WO2010008244A2 - Pharmaceutical preparation - Google Patents

Pharmaceutical preparation Download PDF

Info

Publication number
WO2010008244A2
WO2010008244A2 PCT/KR2009/003969 KR2009003969W WO2010008244A2 WO 2010008244 A2 WO2010008244 A2 WO 2010008244A2 KR 2009003969 W KR2009003969 W KR 2009003969W WO 2010008244 A2 WO2010008244 A2 WO 2010008244A2
Authority
WO
WIPO (PCT)
Prior art keywords
cellulose
release
pharmaceutical formulation
copolymer
mixtures
Prior art date
Application number
PCT/KR2009/003969
Other languages
French (fr)
Korean (ko)
Other versions
WO2010008244A3 (en
Inventor
김성욱
전성수
구자성
김병하
이나영
Original Assignee
한올제약주식회사
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 한올제약주식회사 filed Critical 한올제약주식회사
Publication of WO2010008244A2 publication Critical patent/WO2010008244A2/en
Publication of WO2010008244A3 publication Critical patent/WO2010008244A3/en

Links

Images

Classifications

    • 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
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0002Galenical forms characterised by the drug release technique; Application systems commanded by energy
    • A61K9/0004Osmotic delivery systems; Sustained release driven by osmosis, thermal energy or gas
    • 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

Definitions

  • the present invention relates to a pharmaceutical preparation containing an angiotensin-2 receptor antagonist and a beta adrenergic blocker as an active ingredient, each of which is released over time.
  • Hypertension is a condition that is caused by multiple causes. Therefore, it is difficult to determine in advance what results would result from the use of a single anticompressant [Journal of hypertension 1995: 9: S33-S36]. For this reason, a combination of anti-pressure agents has been increasing for more effective treatment.
  • US Patent Publication No. 2005-0004194 discloses a combination of angiotensin converting enzyme inhibitor and angiotensin-2 receptor antagonist for treating cardiovascular disease by reducing side effects and enhancing the effects on patients with myocardial infarction.
  • Korean Patent Laid-Open Publication No. 2004-0078140 discloses an antihypertensive complex preparation of valsartan and calcium channel blocker, which is an angiotensin-2 receptor antagonist.
  • WO 03/097099 discloses that simple combination therapy in which unit dosage forms of angiotensin-2 receptor blocker and beta receptor blocker are simultaneously administered is effective for the treatment of cardiovascular disease in patients after myocardial infarction.
  • the present inventors have completed the present invention as a result of research to develop a complex formulation for cardiovascular diseases such as hypertension.
  • the problem to be solved by the present invention is to show an equal anti-pressure action and prevent complications, in particular, it is possible to maintain the blood pressure evenly in the dangerous time period of complications, it is useful for patients with hypertension with complications, blood pressure control during sleep, etc. It is intended to provide an agent that reduces the side effects of interaction.
  • the present invention provides a pharmaceutical formulation comprising a prior release compartment comprising an angiotensin-2 receptor antagonist as a pharmacologically active ingredient, and a delayed release compartment comprising a beta adrenergic blocker as a pharmacologically active ingredient.
  • the angiotensin-2 receptor antagonist which is the active ingredient of the pre-release compartment, is prerelease, and preferably, at least 85% of the total amount of the angiotensin-2 receptor antagonist is released within 1 hour after the release of the angiotensin-2 receptor antagonist. Therefore, the desired effect can be generated quickly.
  • the beta adrenergic blocker of the delayed-release compartment in the formulations of the invention is released after a delay, ie 1 to 8 hours after release of the angiotensin-2 receptor antagonist, preferably 1 to 6 hours.
  • the release amount of the beta adrenergic blocker released within 2 hours after the release of the angiotensin-2 receptor antagonist is within 0-10% of the total amount of the beta adrenergic blocker. Can be.
  • the formulation of the present invention may comprise 0.1 to 350 parts by weight of the angiotensin-2 receptor antagonist based on 1 part by weight of the adrenergic blocker.
  • Pre-release compartment refers to the compartment in which the active ingredient is first released in comparison with the delayed-release compartment in the pharmaceutical formulation of the present invention, and may further include a pharmaceutically acceptable additive as necessary in addition to the pharmacologically active ingredient. .
  • the pharmacologically active component of the prior-release compartment is an angiotensin 2 receptor antagonist, and the angiotensin-2 receptor antagonist blocks angiotensin 2, one of the sources of vasoconstriction, with the AT1 receptor in the angiotensin receptor.
  • It is a drug that exerts a blood pressure-lowering effect on both myocardial systolic and diastolic phases, such as losartan, valsartan, telmisartan, irbesartan, candesartan, olmesartan, and / or eprosartan.
  • Isomers of and pharmaceutically acceptable salts thereof may be selected, and may include 1 to 800 mg of the formulation, and preferably 15 to 370 mg. The dose is based on a daily adult (65-75 kg adult male).
  • formulations of the present invention do not interfere with the release of pharmacologically active ingredients from commonly used additives such as pharmaceutically acceptable diluents, binders, disintegrants, lubricants, pH adjusters, dissolution aids and the like without departing from the effect of the present invention. It can be formulated using further within the range which does not.
  • the diluent may be starch, microcrystalline cellulose, lactose, glucose, mannitol, alginate, alkaline earth metal salts, clay, polyethylene glycol, dicalcium phosphate, or a mixture thereof.
  • the binder is starch, microcrystalline cellulose, highly dispersible silica, mannitol, sucrose, lactose, polyethylene glycol, polyvinylpyrrolidone, hydroxypropylmethylcellulose, hydroxypropylcellulose, natural gum, synthetic gum, polyvinylpyrrolidone Copolymers, povidone, gelatin, mixtures thereof, and the like.
  • the disintegrant may be a starch or modified starch such as sodium starch glycolate, corn starch, potato starch or starch gelatinized starch (starch starch); Clay such as bentonite, montmorillonite, or veegum; Celluloses such as microcrystalline cellulose, hydroxypropyl cellulose or carboxymethyl cellulose; Algins such as sodium alginate or alginic acid; Crosslinked celluloses such as croscarmellose sodium; Gums such as guar gum and xanthan gum; Crosslinked polymers such as crosslinked polyvinylpyrrolidone (crospovidone); Effervescent agents such as sodium bicarbonate, citric acid, or mixtures thereof can be used.
  • starch starch such as sodium starch glycolate, corn starch, potato starch or starch gelatinized starch (starch starch); Clay such as bentonite, montmorillonite, or veegum; Celluloses such as microcrystalline cellulose, hydroxypropyl
  • the lubricant is talc, stearic acid, magnesium stearate, calcium stearate, sodium lauryl sulfate, hydrogenated vegetable oil, sodium benzoate, sodium stearyl fumarate, glyceryl behenate, glyceryl monorate, glyceryl monostearate, Glyceryl palmitostearate, polyethylene glycol, and / or mixtures thereof, and the like.
  • the stabilizer may be ascorbic acid, citric acid, butylated hydroxy anisole, butylated hydroxy toluene, and / or tocopherol derivatives.
  • an alkalizing agent which is a salt of an alkali metal, a salt of an alkaline earth metal, or a mixture thereof, may be used.
  • calcium carbonate, sodium carbonate, sodium hydrogen carbonate, magnesium oxide, magnesium carbonate, and / or sodium citrate Etc. can be used.
  • the pH adjusting agent may be an acidifying agent such as acetic acid, adipic acid, ascorbic acid, malic acid, succinic acid, tartaric acid, fumaric acid, citric acid, and a basicizing agent such as precipitated calcium carbonate, aqueous ammonia, and meglumine.
  • an acidifying agent such as acetic acid, adipic acid, ascorbic acid, malic acid, succinic acid, tartaric acid, fumaric acid, citric acid, and a basicizing agent such as precipitated calcium carbonate, aqueous ammonia, and meglumine.
  • the dissolution aid may be used polyoxyethylene sorbitan fatty acid esters such as sodium lauryl sulfate, polysorbate, sodium docusate and the like.
  • a pharmaceutically acceptable additive may be selected and used in the preparation of the present invention as various additives selected from colorants and fragrances.
  • the range of additives usable in the present invention is not limited to the use of such additives, and the above additives may be formulated to contain a range of dosages, usually by selection.
  • the delayed-release compartment refers to a compartment in which the active ingredient is released after a certain time of release of the prior-release compartment active ingredient.
  • the delayed-release compartment comprises (1) a pharmacologically active ingredient and (2-1) release controlling substance or (2-2) an osmotic pressure regulator and a semipermeable membrane coating base, and (3) a pharmaceutically acceptable It may further include an additive.
  • the pharmacologically active component of the delayed-release compartment is beta adrenergic antagonist, and the beta adrenergic blocker blocks the excitability of the sympathetic nervous system due to the stimulation of the adrenergic beta receptors in the cardiovascular and smooth muscle, resulting in hypertension
  • beta adrenergic blocker is alprenolol, acebutorol, amosulolol, arotinol, atenolol, befunolol, betaxolol, bevan Tolol, Bisoprolol, Boffindolol, Bucumolol, Buffololol, Bufuralol, Bunitrolol, Bufrandolol, Butopylolol, Carazolol, Caterolol, Carvedilol, Celylprolol, Cetamolol, Chlor
  • Nebivolol is a fourth-generation beta adrenergic blocker that not only has all the benefits of first-, second- and third-generation beta-adrenergic blockers, but also produces NO, which dilates peripheral blood vessels. It has no side effects such as increase, worsening of diabetes mellitus, cold limbs, and effects on right heart dilatation heart failure. It is particularly suitable for elderly systolic hypertensives, those with heart failure, elderly diabetic hypertension, elderly hyperlipidemic hypertension, and hypertension combined with heart failure.
  • nebivolol can obtain an effective therapeutic effect by combining angiotensin-2 receptor antagonist with hypertension of severe diseases such as heart failure and myocardial infarction.
  • the delayed-release compartment in the pharmaceutical formulation of the present invention comprises a release controlling substance, wherein the release controlling substance of the present formulation is selected from, for example, enteric polymers, water insoluble polymers, hydrophobic compounds, hydrophilic polymers, and mixtures thereof. At least one, preferably at least one selected from water-insoluble polymers and enteric polymers.
  • the release controlling substance of the formulation is 0.1 to 100 parts by weight, preferably 1 to 50 parts by weight, with respect to 1 part by weight of the beta adrenergic blocker, and when it is less than 0.1 parts by weight, it is difficult to have a sufficient delay time. Drug release may be excessively delayed, making it difficult to obtain significant clinical effects.
  • the enteric polymer is insoluble or stable under acidic conditions, and refers to a polymer that is dissolved or decomposed under specific pH conditions of pH 5 or more.
  • the enteric polymer usable in the present invention is at least one selected from the group consisting of an enteric cellulose derivative, an enteric acrylic acid copolymer, an enteric polymethacrylate copolymer, an enteric maleic acid copolymer, an enteric polyvinyl derivative, and a mixture thereof. .
