastatin (FIGURE 4A), which is well understood.
The above results demonstrate that race is an important factor in rosuvastatin PK even after weight was taken into consideration.
Analysis of Rosuvastatin PD response
PubMed database was searched to identify relevant scientific and review articles from September 2001 to June 2015 using the search terms rosuvastatin efficacy, response, effect/clinical trial. Reference lists of included articles were reviewed to locate additional references of interest. PD studies with rosuvastatin calcium dose range from 1 to 80 mg monotherapy were selected. PD response and demographic data from a total of 17,395 patients with dyslipidemia in 52 clinical studies were included in the analysis: Clinical Study Report DW_DWJ1276003 Synopsis 2013; Olsson et al., 2001; Paoletti et al., 2001; Davidson et al., 2002; Brown et al. 2002; Olsson et al. 2002b; Blasetto et al., 2003; Ballantyne et al. 2003; Capuzzi et al., 2003; Saito et al., 2003; Jones et al., 2003; Schneck et al., 2003; Ballantyne et al. 2004; Durrington et al., 2004; Lu et al., 2004; Strandberg et al., 2004; Schuster et al., 2004; Schwartz et al., 2004; Bots and Kastelein 2005; Jukema et al., 2005; Fonseca et al., 2005; Clearfield et al., 2006; Glueck et al., 2006; Ballantyne et al., 2006; Catapano et al., 2006; Milionis et al., 2006; Wongwiwatthananukit et al., 2006; Asztalos et al., 2007; Insull et al., 2007; Leiter et al., 2007; Betteridge et al., 2007; Deedwania et al., 2007; Ballantyne et al., 2007; Saito et al., 2007; Zhu et al., 2007; Stein et al., 2007; Mazza et al., 2008; Talini et al., 2008; Faergeman et al., 2008; Sviridov et al., 2008; Underhill et al., 2008; Laks et al., 2008; Kurabayashi M and Yamazaki 2008; Riesen et al. 2008; Pirro et al., 2009; Qu et al., 2009; McCormack et al., 2010; Saku et al., 2011; Yanagi et al., 2011; Pitt et al., 2012; Hong et al. 2011, and Jang et al., 2015). Hong et al. (2011) and Jang et al. (2015) are studies conducted in Korea.
The mean reduction rate in LDL-C in patients from each study was plotted versus dose (FIGURE 6). The LDL-C reduction rate by ST-101 is similar to the effects by 20 mg of rosuvastatin calcium in Korean, other Asian, and non-Asian patients. Regression analysis showed a significant correlation between LDL-C reduction and dose (p<0.0001) (FIGURE 6). This dose-related response for rosuvastatin calcium has been reported previously (CRESTOR® Prescribing Information 2014).
The interaction between dose and race on the LDL-C reduction was evaluated. The number of subjects in each study was used to assign a weight to each study in the analysis. The slopes of the regression lines relate LDL-C reduction to dose were significantly different between Asian and non-Asian patients (p = 0.02; FIGURE 7). The slope for dose-response in Asian patients is much steeper than that in non-Asian patients, therefore, race is an important determinant of PD response for rosuvastatin.
A dose-related response for rosuvastatin has been described previously in both
Asian and non- Asian patients in a rosuvastatin PD model (Yang et al. 2011). The model has shown that the race differences in PD response of rosuvastatin are consistent with PK, and there is no significant difference in the exposure-response relationship for LDL-C reduction between the 2 groups.