  • the enteric cellulose derivative may be hydroxypropylmethylcellulose acetate succinate (or hypromellose acetate succinate), hydroxypropylmethyl cellulose phthalate (or hypromellose phthalate), hydroxymethylethyl cellulose phthalate , Cellulose acetate phthalate, cellulose acetate succinate, cellulose acetate maleate, cellulose benzoate phthalate, cellulose propionate phthalate, methyl cellulose phthalate, carboxymethylethyl cellulose, ethyl hydroxyethyl cellulose phthalate, methyl hydroxyethyl cellulose and their At least one selected from a mixture;
  • the enteric acrylic acid copolymers include styrene-acrylic acid copolymers, methyl acrylate-acrylic acid copolymers, methyl methacrylate acrylates, butyl acrylate-styrene-acrylic acid copolymers, methyl acrylate-methacrylic acid-o
  • the enteric maleic acid copolymer is vinyl acetate-maleic anhydride copolymer, styrene-maleic anhydride copolymer, styrene-maleic acid monoester copolymer, vinyl methyl ether-maleic anhydride copolymer, ethylene-maleic anhydride copolymer, vinyl butyl ether- At least one selected from maleic anhydride copolymer, acrylonitrile-methyl acrylate maleic anhydride copolymer, butyl styrene-maleic-maleic anhydride copolymer and mixtures thereof;
  • the enteric polyvinyl derivative is preferably at least one selected from polyvinyl alcohol phthalate, polyvin
  • the enteric polymer may be included in an amount of 0.1 parts by weight to 20 parts by weight, preferably 0.5 parts by weight to 10 parts by weight with respect to 1 part by weight of the active ingredient, and less than 0.1 parts by weight may be easily dissolved at a pH of less than 5, If the amount exceeds 20 parts by weight, the total weight of the preparation may be unnecessarily increased or the elution may be excessively delayed.
  • the water insoluble polymer refers to a polymer that is not soluble in pharmaceutically acceptable water that controls the release of the drug.
  • the water-insoluble polymers usable in the present invention are polyvinyl acetate, water-insoluble polymethacrylate copolymers (e.g. poly (ethylacrylate, methyl methacrylate) copolymers (eg Eudragit NE30D), poly (ethylacrylic) Latex, methyl methacrylate, trimethylaminoethyl methacrylate chloride) copolymer (e.g.
  • Eudragit RS, RL), etc. ⁇ ethyl cellulose, cellulose ester, cellulose ether, cellulose acylate, cellulose dicylate, cellulose tri At least one selected from the group consisting of acylate, cellulose acetate, cellulose diacetate, cellulose triacetate and mixtures thereof is preferred, and more preferably polyvinylacetate, ethyl cellulose, poly (ethyl acrylate, methyl methacrylate).
  • Trimethylaminoethyl methacrylate Bit chloride is at least one selected from a copolymer, and cellulose acetate.
  • the water-insoluble polymer may be included in an amount of 0.1 parts by weight to 30 parts by weight, preferably 0.5 parts by weight to 20 parts by weight, and less than 0.1 parts by weight based on 1 part by weight of the active ingredient. If the content is more than 30 parts by weight, the dissolution may be excessively delayed.
  • the hydrophobic compound refers to a substance that does not dissolve in pharmaceutically acceptable water that controls the release of the drug.
  • the hydrophobic compound usable in the present invention is, for example, at least one selected from the group consisting of fatty acids and fatty acid esters, fatty alcohols, waxes, inorganic substances, and mixtures thereof.
  • fatty acids and fatty acid esters are selected from glyceryl palmitostearate, glyceryl stearate, glyceryl distearate, glyceryl bihenate, cetyl palmitate, glyceryl monooleate, stearic acid and mixtures thereof Species or more;
  • Fatty acid alcohols are at least one selected from cetostearyl alcohol, cetyl alcohol, stearyl alcohol and mixtures thereof;
  • the waxes are at least one selected from carnauba wax, beeswax, microcrystalline wax, and mixtures thereof;
  • the inorganic material is preferably at least one selected from talc, precipitated calcium carbonate, calcium dihydrogen phosphate, zinc oxide, titanium oxide, kaolin, bentonite, montmorillonite, bum and mixtures thereof.
  • a more preferred example of the hydrophobic compound is carnauba wax.
  • the hydrophobic compound may be included in an amount of 0.1 parts by weight to 20 parts by weight, preferably 0.5 parts by weight to 10 parts by weight with respect to 1 part by weight of the active ingredient, and when less than 0.1 parts by weight, the release of the drug may not be controlled. In case of more than 20 parts by weight, excessive elution may be delayed.
  • the hydrophilic polymer refers to a polymeric material that is dissolved in pharmaceutically acceptable water that controls the release of the drug.
  • hydrophilic polymers usable in the present invention include 1 selected from the group consisting of sugars, cellulose derivatives, gums, proteins, polyvinyl derivatives, hydrophilic polymethacrylate copolymers, polyethylene derivatives, carboxyvinyl copolymers, and mixtures thereof. More than species.
  • sugars are dextrins, polydextrins, dextran, pectin and pectin derivatives, alginates, polygalacturonic acids, xylans, arabinoxylans, arabinogalactan, starch, hydroxypropylstarches, amylose, amylopectin, and their At least one selected from mixtures;
  • Cellulose derivatives include hydroxypropylmethylcellulose (or hypromellose), hydroxypropylcellulose, hydroxymethylcellulose, hydroxyethylcellulose, methylcellulose, carboxymethylcellulose sodium, hydroxypropyl methylcellulose acetate succinate, At least one selected from hydroxyethyl methyl cellulose, and mixtures thereof;
  • the gum is at least one selected from guar gum, locust bean gum, tragacanta, carrageenan, acacia gum, arabic gum, gellan gum, xanthan gum, and mixtures thereof;
  • Proteins may be at least one selected from gelatin,
  • hydrophilic polymer are selected from polyvinyl pyrrolidone, hydroxypropyl cellulose, hypromellose, and poly (ethyl acrylate-methyl methacrylate-triethylaminoethyl- methacrylate chloride) copolymer. More than species.
  • the hydrophilic polymer may be included in an amount of 0.05 parts by weight to 30 parts by weight, preferably 0.5 to 20 parts by weight with respect to 1 part by weight of the active ingredient, and when it is less than 0.05 parts by weight, the release rate may not be controlled. If the excess is excessive, there is a fear that excessive dissolution is delayed.
  • the delayed-release compartment of the present invention includes an osmotic pressure regulator and may be a compartment coated with a semipermeable membrane coating base.
  • the osmotic pressure regulator is preferably at least one selected from the group consisting of magnesium sulfate, magnesium chloride, sodium chloride, lithium chloride, potassium sulfate, lithium sulfate, sodium sulfate and mixtures thereof.
  • the osmotic pressure control agent may be included in an amount of 0.01 parts by weight to 10 parts by weight, preferably 0.05 parts by weight to 0.5 parts by weight with respect to 1 part by weight of the active ingredient, and there is a concern that it is difficult to obtain sufficient time difference release property at less than 0.01 parts by weight, Drug release may be delayed at more than 10 parts by weight, making it difficult to obtain a significant clinical effect.
  • the semi-permeable membrane coating base is a substance to be blended into the coating layer of the pharmaceutical formulation, and refers to a substance used to form a membrane that allows some components to pass but does not pass other components.
  • the semipermeable membrane coating base may use the above-mentioned water-insoluble polymer.
  • the semipermeable membrane coating base in the present invention is, for example, polyvinyl acetate, polymethacrylate copolymer, poly (ethylacrylate, methyl methacrylate) copolymer, poly (ethylacrylate, methyl methacrylate, trimethylaminoethylmethacrylate Late chloride) copolymer, ethyl cellulose, cellulose ester, cellulose ether, cellulose acylate, cellulose dicylate, cellulose triacylate, cellulose acetate, cellulose diacetate, cellulose triacetate and mixtures thereof The above is mentioned.
  • the semi-permeable membrane coating base may be included in an amount of 0.01 parts by weight to 10 parts by weight, preferably 0.05 parts by weight to 1.25 parts by weight, with respect to 1 part by weight of the active ingredient, and when it is less than 0.01 parts by weight, it may be difficult to have a sufficient delay time. And, if more than 10 parts by weight there is a problem that the release of the drug does not occur or the delay time is long.
  • the formulations of the present invention are diluents, binders, and borates other than those mentioned as pharmaceutically acceptable (2-1) release controlling substances and (2-2) osmotic pressure regulators and semipermeable membrane coating agents within the scope of not impairing the effects of the present invention.
  • Commonly used additives such as release, lubricants, pH adjusters, antifoams, dissolution aids and the like can be formulated further using within a range not departing from the nature of delayed release.
  • the pharmaceutical preparations of the present invention can be prepared in a variety of formulations and can be formulated, for example, in tablets, powders, granules, capsules, and the like, such as uncoated tablets, coated tablets, multilayer tablets, or nucleated tablets.
  • the formulation of the present invention is a tabletting by selectively mixing additives such as granules constituting the pre-release compartment and granules constituting the delayed-release compartment and the like to have a pre-release compartment and a delayed-release compartment in a single tablet, and thus the active ingredient of each compartment.
  • additives such as granules constituting the pre-release compartment and granules constituting the delayed-release compartment and the like to have a pre-release compartment and a delayed-release compartment in a single tablet, and thus the active ingredient of each compartment.
  • This may be in the form of uncoated tablets will be eluted separately to show the respective effects.
  • the formulation of the present invention may be in the form of a two-phase matrix tablet obtained by tableting after the delayed-release compartment and the prior-release compartment are uniformly mixed.
  • the pharmaceutical formulation of the present invention may be in the form of a film coated tablet consisting of a tablet consisting of a delayed-release compartment and a film coating layer consisting of a pre-release compartment surrounding the outside of the tablet, the film coating layer of the film coating layer as it is dissolved
  • the active ingredient is eluted first.
  • the pharmaceutical formulation of the present invention is a delayed-release compartment, obtained by mixing the pharmaceutical additives in the granules constituting the delayed-release compartment and the prior-release compartment, and tableting in a double or triple tablet using a multiple tableting machine and
  • the pre-emitting compartments may be in the form of a multi-layered tablet with a layered structure.
  • This formulation is a tablet for oral administration which is formulated to enable pre-release and delayed release in layers.
  • the pharmaceutical formulation of the present invention may be in the form of a nucleus tablet consisting of an inner layer consisting of a delayed-release compartment and an outer layer consisting of a prior-release compartment surrounding the outer surface of the inner core tablet.
  • the nucleated tablet may be an osmotic nucleated tablet, and the osmotic nucleated tablet contains an osmotic pressure-controlling agent inside the tablet for delayed release, followed by compression, and then coated the surface of the tablet with a semipermeable membrane coating agent to form an inner core.
  • compositions of the present invention may be in the form of capsules comprising particles, granules, pellets, or tablets consisting of delayed-release compartments, and particles, granules, pellets, or tablets consisting of prior release compartments.
  • the tablet consisting of the delayed-release compartment of the capsule may include an osmotic pressure-controlling agent within the tablet and an osmotic coated tablet having a semipermeable membrane coating base on the surface of the tablet.
  • the base of the capsule may be one selected from gelatin, succinate gelatin, or hydroxypropylmethylcellulose, or a mixture thereof.
  • the pharmaceutical formulation of the present invention may be in the form of a kit comprising a delayed-release compartment, and a prior-release compartment
  • the kit comprises (a) a prior-release compartment (b) a delayed-release compartment and (c) the It may consist of a container for filling the pre-release compartment and the delayed-release compartment.
  • the kit prepares the particles, granules, pellets, or tablets constituting the prerelease compartment, and separately prepares the granules, pellets, or tablets constituting the delayed release compartment, and fills them together with foil, blisters, bottles, and the like. It can be prepared in a form that can be taken at the same time.
  • the formulations of the present invention may further form a coating layer on the exterior of the delayed release compartment and / or the prior release compartment.
  • the surface of the particles, granules, pellets, or tablets consisting of delayed-release compartments and / or pre-release compartments may be coated for the purpose of release control or formulation stability.
  • the formulation according to the present invention is also provided in a state such as uncoated tablets without additional coating, but may be in the form of a coated tablet containing a coating layer further by forming a coating layer on the outside of the formulation, if necessary. .
  • a coating layer By forming the coating layer, it is possible to further ensure the stability of the active ingredient.
  • the method of forming the coating layer may be appropriately selected by a person skilled in the art from the method of forming a film-like coating layer on the surface of the tablet layer, a method such as a fluidized bed coating method, a fan coating method may be applied, and preferably Fan coating can be applied.
  • the coating layer may be formed by using a coating agent, a coating aid, or a mixture thereof.
  • the coating agent may be a cellulose derivative such as hydroxypropylmethylcellulose, hydroxypropylcellulose, sugar derivatives, polyvinyl derivatives, waxes, or fatty acids. , Gelatin, or a mixture thereof, and the like, and a coating aid may be polyethylene glycol, ethyl cellulose, glycerides, titanium oxide, talc, diethyl phthalate, triethyl citrate or a mixture thereof.
  • the coating layer may be included in the range of 0.5 to 15% by weight (% w / w) based on the total weight of the tablet.
  • the formulation of the present invention can be administered once a day between 5 to 11 o'clock in the evening, and can exert anti-pressure action and complication prevention evenly for 24 hours.
  • Beta adrenergic blockers are known to have strong antitumor activity after dawn [N EnglJ Med 2003; 348; 2377-8, Chronology International 2006; 23 (4): 813-829], angiotensin-2 receptor antagonists are known to have strong blood pressure lowering effects from midnight to dawn [J. Hypertens, 2005; 23: 1913-1922, Hypertension, 2003; 42: 283-290, Chronobiol. Int. 2005; 22: 755-776], even when the preparation of the present invention is taken once in the evening, the blood pressure is effectively lowered until dawn by an angiotensin-2 receptor antagonist, which is released, and the blood pressure after dawn by a beta-adrenergic blocker delayed release. It effectively lowers the 24 hours even anti-pressure action and prevent complications.
  • the preparation of the present invention By administering the preparation of the present invention in the evening time, blood pressure can be maintained evenly in the time of risk of complications (from dawn to morning time). Therefore, the sympathetic hyper-excited state of the heart should be suppressed evenly for 24 hours. It is useful for patients with high blood pressure with complications.
  • the formulation of the present invention is useful for suppressing non-dipper type hypertension, a type of hypertension in which blood pressure does not decrease during sleep.
  • the non-dipper type hypertension is present in the elderly, diabetics, cardiac hypertrophy, and high blood pressure with a high risk of complications such as stroke, and is useful for suppressing blood pressure during sleep by taking the preparation of the present invention in the evening.
  • the agent of the present invention is an angiotensin-2 receptor antagonist
  • the agent of the present invention is an angiotensin-2 receptor antagonist
  • is first introduced into the liver is activated by the cytochrome P450 enzyme in the liver, after the beta adrenergic blocker passes through the liver between the beta adrenergic blocker and the angiotensin-2 receptor antagonist Blocks metabolic interactions that may occur, reducing side effects of drug interactions.
  • the present invention also provides a method for treating cardiovascular disease, comprising administering the agent of the present invention to a mammal including a human.
  • cardiovascular diseases can be treated.
  • the cardiovascular disease is meant to include hypertension, or high blood pressure complications.
  • compositions of the present invention may be prepared by any suitable method in the art, for example, with reference to methods disclosed in Chronotherapeutics (2003, Peter Redfern, PhP), Remington's Pharmaceutical Science (Recent Edition), Mack Publishing Company, Easton PA, and the like. It can be formulated preferably according to a disease or a component, and can be manufactured by the method containing the following steps specifically ,.
  • the active ingredient of the delayed-release compartment is mixed with, or combined with, one or two release controlling substances selected from an enteric polymer, a water insoluble polymer, a hydrophobic compound, and a hydrophilic polymer, and a conventional additive used in pharmaceuticals.
  • Delayed-release granules or tablets are obtained through drying, granulation or coating, and tableting, or the active ingredient is semipermeable after mixing, associating, drying, granulating or tableting by administering an osmotic agent and a conventional additive which is used pharmaceutically. It is a step of obtaining delayed-release granules or tablets by coating with a membrane coating base.
  • the second step involves the administration of the active ingredient of the prior-release compartment and the conventionally acceptable pharmaceutically acceptable additives to produce the oral solids through mixing, coalescing, drying, granulating or coating, and tableting to produce oral solids. Obtaining extruded granules or tablets.
  • the granules or tablets obtained in the first step and the second step are mixed with pharmaceutical excipients, tableted or filled to obtain a preparation for oral administration.
  • the first step and the second step may be reversed or executed simultaneously.
  • the pharmaceutical formulation of the present invention may be prepared by the above process, and the formulation method of the third step is described in more detail as follows, but is not limited thereto.
  • the particles or granules obtained in the first step are further coated as they are or with a release controlling material, and then mixed with the granules prepared in the second step and compressed into a certain amount of weight to prepare a tablet.
  • the obtained tablet can be film coated as necessary for the purpose of improving stability or property.
  • the coated tablets or granules obtained in the first step are additionally coated as they are or with a release control material, dried, and then compressed into a predetermined amount to prepare tablets as they are or additionally coated, and then the active ingredients of the pre-release compartments are separately coated with a water-soluble film coating solution.
  • the tablet outer layer obtained in step 1 can be used to prepare an orally administered film coating tablet containing the active ingredient in a film coating.
  • the granules obtained in the first step as they are or are additionally coated and dried with a release controlling substance and the granules obtained in the second step can be prepared in double tablets using a tablet press.
  • Coated multi-layered tablets can be prepared by formulating or coating triple or more multi-layered tablets by adding a release aid layer as needed, or by formulation.
  • the coated tablet or granules obtained in the first step are additionally coated as it is or with a release control material, dried, and then compressed into a predetermined amount to be coated as it is or additionally to the inner core, followed by a nucleated tableting machine together with the granules obtained in the second step.
  • the coated nucleated tablet may be prepared by preparing or coating a nucleated tablet having a form in which a pre-release layer is enclosed on the surface of the inner core.
  • the granules obtained in the first step are additionally coated as is or with a release controlling substance, and the dried granules or tablets and the granules or tablets obtained in the second step are placed in a capsule charger and filled into capsules of a predetermined size by an effective amount of each active ingredient in an appropriate amount.
  • Capsules may be prepared by mixing the release control pellets containing the ingredients and filling the capsules with a capsule filling machine.
  • the delayed-release compartment preparation (beta-adrenergic blocker-containing formulation) obtained in the first stage and the prior-release compartment preparation (angiotensin-2 receptor antagonist-containing formulation) obtained in the second stage are filled together with a foil, blister, bottle, etc. It can be made into a kit that can be taken at the same time.
  • the human dosage of the formulation of the present invention is appropriately selected depending on the absorption rate, inactivation rate and excretion rate of the active ingredient in the body, the age, sex and condition of the patient, but in general, the beta adrenergic blocker and angiotensin-2 receptor antagonist in adults In a total amount, 2.0 to 2200 mg per day, preferably 42 to 1900 mg per day can be exerted anti-pressure action and prevent complications.
  • the formulation of the present invention is formulated to be compatible with the theory of drug metabolic enzyme interactions and the principle of time difference administration by allowing time difference dissolution between two components of angiotensin-2 receptor antagonist and beta adrenergic blocker. It can increase the efficacy and reduce the side effects compared with the simultaneous release.
  • the formulation of the present invention applies the principle of chronotherapy and the principle of Xenobiotics of the drug to the expression of pharmacological action of each of the complex components to control release in the body at a specific rate to achieve the most ideal effect upon absorption in the body.
  • the pharmaceutical preparation of the present invention exhibits an equal anti-pressure action and prevents complications, and is particularly useful for hypertension patients with complications, suppressing blood pressure during sleep, and the like. Reduces the side effects of action.
  • Example 1 is a graph showing the dissolution rate of valsartan and nebivolol in the valsartan-hydrochloride nebivolol-containing formulation of Example 2 compared with the reference drug.
  • FIG. 3 is a graph showing the dissolution rate of losartan and nebivolol hydrochloride in the losartan-hydrochloride nebivolol-containing preparation of Example 14 compared with the reference drug.
  • 5 is a graph showing the dissolution rate of losartan in the formulations of Examples 21, 22, 23, and 24 compared with the reference drug.
  • Example 6 is a graph showing the dissolution rate of candesartan cilexetil and atenolol in the candesartan cilexetil-atenolol-containing formulation of Example 34 in comparison with the reference drug.
  • Example 7 is a graph showing the dissolution rate of eprosartan and carvedilol in the eprosartan-carvedilol-containing formulation of Example 36 in comparison with the reference drug.
  • Example 8 is a graph showing the dissolution rate of telmisartan and carvedilol in the telmisartan-carvedilol-containing formulation of Example 37 compared with the reference drug.
  • Example 9 is a graph showing the dissolution rate of losartan and metoprolol in the losartan-metoprolol-containing formulation of Example 39, compared with the reference drug.
  • Valsartan (Ranbaxy, India), lactose (Parmatose, DMV Pharma, Netherlands), microcrystalline cellulose (AvicelPH, FMC Biopolymer, USA), poloxamer 188 (Lutrol-F68, BASF, Germany), starch Sodium glyconate (Primojel, DMV Pharma, The Netherlands) was weighed and inconvenienced as No. 35. Separately, hydroxypropyl cellulose (HPC-L, Nippon soda, Japan) was dissolved in purified water to prepare a binding solution. The mixture was put into a fluid bed granulator GPCG-1 (Glatt, Germany) and granulated by the addition of a binder solution. The fluid bed dryer assembly was dried after assembly was complete.
  • the dried product was sized using an F-type sizer (KYK-60, Korea Medi, Korea) equipped with a No. 20 body, thereby preparing Valsartan pre-release granules.
  • hydrochloric acid nebivolol (Cadila, India), microcrystalline cellulose, sodium lauryl sulfate (Texapon K12P, Cognis Corp, USA), crosslinked polyvinylpyrrolidone (Crospovidone, BASF, Germany) No. 35
  • the apples were sieved, and hydroxypropyl cellulose (HPC-L, Nippon soda, Japan) was separately dissolved in purified water to obtain a binding solution.
  • Fluidized bed granulators and fluidized bed drying are the same as those of valsartan pre-release granules.
  • the dried material was placed in a fluidized bed coater and separately cellulose acetate (acetal group 32%) (Eastman Chemical Company, USA), cellulose acetate (acetal group 39.8%) (Eastman Chemical Company, USA), and hydroxypropylmethylcellulose in ethanol and methylene chloride The melted solution was prepared to coat the granulated product.
  • the product temperature is maintained at 34 ⁇ 38 °C, when the coating is complete, while drying and surface work for about 1 hour while maintaining the product temperature 40 °C, to produce a nebivolol delayed-release granules hydrochloric acid.
  • magnesium stearate was added thereto, and then mixed for 4 minutes and compressed into tablets using a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 10.0 mm diameter punch. .