The above results demonstrate that race is an important factor in rosuvastatin PD. References
Asztalos BF, Le Maulf F, Dallal GE, Stein E, Jones PH, Horvath KV, et al. Comparison of the effects of high doses of rosuvastatin versus atorvastatin on the subpopulations of high-density lipoproteins. Am J Cardiol. 2007 Mar 1; 99(5):681-685. Ballantyne CM, Bertolami M, Hernandez Garcia FIR, Nul D, Stein EA, Theroux
P, et al. Achieving LDL cholesterol, non-FIDL cholesterol, and apolipoprotein B target levels in high-risk patients: Measuring Effective Reductions in Cholesterol Using Rosuvastatin Therapy (MERCURY) II. Am Heart J. 2006 May; 151(5):975 e971-979. Ballantyne CM, Miller E, Chitra R. Efficacy and safety of rosuvastatin alone and in combination with cholestyramine in patients with severe hypercholesterolemia: a randomized, open-label, multi center trial. Clin Ther. 2004 Nov; 26(11): 1855-1864.
Ballantyne CM, Stein EA, Paoletti R, Southworth H, Blasetto JW. Efficacy of rosuvastatin 10 mg in patients with the metabolic syndrome. Am J Cardiol. 2003 Mar 6; 91(5A):25C-27C; discussion 28C.
Ballantyne CM, Weiss R, Moccetti T, Vogt A, Eber B, Sosef F, et al. Efficacy and safety of rosuvastatin 40 mg alone or in combination with ezetimibe in patients at high risk of cardiovascular disease (results from the EXPLORER study). Am J Cardiol. 2007 Mar 1; 99(5):673-680.
Betteridge DJ, Gibson JM, Sager PT. Comparison of effectiveness of rosuvastatin versus atorvastatin on the achievement of combined C-reactive protein (<2 mg/L) and low-density lipoprotein cholesterol (< 70 mg/dl) targets in patients with type 2 diabetes mellitus (from the ANDROMEDA study). Am J Cardiol. 2007 Oct 15; 100(8): 1245-1248. Birmingham BK, Bujac SR, Elsby R, Azumaya CT, Wei C, Chen Y, et al. Impact of ABCG2 and SLC01B1 polymorphisms on pharmacokinetics of rosuvastatin, atorvastatin and simvastatin acid in Caucasian and Asian subjects: a class effect? Eur J Clin Pharmacol. 2015a Mar; 71(3):341-355.
Birmingham BK, Bujac SR, Elsby R, Azumaya CT, Zalikowski J, Chen Y, et al. Rosuvastatin pharmacokinetics and pharmacogenetics in Caucasian and Asian subjects residing in the United States. Eur J Clin Pharmacol. 2015b Mar; 71(3):329-340. Blasetto JW, Stein EA, Brown WV, Chitra R, Raza A. Efficacy of rosuvastatin compared with other statins at selected starting doses in hypercholesterolemic patients and in special population groups. Am J Cardiol. 2003 Mar 6; 91(5A):3C-10C; discussion IOC. Bots AF, Kastelein JJ. Achieving lipid goals in real life: the Dutch DISCOVERY study. Int J Clin Pract. 2005 Dec; 59(12): 1387-1394.
Brown WV, Bays HE, Hassman DR, McKenney J, Chitra R, Hutchinson H, et al. Efficacy and safety of rosuvastatin compared with pravastatin and simvastatin in patients with hypercholesterolemia: a randomized, double-blind, 52-week trial. Am Heart J. 2002 Dec; 144(6): 1036-1043.
Capuzzi DM, Morgan JM, Weiss RJ, Chitra RR, Hutchinson HG, Cressman MD. Beneficial effects of rosuvastatin alone and in combination with extended-release niacin in patients with a combined hyperlipidemia and low high-density lipoprotein cholesterol levels. Am J Cardiol. 2003 Jun 1; 91(11): 1304-1310.
Catapano AL, Davidson MH, Ballantyne CM, Brady WE, Gazzara RA, Tomassini JE, et al. Lipid-altering efficacy of the ezetimibe/simvastatin single tablet versus rosuvastatin in hypercholesterolemic patients. Curr Med Res Opin. 2006 Oct; 22(10):2041-2053.
CDC2011a. CDC 201 la-Cholesterol levels sheet. 2011. CDC. CDC MMWR 2014. 2005-2013.
Clearfield MB, Amerena J, Bassand JP, Hernandez Garcia HR, Miller SS, Sosef FF, et al. Comparison of the efficacy and safety of rosuvastatin 10 mg and atorvastatin 20 mg in high-risk patients with hypercholesterolemia—Prospective study to evaluate the Use of Low doses of the Statins Atorvastatin and Rosuvastatin (PULSAR). Trials. 2006; 7:35. Cooper KJ, Martin PD, Dane AL, Warwick MJ, Schneck DW, Cantarini MV. The effect of fluconazole on the pharmacokinetics of rosuvastatin. Eur J Clin Pharmacol. 2002 Nov; 58(8):527-531.
CRESTOR® (rosuvastatin calcium) Prescribing information. AstraZeneca Pharmaceuticals LP (revised 2014). http://wwwl .astrazeneca-us.com/pi/crestor.pdf
DWJ1276002_CSR_V1.0_20130131_eng./ Internal data
DWJ1276003_CSR_V2.0_20131209(Eng)/Internal data
Davidson M, Ma P, Stein EA, Gotto AM, Jr., Raza A, Chitra R, et al. Comparison of effects on low-density lipoprotein cholesterol and high-density lipoprotein cholesterol with rosuvastatin versus atorvastatin in patients with type Ila or lib hypercholesterolemia. Am J Cardiol. 2002 Feb 1; 89(3):268-275.
Deedwania PC, Gupta M, Stein M, Yeas J, Gold A. Comparison of rosuvastatin versus atorvastatin in South-Asian patients at risk of coronary heart disease (from the IRIS Trial). Am J Cardiol. 2007 Jun 1; 99(11): 1538-1543. Durrington PN, Tuomilehto J, Hamann A, Kallend D, Smith K. Rosuvastatin and fenofibrate alone and in combination in type 2 diabetes patients with combined hyperlipidaemia. Diabetes Res Clin Pract. 2004 May; 64(2): 137-151. Faergeman O, Hill L, Windier E, Wiklund O, Asmar R, Duffield E, et al. Efficacy and tolerability of rosuvastatin and atorvastatin when force-titrated in patients with primary hypercholesterolemia: results from the ECLIPSE study. Cardiology. 2008; l l l(4):219-228.
Fonseca FA, Ruiz A, Cardona-Munoz EG, Silva JM, Fuenmayor N, Marotti M. The DISCOVERY PENTA study: a Direct Statin Comparison of LDL-C Value-an Evaluation of Rosuvastatin therapy compared with atorvastatin. Curr Med Res Opin. 2005 Aug; 21(8): 1307-1315.
Glueck CJ, Aregawi D, Agloria M, Khalil Q, Winiarska M, Munjal J, et al. Rosuvastatin 5 and 10 mg/d: a pilot study of the effects in hypercholesterolemic adults unable to tolerate other statins and reach LDL cholesterol goals with nonstatin lipid-lowering therapies. Clin Ther. 2006 Jun; 28(6):933-942.
Hong YJ, Jeong MH, Hachinohe D, Ahmed K, Choi YH, Cho SH, et al. Comparison of effects of rosuvastatin and atorvastatin on plaque regression in Korean patients with untreated intermediate coronary stenosis. Circ J. 2011; 75(2):398-406. Insull W, Jr., Ghali JK, Hassman DR, JW YA, Gandhi SK, Miller E. Achieving low-density lipoprotein cholesterol goals in high-risk patients in managed care: comparison of rosuvastatin, atorvastatin, and simvastatin in the SOLAR trial. Mayo Clin Proc. 2007 May; 82(5):543-550. Jang JY, Lee SH, Kim BS, Seo HS, Kim WS, Ahn Y, et al. Additive beneficial effects of valsartan combined with rosuvastatin in the treatment of hypercholesterolemic hypertensive patients. Korean Circ J. 2015 May; 45(3):225-233.