  • Tablets that have been tableted are prepared by dissolving and dispersing hydroxypropylmethylcellulose 2910 (Shin-etsu, Japan), polyethylene glycol 6000 (BASF, Germany), and titanium oxide (Tioside Americas, USA) in ethanol and purified water. By coating.
  • magnesium stearate was added to the delayed-release layer granules of (2) nebivolol hydrochloride of Example 1, and finally mixed to prepare a delayed-release layer of nebivolol hydrochloride.
  • magnesium stearate was added to the (1) valsartan pre-release granules of Example 1, and finally mixed to prepare a valsartan pre-release layer.
  • the prepared Navivolol delayed-release layer and valsartan pre-release layer were put into different granule inlet holes of a rotary multi-layer tablet tablet press (MRC-37T: Sejong Pharmatech, Korea) equipped with a 10 mm diameter puncher, and then tableted. Hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were coated using a coating solution prepared by dissolving and dispersing in ethanol and purified water.
  • the tablets were put into different granules, and tableted, the tablets were tableted with hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide in a coating solution prepared by dissolving and dispersing ethanol in purified water.
  • Example 1 (2) After the magnesium stearate was added to the granules of delayed-release layer of nebivolol hydrochloride prepared by the method of Example 1 (2), the final mixture was rotary with a 6 mm diameter punch.
  • MRC-33 Sejong Pharmatech, Korea
  • Magnesium stearate was added to the mixture, and the mixture was compressed into tablets.
  • the tablets having been tableted are coated with a coating solution prepared by dissolving and dispersing hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide in ethanol and purified water to prepare a coated tablet.
  • Valsartan pre-release granules were prepared according to the method of Example 2, except that the components and contents shown in Table 1 do not include lactose.
  • the nebivolol delayed-release layer of Example 2 above It prepared by the same method as the preparation.
  • the valsartan pre-release granules were compressed in a rotary tablet press equipped with a 6 mm diameter punch, and the hydrochloride nebivolol delayed-release granules were compressed in a rotary tablet press equipped with a 5 mm diameter punch (MRC-33: Sejong Pharmatech, Korea). Tableting was completed two tablets were filled capsule No. 1 capsule (SF-40N, Sejong Pharmatech, Korea) into No. 1 capsule (HPMC hard capsule, Seoheung capsule, Korea) to prepare a capsule.
  • the nebivolol delayed-release tablet hydrochloride was prepared according to the preparation method of delayed-release tablet hydrochloride in Example 5, and the valsartan pre-release granules were prepared according to the preparation method of Example 2 (1). After the preparation, each tablet and granules were filled in the same capsule (No. 0 capsule) with a capsule charger to prepare a nebivolol valsartan hydrochloride (tablet + granules).
  • the capsules were prepared by filling the No. 0 capsules with Navivolol delayed-release granules and valsartan granules prepared in the same manner as in Example 2 with a capsule charger.
  • sugar seeds were added to a fluidized bed granulator (GPCG 1: Glatt), and then poloxamer and hydroxypropyl cellulose (HPC-L, Nippon) were mixed separately with purified water and ethanol. soda, Japan) and valsartan were sprayed to form a pellet containing valsartan by spraying a binder solution in which valsartan was dissolved or suspended to prepare valsartan pre-release pellets.
  • GPCG 1 Glatt
  • HPC-L hydroxypropyl cellulose
  • sugar seeds were added to a fluidized bed granulator (GPCG 1: Glatt), and then hydroxypropylmethylcellulose, sodium lauryl sulfate, and nebivolol hydrochloric acid were separately mixed with purified water and ethanol.
  • the binder solution dissolved or suspended was sprayed to form a nebivolol-containing pellet, and dried.
  • the granules were sprayed with phthalic hydroxypropylmethylcellulose in a mixture of ethanol and methylene chloride to prepare nebivolol delayed-release pellets.
  • Capsules were prepared by mixing two pellets prepared in the above (1) and (2) and filling the capsule No. 0 with a capsule charger.
  • valsartan pre-release pellets were prepared in the same manner as in Example (1), and the nebivolol delayed-release tablet of hydrochloric acid was prepared by the method of Example 2 (2).
  • Magnesium stearate was added to the delayed-release layer granules of the ball roll, and the final mixture was compressed into tablets and pellets by tableting with a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 6 mm diameter punch.
  • the capsules were filled in capsule No. 0 with a capsule charger to prepare a capsule.
  • valsartan, lactose, poloxamer, microcrystalline cellulose, sodium starch glycolate were weighed and appled into No. 35 sieve and mixed for 20 minutes in a double cone mixer to prepare a mixture.
  • hydroxypropyl methyl cellulose was dissolved in 80% ethanol, and acrylic acid (Acryl-eze, Colorcon, USA) was dispersed in purified water to prepare a coating solution. After the preparation of the coating solution was completed, the granules were administered to the fluidized bed coater and subjected to the first coating (hydroxypropylmethylcellulose coating solution), followed by a second coating (acrylic coating solution).
  • magnesium stearate was added thereto, mixed for 4 minutes, and compressed using a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 12.0 mm diameter punch. Tablet-finished tablets were coated with a coating solution in which hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were dissolved and dispersed in ethanol and purified water.
  • valsartan lactose, poloxamer 188, microcrystalline cellulose, sodium starch glycolate, starch gelatinized starch (Starch 1500G, Colorcon, USA) and weigh apples in No. 35 with the ingredients and contents shown in Table 2.
  • the mixture was prepared by mixing for 20 minutes at. After completion of mixing, magnesium stearate was added and mixed for 4 minutes to prepare valsartan pre-release granules.
  • nebivolol hydrochloride, microcrystalline cellulose, sodium lauryl sulfate was appled with No. 35 sieve, and mixed for 5 minutes with a double cone mixer to prepare a mixture.
  • hydroxypropyl cellulose was dissolved in purified water to obtain a binding solution.
  • the above mixture was administered to a fluidized bed granulator (GPCG-1, Glatt, Germany), followed by spraying the binding solution to prepare granules.
  • the granules were dried in a fluidized bed dryer (GPCC-1, Glatt, Germany), and then carbomer 71G (carboxyvinyl polymer, lubrizol) was added to the granules in a powder state and mixed for 10 minutes. I passed. After sieving, magnesium stearate was added and mixed for 4 minutes to prepare nebivolol delayed-release granules.
  • GPCC-1 fluidized bed dryer
  • Valsartan pre-release granules are compressed into a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 6 mm diameter punch.Navivolol delayed-release granules hydrochloric acid rotary tablets (RC-30: Sejong Pharmatech, equipped with a 5 mm diameter punch) Korea). Tablet-finished tablets were coated with a coating solution in which hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were dissolved and dispersed in ethanol and purified water. Each coated tablet was packaged in a single press through pack (PTP) packaging container with a packaging machine (WiderVII, Bucheon machine, Korea) to prepare a packaging kit for simultaneous use.
  • PTP press through pack
  • nebivolol hydrochloride, microcrystalline cellulose and sodium lauryl sulfate were appled in a No. 35 sieve, mixed in a double cone mixer, and then poured into a fluidized bed granulator (GPCG 1: Glatt), and separately hydroxypropyl cellulose. After spraying the binder solution dissolved in water to form granules, drying was completed. Carbomer 71G was added to the granules in a powder state and mixed for 10 minutes. Magnesium stearate was added thereto and mixed with a final double cone mixer, and the final mixture was rotary tablet press (MRC-33: Sejong Pharmatech, Korea). Uncoated tablet was prepared by tableting with a 7 mm diameter punch.
  • the valsartan coating solution was prepared by dissolving valsartan, colloidal silicon oxide, poloxamer, and hydroxypropylmethylcellulose in a mixture of ethanol and methylene chloride with the components and contents shown in Table 2.
  • Hydrochloric acid nebivolol uncoated tablets were administered to a high coater (SFC-30F Sejong Machinery, Korea) and coated with valsartan coating solution. After the drug coating was completed, hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were coated using a coating solution dissolved and dispersed in ethanol and purified water.
  • Valsartan pre-release granules were prepared in the same manner as in Example 11 (1), with the components and contents shown in Table 2.
  • nucleated tablet tableting machine Kilian
  • hydrochloric acid nebivolol osmotic nuclear tablets and valsartan pre-release granules as outer layers, tablets were made with a 12 mm diameter punch and then a high coater ) To form a film coating layer to prepare a nucleated tablet. Tablets having been tableted were coated with a coating solution in which hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were dissolved and dispersed in ethanol and purified water.
  • Example 2 (2) Prepared in the same manner as in Example 2 (2), except that Eudragit RL was used instead of cellulose acetate (acetal group 32%) and cellulose acetate (39.8%) with the ingredients and contents shown in Table 3. It was.
  • apple nebivolol hydrochloride, microcrystalline cellulose and sodium lauryl sulfate were appled in a No. 35 sieve, mixed in a double cone mixer, and then put into a fluidized bed granulator (GPCG 1: Glatt), and separately hydroxypropyl cellulose.
  • GPCG 1 Glatt
  • Carbomer 71G was added to the granules in a powder state and mixed for 10 minutes.
  • Magnesium stearate was added thereto and mixed in a final double cone mixer, and the final mixture was mixed with a rotary tablet press (MRC-33: Sejong Pharmatech).
  • the uncoated tablet thus prepared was coated with a coating solution in which phthalic acid hydroxypropylmethylcellulose (Shin-etsu, Japan) was dissolved in a mixed solvent of methylene chloride and ethanol to prepare a core tablet.
  • phthalic acid hydroxypropylmethylcellulose Shin-etsu, Japan
  • telmisartan (Ranbaxy, India) was used instead of mbvalsartan.
  • Example 5 The ingredients and contents shown in Table 5 were prepared according to the method of Example 1 (2), except that Eudragit RS was used instead of cellulose acetate (acetal group 32%) and cellulose acetate (39.8% acetal group). .
  • sugar seeds were added to a fluidized bed granulator (GPCG 1: Glatt), and then poloxamer, sodium hydroxide, hydroxypropylmethylcellulose and candee were mixed with purified water and ethanol.
  • GPCG 1 Glatt
  • poloxamer, sodium hydroxide, hydroxypropylmethylcellulose and candee were mixed with purified water and ethanol.
  • cellulose acetate (acetal group 32%) (Eastman Chemical Company, USA) and cellulose acetate (acetal group 39.8%) (Eastman Chemical Company, USA) are dissolved in a mixed solvent of ethanol and methylene chloride After the preparation, the tablet was coated with a high coater (SFC-30F, Sejong Machinery, Korea) to prepare an osmotic tablet.
  • a high coater SFC-30F, Sejong Machinery, Korea
  • the candesartan cilexetil pre-release pellet and atenolol delayed-release tablets were filled into capsule 0 using a capsule charger to prepare a capsule.
  • Athenolol, microcrystalline cellulose (AvicelPH, FMC Biopolymer, USA), sodium lauryl sulfate, hydroxypropyl cellulose, carbomer 71G, and crosslinked vinylpyrrolidone were weighed into the ingredients and contents shown in Table 6.
  • the mixture was prepared by mixing for 20 minutes in a cone mixer. Magnesium stearate was added to the mixture for final mixing, followed by compression with a 5 mm diameter punch using a rotary tablet press (Sejong Pharmatech-30: Sejong).
  • candesartan cilexetil, colloidal silicon oxide, poloxamer, and hydroxypropyl cellulose were dissolved in a mixture of ethanol and methylene chloride to prepare a candesartan cilexetil coating solution.