Jones PH, Davidson MH, Stein EA, Bays HE, McKenney JM, Miller E, et al. Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* Trial). Am J Cardiol. 2003 Jul 15; 92(2): 152-160. Jukema JW, Liem AH, Dunselman PH, van der Sloot JA, Lok DJ, Zwinderman AH. LDL-C/HDL-C ratio in subjects with cardiovascular disease and a low HDL-C: results of the RADAR (Rosuvastatin and Atorvastatin in different Dosages And Reverse cholesterol transport) study. Curr Med Res Opin. 2005 Nov; 21(11): 1865-1874.
Kurabayashi M, Yamazaki T, Group SS. Superior benefit of aggressive lipid-lowering therapy for high- risk patients using statins: the SUBARU study—more hypercholesterolemic patients achieve Japan Atherosclerosis Society LDL-C goals with rosuvastatin therapy than with atorvastatin therapy. J Atheroscler Thromb. 2008 Dec; 15(6):314-323.
Laks T, Keba E, Leiner M, Merilind E, Petersen M, Reinmets S, et al. Achieving lipid goals with rosuvastatin compared with simvastatin in high risk patients in real clinical practice: a randomized, open-label, parallel-group, multi-center study: the DISCOVERY-Beta study. Vase Health Risk Manag. 2008; 4(6): 1407-1416.
Lee E, Ryan S, Birmingham B, Zalikowski J, March R, Ambrose H, et al. Rosuvastatin pharmacokinetics and pharmacogenetics in white and Asian subjects residing in the same environment. Clin Pharmacol Ther. 2005 Oct; 78(4):330-341.
Leiter LA, Rosenson RS, Stein E, Reckless JP, Schulte KL, Schleman M, et al. Efficacy and safety of rosuvastatin 40 mg versus atorvastatin 80 mg in high-risk patients with hypercholesterolemia: results of the POLARIS study. Atherosclerosis. 2007 Oct; 194(2):el 54-164.
Li XN, Xu HR, Chen WL, Chu NN, Zhu JR. Pharmacokinetics of rosuvastatin in healthy Chinese volunteers living in China: a randomized, open-label, ascending single- and multiple-dose study. Clin Ther. 2010 Mar; 32(3):575-587.
Li Y, Jiang X, Lan K, Zhang R, Li X, Jiang Q. Pharmacokinetic properties of rosuvastatin after single-dose, oral administration in Chinese volunteers: a randomized, open-label, three-way crossover study. Clin Ther. 2007 Oct; 29(10):2194-2203. Lu TM, Ding YA, Leu HB, Yin WH, Sheu WH, Chu KM. Effect of rosuvastatin on plasma levels of asymmetric dimethylarginine in patients with hypercholesterolemia. Am J Cardiol. 2004 Jul 15; 94(2): 157-161. Martin PD, Warwick MJ, Dane AL, Brindley C, Short T. Absolute oral bioavailability of rosuvastatin in healthy white adult male volunteers. Clin Ther. 2003a Oct; 25(10):2553-2563.
Martin PD, Warwick MJ, Dane AL, Cantarini MV. A double-blind, randomized, incomplete crossover trial to assess the dose proportionality of rosuvastatin in healthy volunteers. Clin Ther. 2003b Aug; 25(8):2215-2224.
Mazza F, Stefanutti C, Di Giacomo S, Vivenzio A, Fraone N, Mazzarella B, et al. Effects of low-dose atorvastatin and rosuvastatin on plasma lipid profiles: a long-term, randomized, open-label study in patients with primary hypercholesterolemia. Am J Cardiovasc Drugs. 2008; 8(4):265-270.
McCormack T, Harvey P, Gaunt R, Allgar V, Chipperfield R, Robinson P. Incremental cholesterol reduction with ezetimibe/simvastatin, atorvastatin and rosuvastatin in UK General Practice (IN-PRACTICE): randomised controlled trial of achievement of Joint British Societies (JBS-2) cholesterol targets. Int J Clin Pract. 2010 Jul; 64(8): 1052-1061.
Milionis HJ, Rizos E, Kostapanos M, Filippatos TD, Gazi IF, Ganotakis ES, et al. Treating to target patients with primary hyperlipidaemia: comparison of the effects of ATOrvastatin and ROSuvastatin (the ATOROS study). Curr Med Res Opin. 2006 Jun; 22(6): 1123-1131.
Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, et al. Heart disease and stroke statistics—2015 update: a report from the American Heart Association. Circulation. 2015 Jan 27; 131(4):e29-322. Olsson AG, Istad H, Luurila O, Ose L, Stender S, Tuomilehto J, et al. Effects of rosuvastatin and atorvastatin compared over 52 weeks of treatment in patients with hypercholesterolemia. Am Heart J. 2002 Dec; 144(6): 1044-1051. Olsson AG, Pears J, McKellar J, Mizan J, Raza A. Effect of rosuvastatin on low- density lipoprotein cholesterol in patients with hypercholesterolemia. Am J Cardiol. 2001 Sep 1; 88(5):504-508.
Paoletti R, Fahmy M, Mahla G, Mizan J, Southworth H. Rosuvastatin demonstrates greater reduction of low-density lipoprotein cholesterol compared with pravastatin and simvastatin in hypercholesterolaemic patients: a randomized, double- blind study. J Cardiovasc Risk. 2001 Dec; 8(6):383-390.
Pirro M, Schillaci G, Romagno PF, Mannarino MR, Bagaglia F, Razzi R, et al. Influence of short-term rosuvastatin therapy on endothelial progenitor cells and endothelial function. J Cardiovasc Pharmacol Ther. 2009 Mar; 14(1): 14-21.
Pitt B, Loscalzo J, Monyak J, Miller E, Raichlen J. Comparison of lipid- modifying efficacy of rosuvastatin versus atorvastatin in patients with acute coronary syndrome (from the LUNAR study). Am J Cardiol. 2012 May 1; 109(9): 1239-1246.
Qu HY, Xiao YW, Jiang GH, Wang ZY, Zhang Y, Zhang M. Effect of atorvastatin versus rosuvastatin on levels of serum lipids, inflammatory markers and adiponectin in patients with hypercholesterolemia. Pharm Res. 2009 Apr; 26(4):958-964.
Riesen WF, Noll G, Dariolo R. Impact of enhanced compliance initiatives on the efficacy of rosuvastatin in reducing low density lipoprotein cholesterol levels in patients with primary hypercholesterolaemia. Swiss Med Wkly. 2008 Jul 26; 138(29-30):420-426. Saito Y, Goto Y, Dane A, Strutt K, Raza A. Randomized dose-response study of rosuvastatin in Japanese patients with hypercholesterolemia. J Atheroscler Thromb. 2003; 10(6):329-336. Saito Y, Yamada N, Shirai K, Sasaki J, Ebihara Y, Yanase T, et al. Effect of rosuvastatin 5-20mg on triglycerides and other lipid parameters in Japanese patients with hypertriglyceridemia. Atherosclerosis. 2007 Oct; 194(2):505-511. Saku K, Zhang B, Noda K, Investigators TPT. Randomized head-to-head comparison of pitavastatin, atorvastatin, and rosuvastatin for safety and efficacy (quantity and quality of LDL): the PATROL trial. Circ J. 2011; 75(6): 1493-1505.
Schneck DW, Birmingham BK, Zalikowski JA, Mitchell PD, Wang Y, Martin PD, et al. The effect of gemfibrozil on the pharmacokinetics of rosuvastatin. Clin Pharmacol Ther. 2004 May; 75(5):455-463.
Schneck DW, Knopp RH, Ballantyne CM, McPherson R, Chitra RR, Simonson SG. Comparative effects of rosuvastatin and atorvastatin across their dose ranges in patients with hypercholesterolemia and without active arterial disease. Am J Cardiol. 2003 Jan 1; 91(1):33-41.