  • Atenolol delayed-release tablets were administered to a high coater (SFC-30F Sejong Machinery, Korea) and coated with the candesartan cilexetil coating solution of (2). After the drug coating was completed, hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were coated with a coating solution dissolved and dispersed in ethanol and purified water.
  • Carvedilol delayed-release granules were prepared in the same manner as the method of preparing granules of delayed-release layer granules of (2) nebivolol hydrochloride of Example 1, except that carvedilol was used instead of nebivolol hydrochloride in the ingredients and contents shown in Table 6. Prepared.
  • the carvedilol delayed-release layer granules prepared in (2) were compressed into tablets with a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 5 mm diameter punch to prepare an inner core, followed by a 12 mm punch equipped oil. It was compressed with the eprosartan fast-release granules of (1) in a nuclear tablet press (RUD-I Killian, Germany). Tablets are finished tablets hydroxypropyl methyl cellulose 2910, polyethylene glycol 6000, titanium oxide was dissolved and dispersed in ethanol and purified water coating the tablet with a coating solution to prepare a nucleated tablet.
  • MRC-33 Sejong Pharmatech, Korea
  • Example 6 The ingredients and contents shown in Table 6 were prepared according to the method of Example 36 (1), except that candesartan cilexetil (Ranbaxy, India) was used instead of eprosartan.
  • Comparative dissolution tests were performed using valsartan (Novartis: Diovan, Valsartan monolith) and nebivolol (Forest labs: Bystolic, nebivolol monolith) as the tablets and the control agent obtained in Example 2.
  • the dissolution test was performed by changing the elution solution from 0.1 N-hydrochloric acid solution (acidic environment) to pH 6.8 (phosphate solution).
  • phosphate buffer was used.
  • the dissolution test method for each component is as follows, and the results are shown in FIG. 1 (the number of test samples is 12 pieces each).
  • the tablet of Example 2 was found to have almost the same elution characteristics as the valsartan component in the dissolution test under the following conditions, compared to the control agent Diovan, and the nebivolol component was compared with the control agent Bystolic. Very slow elution rates can be seen.
  • the nebivolol / valsartan double tablets of Example 2 had a dissolution rate of all of the nebivolol components up to 2 hours within 10%, much slower than the dissolution rate of the control formulation (about 99%).
  • the tablets of the nebivolol / valsartan of the present invention are unlikely to be metabolized in the liver before the valsartan because the initial release of the nebivolol is much slower than the valsartan, in contrast to the control drug nevivolol single agent.
  • Dissolution test basis Dissolution test method of General Test Method
  • Test method Paddle method, 50 revolutions / minute
  • Test solution 0.1 N hydrochloric acid solution, 500 mL (0-2 hours),
  • Test method Paddle method, 50 revolutions / minute
  • Formulations of various formulations obtained in Examples 2, 3, 4, and 5 were carried out according to the nebivolol comparative elution method of Experimental Example 1, and the results are shown in FIG. 2 (the number of test samples was 12 each).
  • the preparation prepared in Examples 2 to 5 for the purpose of the present invention can control the blood pressure during the day time of nebivolol delayed eluting during oral administration during the evening.
  • the formulation of the present invention can be developed in all formulations of the examples.
  • Example 14 Using the tablets prepared in Example 14 and the control agent Losartan (Merck: Cozaar, Losartan single agent) and nebivolol (Forest labs: Bystolic, Nebivolol single agent) according to the comparative test method of Navivolol of Experimental Example 1 Losartan comparative dissolution test method was carried out as follows, each result is shown in Figure 3 (the number of test samples each 12).
  • FIG. 3 is a result of comparative dissolution test of the tablet of Example 14, a combination agent of losartan and nebivolol, which is an angiotensin-2 receptor antagonist drug, and a control drug of losartan and nebivolol.
  • the drug began to elute 2 hours after the start and it was confirmed that more than 80% eluted within 1 hour.
  • Dissolution test basis 'Losartan Potassium tablet' in Dissolution Methods for Drug Products (FDA)
  • Test method Paddle method, 50 revolutions / minute
  • Test liquid water (degassing), 900 mL
  • Figure 4 shows the comparative dissolution test results of the formulations of Examples 21, 22, 23, 24 and the nebivolol control, the nebivolol is 1 hour after starting the drug dissolution 2 hours after the start of the dissolution test intended by the present invention It was confirmed that more than 80% eluted within.
  • Example 34 A comparative dissolution test was conducted using the tablet prepared in Example 34 and the control agent candesartan (Astrazeneca: Atacand, candesartan mono) and atenolol (modern drugs: Hyundai tenolmin tablet, atenolol mono). It was.
  • the dissolution test was performed by changing the eluate from 0.1 N hydrochloric acid solution (acidic environment) to pH 6.8 (phosphate) solution.
  • the dissolution test method for each component is as follows, and the results are shown in FIG. 6 (the number of test samples is 12 each).
  • Example 34 the tablet of Example 34 was confirmed that the candesartan component in the dissolution test under the following conditions compared to the control agent atacane (Astrageneca: Atacand, Candesartan single agent), but almost the same elution characteristics, The norol component can be seen that the initial drug release is delayed when comparing the dissolution results with the control agent Hyundaitenolmin.
  • the atenolol / candesartan tablet of the present invention is unlikely to be metabolized in the liver prior to candesartan because the initial release of atenolol is much slower than candesartan, unlike the atenolol monotherapy, which is a reference drug.
  • Dissolution test basis Dissolution test method of General Test Method
  • Test method Paddle method, 50 revolutions / minute
  • Test solution 0.1 N hydrochloric acid solution, 500 mL (0-2 hours),
  • Dissolution test basis 'Candesartan Cilexetil tablet' in Dissolution Methods for Drug Products (FDA)
  • Test method Paddle method, 50 revolutions / minute
  • Formulations of the various formulations obtained in Examples 36, 37 and 39 and the control agent eprosartan (Abbott: Teveten, eprosartan monolith), carvedilol (common root: dilatrene, carvedilol monolith), telme Comparative dissolution tests were carried out using Sartan (Boehringer ingelheim: Micardis, Telmisartan mono), Metoprolol (Astrazeneca: Toprol-XL, Metoprolol sustained-release tablet mono).
  • the dissolution test was performed by changing the eluate from 0.1 N hydrochloric acid solution (acid environment) to pH 6.8 (phosphate solution).
  • the dissolution test method for each component is as follows, and the results are shown in FIGS. 7, 8, and 9 (the test population is 12 each).
  • carvedilol and metoprolol which are adrenergic beta receptor blocker-based drugs other than nebivolol, also have an intended release pattern.
  • Dissolution test basis Dissolution test method of General Test Method
  • Test method Paddle method, 50 revolutions / minute
  • Test solution 0.1 N hydrochloric acid solution, 500 mL (0-2 hours),
  • Test method Paddle method, 50 revolutions / minute
  • Test solution 0.1 N hydrochloric acid solution, 500 mL (0-2 hours),
  • Dissolution test basis 'Eprosartan Mesylate / Hydrochlorothiazide tablet' in Dissolution Methods for Drug Products (FDA)
  • Test method Paddle method, 75 revolutions / minute
  • Dissolution test basis 'Telmisartan tablet' in Dissolution Methods for Drug Products (FDA)
  • Test method Paddle method, 75 revolutions / minute
  • the formulation of the present invention exhibits an equal anti-pressure action and prevents complications, and is particularly useful for hypertension patients with complications, suppressing blood pressure during sleep, and for drug interactions. Reduce side effects.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention provides a pharmaceutical preparation comprising a prerelease compartment that includes an angiotension-2 receptor antagonist as its pharmacologically active ingredient and a delay-release compartment that includes a beta adrenalin blocker as its pharmacologically active ingredient; the preparation of the present invention acts to maintain a uniform constant pressure and prevent complications, and in particular, enables holding blood pressure uniform during the risk period for complications, so that it has the advantage of being useful for hypertension patients with complications, patients requiring blood pressure suppression during sleep, etc., and of reducing adverse reactions caused by drug interactions.

Description

약제학적 제제Pharmaceutical preparations
본 발명은 안지오텐신-2 수용체 길항제와 베타 아드레날린 차단제를 유효성분으로 함유하며, 각각의 성분이 시간차를 두고 방출하는 약제학적 제제에 관한 것이다.The present invention relates to a pharmaceutical preparation containing an angiotensin-2 receptor antagonist and a beta adrenergic blocker as an active ingredient, each of which is released over time.
고혈압은 매우 다양한 원인에 의해 중복적으로 유발되는 증상이다. 따라서 단일 항압제를 사용하는 경우 어떤 결과가 나오는 지는 미리 판단하기 어렵다[Journal of hypertension 1995: 9: S33-S36]. 이러한 이유로 보다 효과적인 치료를 위하여 항압제들의 복합 처방이 계속 증가하여 왔다. Hypertension is a condition that is caused by multiple causes. Therefore, it is difficult to determine in advance what results would result from the use of a single anticompressant [Journal of hypertension 1995: 9: S33-S36]. For this reason, a combination of anti-pressure agents has been increasing for more effective treatment.
지난 30년 동안 발표된 대단위 임상(예: Hypertension Optimal Treatment, United Kingdom Prospective Diabetes Study 등) 보고서에도 경증 내지 중등증 고혈압때부터 복합 처방으로 치료함으로써 합병증 발병 및 악화를 예방하고 장수를 보장받을 수 있다는 입증자료가 계속 증가해 왔다[미국 고혈압 대책 위원회 고혈압 치료 지침, JNC(Joint National Committee) Report 6 and 7, World Health Organization-International Society of Hypertension (1999)]. Large clinical reports (e.g. Hypertension Optimal Treatment, United Kingdom Prospective Diabetes Study, etc.) published over the past 30 years demonstrate that treatment with complications from mild to moderate hypertension prevents complications and worsens and ensures longevity. Data continues to increase [US Hypertension Measures Guidelines for Hypertension Treatment, Joint National Committee Report 6 and 7, World Health Organization-International Society of Hypertension (1999).
심혈관 질환 치료용 복합제제에 관하여, 미국 공개특허 제 2005-0004194호에는 심근 경색 환자에 대해 부작용을 줄이고 효과를 높여 심혈관 질환을 치료하기 위한 안지오텐신 전환효소 저해제와 안지오텐신-2 수용체 길항제의 복합처방이 개시되어 있고, 대한민국 공개특허 제2004-0078140호에는 안지오텐신-2 수용체 길항제인 발사르탄과 칼슘 채널 차단제의 항 고혈압 복합제제가 개시되어 있다. Regarding the combination preparation for cardiovascular disease, US Patent Publication No. 2005-0004194 discloses a combination of angiotensin converting enzyme inhibitor and angiotensin-2 receptor antagonist for treating cardiovascular disease by reducing side effects and enhancing the effects on patients with myocardial infarction. Korean Patent Laid-Open Publication No. 2004-0078140 discloses an antihypertensive complex preparation of valsartan and calcium channel blocker, which is an angiotensin-2 receptor antagonist.
또한, 국제공개공보 WO03/097099에는 안지오텐신-2 수용체 차단제 및 베타수용체 차단제 각각의 단위 제형을 동시에 투여하는 단순 병용 요법이 심근경색증 후 환자의 심혈관 질환의 치료에 효과적임이 개시되어 있다. In addition, WO 03/097099 discloses that simple combination therapy in which unit dosage forms of angiotensin-2 receptor blocker and beta receptor blocker are simultaneously administered is effective for the treatment of cardiovascular disease in patients after myocardial infarction.