Schuster H, Barter PJ, Stender S, Cheung RC, Bonnet J, Morrell JM, et al. Effects of switching statins on achievement of lipid goals: Measuring Effective Reductions in Cholesterol Using Rosuvastatin Therapy (MERCURY I) study. Am Heart J. 2004 Apr; 147(4):705-713.
Schwartz GG, Bolognese MA, Tremblay BP, Caplan R, Hutchinson H, Raza A, et al. Efficacy and safety of rosuvastatin and atorvastatin in patients with hypercholesterolemia and a high risk of coronary heart disease: a randomized, controlled trial. Am Heart J. 2004 Jul; 148(l):e4.
Stein EA, Amerena J, Ballantyne CM, Brice E, Farnier M, Guthrie RM, et al. Long-term efficacy and safety of rosuvastatin 40 mg in patients with severe hypercholesterolemia. Am J Cardiol. 2007 Nov 1; 100(9): 1387-1396.
Strandberg TE, Feely J, Sigurdsson EL. Twelve-week, multicenter, randomized, open-label comparison of the effects of rosuvastatin 10 mg/d and atorvastatin 10 mg/d in high-risk adults: a DISCOVERY study. Clin Ther. 2004 Nov; 26(11): 1821-1833. Sviridov D, Hoang A, Ooi E, Watts G, Barrett PH, Nestel P. Indices of reverse cholesterol transport in subjects with metabolic syndrome after treatment with rosuvastatin. Atherosclerosis. 2008 Apr; 197(2):732-739.
Talini E, Di Bello V, Bianchi C, Palagi C, Delle Donne MG, Penno G, et al. Early impairment of left ventricular function in hypercholesterolemia and its reversibility after short term treatment with rosuvastatin A preliminary echocardiographic study. Atherosclerosis. 2008 Mar; 197(l):346-354.
Trabelsi F, Bartunek A, Vlavonou R, Navratilova L, Dube C, Tanguay M, et al. Single-dose, 2-way crossover, bioequivalence study of two rosuvastatin formulations in normal healthy subjects under fasting conditions. Int J Clin Pharmacol Ther. 2012 Oct; 50(10):741-750.
Tzeng TB, Schneck DW, Birmingham BK, Mitchell PD, Zhang H, Martin PD, et al. Population pharmacokinetics of rosuvastatin: implications of renal impairment, race, and dyslipidaemia. Curr Med Res Opin. 2008 Sep; 24(9):2575-2585. Underhill HR, Yuan C, Zhao XQ, Kraiss LW, Parker DL, Saam T, et al. Effect of rosuvastatin therapy on carotid plaque morphology and composition in moderately hypercholesterolemic patients: a high-resolution magnetic resonance imaging trial. Am Heart J. 2008 Mar; 155(3):584 e581-588. Wongwiwatthananukit S, Sansanayudh N, Dhummauppakorn R, Kitiyadisai C.
Efficacy and safety of rosuvastatin every other day compared with once daily in patients with hypercholesterolemia. Ann Pharmacother. 2006 Nov; 40(11): 1917-1923.
Yanagi K, Monden T, Ikeda S, Matsumura M, Kasai K. A crossover study of rosuvastatin and pitavastatin in patients with type 2 diabetes. Adv Ther. 2011 Feb; 28(2): 160-171. Yang J, Li LJ, Wang K, He YC, Sheng YC, Xu L, et al. Race differences: modeling the pharmacodynamics of rosuvastatin in Western and Asian hypercholesterolemia patients. Acta Pharmacol Sin. 2011 Jan; 32(1): 116-125.
Zhu JR, Tomlinson B, Ro YM, Sim KH, Lee YT, Sriratanasathavorn C. A randomised study comparing the efficacy and safety of rosuvastatin with atorvastatin for achieving lipid goals in clinical practice in Asian patients at high risk of cardiovascular disease (DISCOVERY-Asia study). Curr Med Res Opin. 2007 Dec; 23(12):3055-3068.
While the preferred embodiment of the invention has been illustrated and described, it will be appreciated that various changes can be made therein without departing from the spirit and scope of the invention.