본 발명자들은 고혈압 등의 심혈관질환에 효과적인 복합제제를 개발하기 위한 연구결과 본 발명을 완성하였다. The present inventors have completed the present invention as a result of research to develop a complex formulation for cardiovascular diseases such as hypertension.
본 발명이 해결하고자 하는 과제는 균등한 항압작용과 합병증 예방 작용을 나타내며, 특히 합병증 발생 위험시간대에 혈압을 균등하게 유지시켜 줄 수 있어 합병증을 지닌 고혈압 환자, 수면 중 혈압억제 등에 유용하고, 약물간 상호작용에 따른 부작용을 감소시켜 주는 제제를 제공하고자 한다.The problem to be solved by the present invention is to show an equal anti-pressure action and prevent complications, in particular, it is possible to maintain the blood pressure evenly in the dangerous time period of complications, it is useful for patients with hypertension with complications, blood pressure control during sleep, etc. It is intended to provide an agent that reduces the side effects of interaction.
본 발명은 약리학적 활성성분으로 안지오텐신-2 수용체 길항제를 포함하는 선방출성 구획, 및 약리학적 활성성분으로 베타 아드레날린 차단제를 포함하는 지연방출성 구획을 포함하는 약제학적 제제를 제공한다. The present invention provides a pharmaceutical formulation comprising a prior release compartment comprising an angiotensin-2 receptor antagonist as a pharmacologically active ingredient, and a delayed release compartment comprising a beta adrenergic blocker as a pharmacologically active ingredient.
본 발명의 제제 중 선방출성 구획의 활성성분인 안지오텐신-2 수용체 길항제는 선방출성이며, 바람직하게는 안지오텐신-2 수용체 길항제의 방출개시 후1시간 이내에 안지오텐신-2 수용체 길항제 총량의 85%이상이 방출되어, 원하는 약효를 신속하게 발생할 수 있다.In the preparation of the present invention, the angiotensin-2 receptor antagonist, which is the active ingredient of the pre-release compartment, is prerelease, and preferably, at least 85% of the total amount of the angiotensin-2 receptor antagonist is released within 1 hour after the release of the angiotensin-2 receptor antagonist. Therefore, the desired effect can be generated quickly.
본 발명의 제제 중 지연방출성 구획의 베타 아드레날린 차단제는 지연시간 경과 후, 즉, 안지오텐신-2 수용체 길항제 방출 1시간 내지 8시간 후, 바람직하게는 1시간 내지 6시간 후에 방출된다.The beta adrenergic blocker of the delayed-release compartment in the formulations of the invention is released after a delay, ie 1 to 8 hours after release of the angiotensin-2 receptor antagonist, preferably 1 to 6 hours.
본 발명 제제는 안지오텐신-2수용체 길항제 방출개시 후 2시간 이내에 방출되는 베타 아드레날린 차단제의 방출량이 베타 아드레날린 차단제 총량의 0~10% 이내로 하여, 베타 아드레날린 차단제 방출이 지연되어 원하는 약효를 지연시간 경과 후발생할 수 있다.In the present invention, the release amount of the beta adrenergic blocker released within 2 hours after the release of the angiotensin-2 receptor antagonist is within 0-10% of the total amount of the beta adrenergic blocker. Can be.
또한, 본 발명의 제제는 상기 아드레날린 차단제 1중량부에 대하여 안지오텐신-2 수용체 길항제를 0.1~350중량부 포함할 수 있다.In addition, the formulation of the present invention may comprise 0.1 to 350 parts by weight of the angiotensin-2 receptor antagonist based on 1 part by weight of the adrenergic blocker.
본 발명 제제의 각 구획을 보다 상세히 설명하면 다음과 같다. Each compartment of the formulation of the present invention will be described in more detail as follows.
1. 선(先)방출성 구획 1.Preventive compartment
선방출성 구획은 본 발명의 약제학적 제제에 있어서 지연방출성 구획에 비해 먼저 활성성분이 방출되는 구획을 의미하며, 약리학적 활성성분 외에 필요에 따라 약제학적으로 허용 가능한 첨가제를 추가로 포함할 수 있다. Pre-release compartment refers to the compartment in which the active ingredient is first released in comparison with the delayed-release compartment in the pharmaceutical formulation of the present invention, and may further include a pharmaceutically acceptable additive as necessary in addition to the pharmacologically active ingredient. .
(1) 약리학적 활성성분 (1) pharmacologically active ingredients
선방출성 구획의 약리학적 활성성분은 안지오텐신-2 수용체 길항제(Angiotensin 2 receptor blocker)로 상기 안지오텐신-2 수용체 길항제는 혈관수축을 일으키는 근원물질 중 하나인 안지오텐신 2가 안지오텐신 수용체 중 AT1 수용체와 결합하는 것을 차단하여, 심근의 수축기와 확장기 모두에 있어 혈압강하효과를 발휘하는 약물이며, 로사르탄, 발사르탄, 텔미사르탄, 이르베사르탄, 칸데사르탄, 올메사르탄, 및/또는 에프로사르탄 등과 이들의 이성질체 및 이들의 약학적으로 허용 가능한 염 중에서 선택될 수 있으며, 제제 중 1 ~ 800mg 포함할 수 있고, 바람직하게는 15 ~ 370mg을 포함한다. 상기 용량은 1일 성인(체중 65~75kg 성인남자) 기준의 용량이다.The pharmacologically active component of the prior-release compartment is an angiotensin 2 receptor antagonist, and the angiotensin-2 receptor antagonist blocks angiotensin 2, one of the sources of vasoconstriction, with the AT1 receptor in the angiotensin receptor. It is a drug that exerts a blood pressure-lowering effect on both myocardial systolic and diastolic phases, such as losartan, valsartan, telmisartan, irbesartan, candesartan, olmesartan, and / or eprosartan. Isomers of and pharmaceutically acceptable salts thereof may be selected, and may include 1 to 800 mg of the formulation, and preferably 15 to 370 mg. The dose is based on a daily adult (65-75 kg adult male).
(2) 약제학적으로 허용가능한 첨가제 (2) pharmaceutically acceptable additives
본 발명의 제제는 본 발명의 효과를 해치지 않는 범위에서 약학적으로 허용 가능한 희석제, 결합제, 붕해제, 윤활제, pH 조절제, 용해보조제 등의 통상적으로 사용되는 첨가제를 약리학적 활성성분의 방출을 방해하지 않는 범위 내에서 추가로 사용하여 제제화할 수 있다.The formulations of the present invention do not interfere with the release of pharmacologically active ingredients from commonly used additives such as pharmaceutically acceptable diluents, binders, disintegrants, lubricants, pH adjusters, dissolution aids and the like without departing from the effect of the present invention. It can be formulated using further within the range which does not.
상기 희석제는 전분, 미세결정 셀룰로오스, 유당, 포도당, 만니톨, 알기네이트, 알칼리토금속류염, 클레이, 폴리에틸렌글리콜, 디칼슘포스페이트, 또는 이들의 혼합물 등을 사용할 수 있다. The diluent may be starch, microcrystalline cellulose, lactose, glucose, mannitol, alginate, alkaline earth metal salts, clay, polyethylene glycol, dicalcium phosphate, or a mixture thereof.
상기 결합제는 전분, 미세결정 셀룰로오스, 고분산성 실리카, 만니톨, 자당, 유당, 폴리에틸렌글리콜, 폴리비닐피롤리돈, 히드록시프로필메틸셀룰로오스, 히드록시프로필셀룰로오스, 천연검, 합성검, 폴리비닐피롤리돈 공중합체, 포비돈, 젤라틴, 또는 이들의 혼합물 등을 사용할 수 있다. The binder is starch, microcrystalline cellulose, highly dispersible silica, mannitol, sucrose, lactose, polyethylene glycol, polyvinylpyrrolidone, hydroxypropylmethylcellulose, hydroxypropylcellulose, natural gum, synthetic gum, polyvinylpyrrolidone Copolymers, povidone, gelatin, mixtures thereof, and the like.
상기 붕해제는 나트륨전분글리콜레이트, 옥수수전분, 감자전분 또는 전젤라틴화전분(전호화전분) 등의 전분 또는 변성전분; 벤토나이트, 몬모릴로나이트, 또는 비검(veegum) 등의 클레이; 미결정셀룰로오스, 히드록시프로필셀룰로오스 또는 카르복시메틸셀룰로오스 등의 셀룰로오스류; 알긴산나트륨 또는 알긴산 등의 알긴류; 크로스카멜로스(croscarmellose)나트륨 등의 가교 셀룰로오스류; 구아검, 잔탄검 등의 검류; 가교 폴리비닐피롤리돈(crospovidone) 등의 가교 중합체; 중탄산나트륨, 시트르산 등의 비등성 제제, 또는 이들의 혼합물을 사용할 수 있다.The disintegrant may be a starch or modified starch such as sodium starch glycolate, corn starch, potato starch or starch gelatinized starch (starch starch); Clay such as bentonite, montmorillonite, or veegum; Celluloses such as microcrystalline cellulose, hydroxypropyl cellulose or carboxymethyl cellulose; Algins such as sodium alginate or alginic acid; Crosslinked celluloses such as croscarmellose sodium; Gums such as guar gum and xanthan gum; Crosslinked polymers such as crosslinked polyvinylpyrrolidone (crospovidone); Effervescent agents such as sodium bicarbonate, citric acid, or mixtures thereof can be used.
상기 윤활제는 탈크, 스테아르산, 스테아르산 마그네슘, 스테아르산 칼슘, 라우릴황산나트륨, 수소화식물성오일, 나트륨벤조에이트, 나트륨스테아릴푸마레이트, 글리세릴 베헤네이트, 글리세릴 모노레이트, 글리세릴모노스테아레이트, 글리세릴 팔미토스테아레이트, 폴리에틸렌글리콜 및/또는 이들의 혼합물 등을 사용할 수 있다. The lubricant is talc, stearic acid, magnesium stearate, calcium stearate, sodium lauryl sulfate, hydrogenated vegetable oil, sodium benzoate, sodium stearyl fumarate, glyceryl behenate, glyceryl monorate, glyceryl monostearate, Glyceryl palmitostearate, polyethylene glycol, and / or mixtures thereof, and the like.
상기 안정화제는 아스코르빈산, 구연산, 부틸레이티드히드록시 아니솔, 부틸레이티드히드록시 톨루엔, 및/또는 토코페롤 유도체 등을 사용할 수 있다. 또한, 안정화제로서 알칼리금속의 염, 알칼리토금속의 염, 또는 이들의 혼합물인 알칼리화제를 사용할 수 있으며, 바람직하게는 탄산칼슘, 탄산나트륨, 탄산수소나트륨, 산화마그네슘, 탄산마그네슘, 및/또는 구연산나트륨 등을 사용할 수 있다. 상기 pH 조절제는 초산, 아디프산, 아스코르브산, 사과산, 숙신산, 주석산, 푸마르산, 구연산과 같은 산성화제와 침강 탄산 칼슘, 암모니아수, 메글루민과 같은 염기성화제 등을 사용할 수 있다. The stabilizer may be ascorbic acid, citric acid, butylated hydroxy anisole, butylated hydroxy toluene, and / or tocopherol derivatives. In addition, as a stabilizer, an alkalizing agent, which is a salt of an alkali metal, a salt of an alkaline earth metal, or a mixture thereof, may be used. Preferably, calcium carbonate, sodium carbonate, sodium hydrogen carbonate, magnesium oxide, magnesium carbonate, and / or sodium citrate Etc. can be used. The pH adjusting agent may be an acidifying agent such as acetic acid, adipic acid, ascorbic acid, malic acid, succinic acid, tartaric acid, fumaric acid, citric acid, and a basicizing agent such as precipitated calcium carbonate, aqueous ammonia, and meglumine.
상기 용해보조제는 라우릴황산나트륨, 폴리소르베이트 등의 폴리옥시에틸렌 소르비탄 지방산 에스테르, 도큐세이트 나트륨 등을 사용할 수 있다. The dissolution aid may be used polyoxyethylene sorbitan fatty acid esters such as sodium lauryl sulfate, polysorbate, sodium docusate and the like.
이외에도 착색제, 향료 중에서 선택된 다양한 첨가제로서 약학적으로 허용 가능한 첨가제를 선택 사용하여 본 발명의 제제를 제제화할 수 있다. In addition, a pharmaceutically acceptable additive may be selected and used in the preparation of the present invention as various additives selected from colorants and fragrances.
본 발명에서 사용가능한 첨가제의 범위가 상기 첨가제를 사용하는 것으로 한정되는 것은 아니며, 상기한 첨가제는 선택에 의하여 통상 범위의 용량을 함유하여 제제화할 수 있다.The range of additives usable in the present invention is not limited to the use of such additives, and the above additives may be formulated to contain a range of dosages, usually by selection.
2. 지연방출성 구획 2. Delayed release block
본 발명에서 지연방출성 구획은 선방출성 구획 활성성분의 방출 일정 시간 후에 그 활성성분이 방출되는 구획을 의미한다. 지연방출성 구획은 (1)약리학적 활성성분 및 (2-1)방출제어물질 또는 (2-2)삼투압 조절제 및 반투과성막 코팅기제를 포함하며, 필요에 따라, (3) 약제학적으로 허용 가능한 첨가제를 추가로 포함할 수 있다. In the present invention, the delayed-release compartment refers to a compartment in which the active ingredient is released after a certain time of release of the prior-release compartment active ingredient. The delayed-release compartment comprises (1) a pharmacologically active ingredient and (2-1) release controlling substance or (2-2) an osmotic pressure regulator and a semipermeable membrane coating base, and (3) a pharmaceutically acceptable It may further include an additive.
(1) 약리학적 활성성분 (1) pharmacologically active ingredients
지연방출성 구획의 약리학적 활성성분은 베타아드레날린차단제(beta adrenergic antagonist)로, 상기 베타아드레날린차단제는 심혈관 및 평활근에 많이 존재하는 β수용체가 아드레날린에 의한 자극으로 교감신경계가 흥분되는 것을 차단하여 고혈압, 협심증, 심부정맥을 포함하는 여러가지 심장질환을 치료하는 데 사용되는 약물이며, 상기 베타아드레날린차단제는 알프레놀롤, 아세부토롤, 아모술랄롤, 아로티놀롤, 아테놀롤, 베푸놀롤, 베탁솔롤, 베반톨롤, 비소프롤롤, 보핀돌롤, 부쿠몰롤, 뷔페톨롤, 부푸랄롤, 부니트롤롤, 부프란돌롤, 부토필롤롤, 카라졸롤, 카테올롤, 카르베딜롤, 셀릴프롤롤, 세타몰롤, 클로라놀롤, 딜레발롤, 에파놀롤, 인데놀롤, 라베탈롤, 레보부놀롤, 메핀돌롤, 메티프라놀롤, 메토프롤롤, 모프롤롤, 나돌롤, 나독솔롤, 염산 네비볼롤, 니프라딜롤, 옥스프레놀롤, 퍼부톨롤, 핀돌롤, 프랙톨롤, 프로네탈롤, 프로프라놀롤, 소탈롤, 수피날롤, 탈린돌, 테르타톨롤, 틸리솔롤, 티몰롤, 톨리프롤롤, 및/또는 지베놀롤 등, 이들의 이성질체, 이들의 약학적으로 허용 가능한 염 등에서 선택될 수 있으며, 네비볼롤이 바람직하다. 상기 베타아드레날린 차단제는 제제 중1 ~ 1400mg 포함할 수 있고, 바람직하게는 2 ~ 300mg을 포함한다. 상기 용량은 1일 성인(체중 65~75kg 성인남자) 기준의 용량이다.The pharmacologically active component of the delayed-release compartment is beta adrenergic antagonist, and the beta adrenergic blocker blocks the excitability of the sympathetic nervous system due to the stimulation of the adrenergic beta receptors in the cardiovascular and smooth muscle, resulting in hypertension It is a drug used to treat various heart diseases including angina and deep vein, and the beta adrenergic blocker is alprenolol, acebutorol, amosulolol, arotinol, atenolol, befunolol, betaxolol, bevan Tolol, Bisoprolol, Boffindolol, Bucumolol, Buffololol, Bufuralol, Bunitrolol, Bufrandolol, Butopylolol, Carazolol, Caterolol, Carvedilol, Celylprolol, Cetamolol, Chlorolol , Dilevalol, epanolol, indenolol, labetalol, levobunol, mepindolol, metipranolol, metoprolol, moprolol, nadolol, nadoxolol, and nebivolol hydrochloride , Nipradilol, oxprenolol, perbutolol, pindolol, fractolol, pronetrol, propranolol, sotalol, supinalol, tallinol, tertatolol, tilisolol, timolol, toliprolol, and / or Gibenolol and the like, isomers thereof, pharmaceutically acceptable salts thereof, and the like, and nebivolol is preferred. The beta adrenergic blocker may comprise 1 to 1400 mg of the formulation, preferably 2 to 300 mg. The dose is based on a daily adult (65-75 kg adult male).
네비볼롤은 제4세대 베타 아드레날린 차단제로, 1, 2, 3세대 베타 아드레날린 차단제가 갖는 모든 장점을 가질 뿐만 아니라, NO를 생성시켜 말초 혈관을 확장시켜 주므로, 다른 베타 아드레날린 차단제가 갖는 성기능 퇴화, 지질증가, 당뇨병 악화, 사지 냉감 등의 부작용이 없으며, 우심 확장성 심부전에도 효과를 나타낸다. 특히 노인성 수축기 고혈압자로서 심부전을 지닌 자, 노인성 당뇨병성 고혈압자, 노인성 고지질성 고혈압자, 심부전을 겸한 고혈압자에게 적합하다. Nebivolol is a fourth-generation beta adrenergic blocker that not only has all the benefits of first-, second- and third-generation beta-adrenergic blockers, but also produces NO, which dilates peripheral blood vessels. It has no side effects such as increase, worsening of diabetes mellitus, cold limbs, and effects on right heart dilatation heart failure. It is particularly suitable for elderly systolic hypertensives, those with heart failure, elderly diabetic hypertension, elderly hyperlipidemic hypertension, and hypertension combined with heart failure.
따라서, 네비볼롤은 심부전, 심근 경색 등 중증 질환자의 고혈압에 안지오텐신-2 수용체 길항제를 복합함으로써, 유효한 치료효과를 얻을 수 있다. Therefore, nebivolol can obtain an effective therapeutic effect by combining angiotensin-2 receptor antagonist with hypertension of severe diseases such as heart failure and myocardial infarction.
(2-1) 방출제어물질 (2-1) Release Control Substances
본 발명의 약제학적 제제 중 지연방출성 구획은 방출제어물질을 포함하며, 본 발명 제제의 방출제어 물질은 예를 들어, 장용성 고분자, 수불용성 중합체, 소수성 화합물, 친수성 고분자, 및 이들의 혼합물 중에서 선택된 하나 이상으로, 바람직하게는 수불용성 중합체 및 장용성 고분자 중에서 선택된 하나 이상이다. 상기 제제의 방출제어물질은 베타 아드레날린 차단제 1 중량부에 대하여 0.1 ~ 100 중량부, 바람직하게는 1~50중량부이며, 0.1중량부 미만일 경우 충분한 지연시간을 갖기 어려울 염려가 있고, 100중량부 초과시 약물방출이 과도하게 지연되어 유의성 있는 임상적 효과를 얻기 어려울 수 있다. The delayed-release compartment in the pharmaceutical formulation of the present invention comprises a release controlling substance, wherein the release controlling substance of the present formulation is selected from, for example, enteric polymers, water insoluble polymers, hydrophobic compounds, hydrophilic polymers, and mixtures thereof. At least one, preferably at least one selected from water-insoluble polymers and enteric polymers. The release controlling substance of the formulation is 0.1 to 100 parts by weight, preferably 1 to 50 parts by weight, with respect to 1 part by weight of the beta adrenergic blocker, and when it is less than 0.1 parts by weight, it is difficult to have a sufficient delay time. Drug release may be excessively delayed, making it difficult to obtain significant clinical effects.
상기 장용성 고분자는 산성 조건하에서 불용성이거나 또는 안정한 것으로, pH5 이상인 특정 pH조건하에서 용해되거나 또는 분해되는 고분자를 말한다. 본 발명에서 사용가능한 장용성 고분자는 장용성 셀룰로오스 유도체, 장용성 아크릴산계 공중합체, 장용성 폴리메타크릴레이트 공중합체, 장용성 말레인산계 공중합체, 장용성 폴리비닐 유도체, 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상이다. The enteric polymer is insoluble or stable under acidic conditions, and refers to a polymer that is dissolved or decomposed under specific pH conditions of pH 5 or more. The enteric polymer usable in the present invention is at least one selected from the group consisting of an enteric cellulose derivative, an enteric acrylic acid copolymer, an enteric polymethacrylate copolymer, an enteric maleic acid copolymer, an enteric polyvinyl derivative, and a mixture thereof. .