Claims

CLAIMS The embodiments of the invention in which an exclusive property or privilege is claimed are defined as follows:
1. A method for rosuvastatin dosing by AUC, comprising:
(a) administering rosuvastatin at a first dose to a subject in need of therapy;
(b) determining the concentration of rosuvastatin in the subject's blood at one or more time points after rosuvastatin administration to provide a set of rosuvastatin concentration/time data points;
(c) transforming the set of rosuvastatin concentration/time data points to provide area-under-the-curve (AUC); and
(d) administering rosuvastatin to the subject at one or more subsequent doses to achieve a target rosuvastatin AUC of 80 ng*hr/mL.
2. The method of Claim 1, wherein the target AUC is ±15% of the target value.
3. The method of Claim 1, wherein the target AUC is ±20% of the target value.
4. The method of Claim 1, wherein the target AUC is ±30% of the target value.
5. The method of Claim 1, wherein the second dose is substantially the same as the first dose.
6. The method of Claim 1, wherein the second dose is greater than the first dose.
7. The method of Claim 1, wherein the second dose is less than the first dose.
8. The method of Claim 1 further comprising repeating steps (a)-(d) until target rosuvastatin AUC is achieved.
9. The method of Claim 1, wherein the subject is in need of treatment for hypertension and dyslipidemia, and the method comprises administration of a single dosage form that comprises olmesartan and an anti-dyslipidemia drug.
10. The method of Claim 9, wherein single dosage form comprises olmesartan and rosuvastatin.
11. The method of Claim 1, wherein the hypertension is resistant hypertension.
12. A method for anti-dyslipidemia drug dosing by one or more pharmacokinetic parameters, comprising:
(a) administering an anti-dyslipidemia drug at a first dose to a subject in need of therapy;
(b) determining the concentration of the anti-dyslipidemia drug in the subject's blood at one or more time points after anti-dyslipidemia drug administration to provide a set of anti-dyslipidemia drug concentration/time data points;
(c) transforming the set of anti-dyslipidemia drug concentration/time data points to provide one or more pharmacokinetic parameters; and
(d) administering the anti-dyslipidemia drug to the subject at one or more subsequent doses to achieve a target optimal value for the one or more pharmacokinetic parameters.
13. The method of Claim 12, wherein the one or more pharmacokinetic parameters is selected from the group consisting of concentration time course, peak concentration (Cj^), and time after administration to peak concentration, terminal half-life, area-under-the-curve (AUC), bioavailability, absorption, distribution, metabolism, excretion, biotransformation, and combinations thereof.
14. The method of Claim 12, wherein the one or more pharmacokinetic parameters is area-under-the-curve (AUC).
15. The method of Claim 12, wherein the target optimal value is ±15% of the target optimal value.
16. The method of Claim 12 further comprising repeating steps (a)-(d) until dyslipidemia control is achieved.
17. The method of Claim 12, wherein the anti-dyslipidemia drug is a statin.
18. The method of Claim 12, wherein the anti-dyslipidemia drug is rosuvastatin.
19. The method of Claim 12, wherein the subject is in need of treatment for hypertension and dyslipidemia, and the method comprises administration of an antihypertensive drug and an anti-dyslipidemia drug.
20. The method of 12, wherein the subject is in need of treatment for hypertension and dyslipidemia, and the method comprises administration of a single dosage form that comprises an antihypertensive drug and an anti-dyslipidemia drug.
21. The method of Claim 20, wherein single dosage form comprises olmesartan and rosuvastatin.
22. The method of Claim 12, wherein the hypertension is resistant hypertension.
PCT/US2017/041162 2016-07-08 2017-07-07 Methods for rosuvastatin dosing by auc WO2018009842A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662360262P 2016-07-08 2016-07-08
US62/360,262 2016-07-08

Publications (1)

Publication Number Publication Date
WO2018009842A1 true WO2018009842A1 (en) 2018-01-11

Family

ID=60912300

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/041162 WO2018009842A1 (en) 2016-07-08 2017-07-07 Methods for rosuvastatin dosing by auc

Country Status (2)

Country Link
TW (1) TW201803565A (en)
WO (1) WO2018009842A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013101830A1 (en) * 2011-12-27 2013-07-04 Ironwood Pharmaceuticals, Inc. 2 - benzyl, 3 - (pyrimidin- 2 -yl) substituted pyrazoles useful as sgc stimulators
US20140349862A1 (en) * 2011-05-30 2014-11-27 Vuong Ngoc Trieu Methods and compositions for therapeutic drug monitoring and dosing by point of care pharmacokinetic profiling
US20150050333A1 (en) * 2012-03-30 2015-02-19 Daewoong Pharmaceutical Co., Ltd. Pharmaceutical composition comprising olmesartan medoxomil and rosuvastatin or its salt

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140349862A1 (en) * 2011-05-30 2014-11-27 Vuong Ngoc Trieu Methods and compositions for therapeutic drug monitoring and dosing by point of care pharmacokinetic profiling
WO2013101830A1 (en) * 2011-12-27 2013-07-04 Ironwood Pharmaceuticals, Inc. 2 - benzyl, 3 - (pyrimidin- 2 -yl) substituted pyrazoles useful as sgc stimulators
US20150050333A1 (en) * 2012-03-30 2015-02-19 Daewoong Pharmaceutical Co., Ltd. Pharmaceutical composition comprising olmesartan medoxomil and rosuvastatin or its salt

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JHARAHAP ET AL.: "Bioequivalence study of two rosuvastatin tablet formulations in healthy Indonesian subjects", INT J CLIN PHARMACOL THER, vol. 54, no. 3, 15 June 2015 (2015-06-15), pages 212 - 216 *
SON ET AL.: "Pharmacokinetics of rosuvastatin/olmesartan fixed-dose combination: a single-dose, randomized, open-label, 2-period crossover study in healthy Korean subjects", CLIN THER, vol. 35, no. 7, 27 June 2013 (2013-06-27), pages 915 - 922, XP028680042 *
ZAID ET AL.: "Investigation of the Bioequivalence of Rosuvastatin 20 mg Tablets after a Single ''Oral Administration in Mediterranean Arabs Using a Validated LC-MS/MS Method", SCI PHARM, vol. 84, no. 3, 30 June 2016 (2016-06-30), pages 536 - 546, XP055451935 *

Also Published As

Publication number Publication date
TW201803565A (en) 2018-02-01

Similar Documents

Publication Publication Date Title
US20230414521A1 (en) Sustained-release formulations of colchicine and methods of using same
US8758815B2 (en) Pharmaceutical compositions comprising a combination of metformin and sitagliptin
JP6068765B2 (en) Pharmaceutical combination preparation
JP2018507875A (en) Tesofensin and beta blocker combination
JP2011521977A (en) Modified release niacin formulation
CN114209667A (en) Delayed release pharmaceutical formulations comprising valproic acid and uses thereof
KR102306005B1 (en) Complex formulation comprising valsartan and rosuvastatin calcium and method for the preparation thereof
WO2018009842A1 (en) Methods for rosuvastatin dosing by auc
US20160199354A1 (en) Methods for olmesartan dosing by auc
JP6697539B2 (en) Colchicine sustained-release preparation and method of using the same
EP2763662A1 (en) Pharmaceutical compositions of antihypertensives
TW202224681A (en) Single dosage form of a pharmaceutical composition for the treatment or prevention of hypertension and hypercholesterolemia
Novaković et al. New drugs for the treatment of dyslipidemia
TW202313072A (en) Pediatric formulations of ferric citrate
US20120064161A1 (en) Modified release niacin pharmaceutical formulations
KR20110132171A (en) Combination sustained-release formulation containing nicain

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17825008

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17825008

Country of ref document: EP

Kind code of ref document: A1