여기서, 장용성 셀룰로오스 유도체는 히드록시프로필메틸셀룰로오스아세테이트숙시네이트(또는 히프로멜로오스아세테이트숙시네이트이라 함), 히드록시프로필메틸셀룰로오스프탈레이트(또는 히프로멜로오스프탈레이트이라 함), 히드록시메틸에틸셀룰로오스프탈레이트, 셀룰로오스아세테이트프탈레이트, 셀룰로오스아세테이트숙시네이트, 셀룰로오스아세테이트말레이트, 셀룰로오스벤조에이트프탈레이트, 셀룰로오스프로피오네이트프탈레이트, 메틸셀룰로오스프탈레이트, 카르복시메틸에틸셀룰로오스, 에틸히드록시에틸셀룰로오스프탈레이트, 메틸히드록시에틸셀룰로오스 및 이들의 혼합물 중에서 선택된 1종 이상이며; 상기 장용성 아크릴산계 공중합체는 스티렌-아크릴산 공중합체, 아크릴산메틸-아크릴산 공중합체, 아크릴산메틸메타크릴산 공중합체, 아크릴산부틸-스티렌-아크릴산 공중합체, 아크릴산메틸-메타크릴산-아크릴산옥틸공중합체 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 장용성 폴리메타크릴레이트 공중합체는 폴리(메타크릴산-메틸메타크릴레이트) 공중합체(예컨대, 유드라짓 L 100, 유드라짓 S, 에보닉, 독일), 폴리(메타크릴산ㆍ에틸아크릴레이트)공중합체(예컨대, 유드라짓 L 100-55, 에보닉, 독일 아크릴이즈, Colorcon, USA) 및 이들의 혼합물 중에서 선택된 1종이상이고; 상기 장용성 말레인산계 공중합체는 아세트산비닐-말레인산 무수물 공중합체, 스티렌-말레인산 무수물 공중합체, 스티렌-말레인산모노에스테르 공중합체, 비닐메틸에테르-말레인산 무수물 공중합체, 에틸렌-말레인산 무수물 공중합체, 비닐부틸에테르-말레인산 무수물 공중합체, 아크릴로니트릴-아크릴산메틸ㆍ말레인산 무수물 공중합체, 아크릴산부틸-스티렌-말레인산 무수물 공중합체 및 이들의 혼합물 중에서 선택된 1종 이상이며; 상기 장용성 폴리비닐 유도체는 폴리비닐알콜프탈레이트, 폴리비닐아세테이트프탈레이트, 폴리비닐부티레이트프탈레이트, 폴리비닐아세트아세탈프탈레이트 및 이들의 혼합물 중에서 선택된 1종 이상이 바람직하다. 상기 장용성 고분자로 더욱 바람직한 예는 히드록시프로필메틸셀룰로오스 프탈레이트 및 아크릴산메틸메타크릴산 공중합체 중에서 선택된 하나 이상이다.Here, the enteric cellulose derivative may be hydroxypropylmethylcellulose acetate succinate (or hypromellose acetate succinate), hydroxypropylmethyl cellulose phthalate (or hypromellose phthalate), hydroxymethylethyl cellulose phthalate , Cellulose acetate phthalate, cellulose acetate succinate, cellulose acetate maleate, cellulose benzoate phthalate, cellulose propionate phthalate, methyl cellulose phthalate, carboxymethylethyl cellulose, ethyl hydroxyethyl cellulose phthalate, methyl hydroxyethyl cellulose and their At least one selected from a mixture; The enteric acrylic acid copolymers include styrene-acrylic acid copolymers, methyl acrylate-acrylic acid copolymers, methyl methacrylate acrylates, butyl acrylate-styrene-acrylic acid copolymers, methyl acrylate-methacrylic acid-octyl acrylate copolymers, and these At least one selected from a mixture of; The enteric polymethacrylate copolymer is a poly (methacrylic acid-methylmethacrylate) copolymer (e.g. Eudragit L 100, Eudragit S, Evonik, Germany), poly (methacrylic acid ethylacryl At least one selected from copolymers (eg Eudragit L 100-55, Evonik, Acrylics, Germany, Colorcon, USA) and mixtures thereof; The enteric maleic acid copolymer is vinyl acetate-maleic anhydride copolymer, styrene-maleic anhydride copolymer, styrene-maleic acid monoester copolymer, vinyl methyl ether-maleic anhydride copolymer, ethylene-maleic anhydride copolymer, vinyl butyl ether- At least one selected from maleic anhydride copolymer, acrylonitrile-methyl acrylate maleic anhydride copolymer, butyl styrene-maleic-maleic anhydride copolymer and mixtures thereof; The enteric polyvinyl derivative is preferably at least one selected from polyvinyl alcohol phthalate, polyvinylacetate phthalate, polyvinyl butyrate phthalate, polyvinylacetacetal phthalate and mixtures thereof. More preferred examples of the enteric polymer are at least one selected from hydroxypropylmethylcellulose phthalate and methyl methacrylate acrylate.
여기서, 장용성 고분자는 활성성분1 중량부에 대해서 0.1 중량부 ~ 20 중량부, 바람직하게는 0.5 중량부 ~ 10 중량부로 포함될 수 있으며, 0.1 중량부 미만시 pH 5 미만에서 쉽게 용해될 염려가 있고, 20 중량부 초과시 불필요하게 제제 총중량이 커지거나 과도하게 용출이 지연될 염려가 있다. Here, the enteric polymer may be included in an amount of 0.1 parts by weight to 20 parts by weight, preferably 0.5 parts by weight to 10 parts by weight with respect to 1 part by weight of the active ingredient, and less than 0.1 parts by weight may be easily dissolved at a pH of less than 5, If the amount exceeds 20 parts by weight, the total weight of the preparation may be unnecessarily increased or the elution may be excessively delayed.
상기 수불용성 중합체는 약물의 방출을 제어하는 약제학적으로 허용가능한 물에 용해되지 않는 고분자를 말한다. 본 발명에서 사용가능한 수불용성 중합체는 폴리비닐 아세테이트, 수불용성 폴리메타크릴레이트 공중합체{예: 폴리(에틸아크릴레이트,메틸 메타크릴레이트) 공중합체(예컨대, 유드라짓NE30D), 폴리(에틸아크릴레이트,메틸 메타크릴레이트,트리메틸아미노에틸메타크릴레이트클로라이드)공중합체(예컨대, 유드라짓 RS, RL) 등}, 에틸셀룰로오스, 셀룰로오스 에스테르, 셀룰로오스 에테르, 셀룰로오스 아실레이트, 셀룰로오스 디아실레이트, 셀룰로오스 트리아실레이트, 셀룰로오스 아세테이트, 셀룰로오스 디아세테이트, 셀룰로오스 트리아세테이트 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상이 바람직하며, 더욱 바람직하게는 폴리비닐아세테이트, 에틸셀룰로오스, 폴리(에틸아크릴레이트,메틸 메타크릴레이트, 트리메틸아미노에틸메타크릴레이트 클로라이드)공중합체, 및 셀룰로오스 아세테이트 중에서 선택된 하나 이상이다. 여기서, 수불용성 중합체는 활성성분 1 중량부에 대해서 0.1 중량부 ~ 30 중량부, 바람직하게는 0.5 중량부 ~ 20 중량부로 포함될 수 있으며, 0.1 중량부 미만인 경우에는 약물의 방출이 제어되지 않을 염려가 있고, 30 중량부 초과인 경우에는 과도하게 용출이 지연될 염려가 있다. The water insoluble polymer refers to a polymer that is not soluble in pharmaceutically acceptable water that controls the release of the drug. The water-insoluble polymers usable in the present invention are polyvinyl acetate, water-insoluble polymethacrylate copolymers (e.g. poly (ethylacrylate, methyl methacrylate) copolymers (eg Eudragit NE30D), poly (ethylacrylic) Latex, methyl methacrylate, trimethylaminoethyl methacrylate chloride) copolymer (e.g. Eudragit RS, RL), etc.}, ethyl cellulose, cellulose ester, cellulose ether, cellulose acylate, cellulose dicylate, cellulose tri At least one selected from the group consisting of acylate, cellulose acetate, cellulose diacetate, cellulose triacetate and mixtures thereof is preferred, and more preferably polyvinylacetate, ethyl cellulose, poly (ethyl acrylate, methyl methacrylate). , Trimethylaminoethyl methacrylate Bit chloride) is at least one selected from a copolymer, and cellulose acetate. Herein, the water-insoluble polymer may be included in an amount of 0.1 parts by weight to 30 parts by weight, preferably 0.5 parts by weight to 20 parts by weight, and less than 0.1 parts by weight based on 1 part by weight of the active ingredient. If the content is more than 30 parts by weight, the dissolution may be excessively delayed.
상기 소수성 화합물은 약물의 방출을 제어하는 약제학적으로 허용가능한 물에 용해되지 않는 물질을 말한다. 본 발명에서 사용가능한 소수성 화합물은 예를 들어, 지방산 및 지방산 에스테르류, 지방산 알코올류, 왁스류, 무기물질, 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상이다. 여기서, 지방산 및 지방산 에스테르류는 글리세릴 팔미토스테아레이트, 글리세릴 스테아레이트, 글리세릴 디스테아레이트, 글리세릴 비헤네이트, 세틸 팔미테이트, 글리세릴 모노 올레이트, 스테아르산 및 이들의 혼합물 중에서 선택된 1종 이상이고; 지방산 알코올류는 세토스테아릴 알코올, 세틸알코올, 스테아릴알코올 및 이들의 혼합물 중에서 선택된 1종 이상이며; 왁스류는 카르나우바왁스, 밀납, 미결정왁스, 및 이들의 혼합물 중에서 선택된 1종 이상이고; 무기물질은 탈크, 침강탄산칼슘, 인산일수소칼슘, 산화아연, 산화티탄, 카올린, 벤토나이트, 몬모릴로나이트, 비검 및 이들의 혼합물 중에서 선택된 1종 이상인 것이 바람직하다. 상기 소수성 화합물의 더욱 바람직한 예는 카르나우바왁스이다. 여기서, 소수성 화합물은 활성성분 1 중량부에 대해서 0.1 중량부 ~ 20 중량부, 바람직하게는 0.5 중량부~ 10 중량부로 포함될 수 있으며, 0.1 중량부 미만인 경우에는 약물의 방출이 제어되지 않을 염려가 있고, 20 중량부 초과인 경우에는 과도하게 용출이 지연될 염려가 있다. The hydrophobic compound refers to a substance that does not dissolve in pharmaceutically acceptable water that controls the release of the drug. The hydrophobic compound usable in the present invention is, for example, at least one selected from the group consisting of fatty acids and fatty acid esters, fatty alcohols, waxes, inorganic substances, and mixtures thereof. Wherein the fatty acids and fatty acid esters are selected from glyceryl palmitostearate, glyceryl stearate, glyceryl distearate, glyceryl bihenate, cetyl palmitate, glyceryl monooleate, stearic acid and mixtures thereof Species or more; Fatty acid alcohols are at least one selected from cetostearyl alcohol, cetyl alcohol, stearyl alcohol and mixtures thereof; The waxes are at least one selected from carnauba wax, beeswax, microcrystalline wax, and mixtures thereof; The inorganic material is preferably at least one selected from talc, precipitated calcium carbonate, calcium dihydrogen phosphate, zinc oxide, titanium oxide, kaolin, bentonite, montmorillonite, bum and mixtures thereof. A more preferred example of the hydrophobic compound is carnauba wax. Here, the hydrophobic compound may be included in an amount of 0.1 parts by weight to 20 parts by weight, preferably 0.5 parts by weight to 10 parts by weight with respect to 1 part by weight of the active ingredient, and when less than 0.1 parts by weight, the release of the drug may not be controlled. In case of more than 20 parts by weight, excessive elution may be delayed.
상기 친수성 고분자는 약물의 방출을 제어하는 약제학적으로 허용가능한 물에 용해되는 고분자 물질을 말한다. 본 발명에서 사용가능한 친수성 고분자의 예는 당류, 셀룰로오스 유도체, 검류, 단백질류, 폴리비닐 유도체, 친수성 폴리메타크릴레이트 공중합체, 폴리에틸렌 유도체, 카르복시비닐공중합체, 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상이다. 여기서 당류는 덱스트린, 폴리덱스트린, 덱스트란, 펙틴 및 펙틴 유도체, 알긴산염, 폴리갈락투론산, 자일란, 아라비노자일란, 아라비노갈락탄, 전분, 히드록시프로필스타치, 아밀로오스, 아밀로펙틴, 및 이들의 혼합물 중에서 선택된 1종 이상; 셀룰로오스 유도체는 히드록시프로필메틸셀룰로오스(또는 히프로멜로오스라 함), 히드록시프로필셀룰로오스, 히드록시메틸셀룰로오스, 히드록시에틸셀룰로오스, 메틸셀룰로오스, 카르복시메틸셀룰로오스 나트륨, 히드록시프로필 메틸셀룰로오스 아세테이트 숙시네이트, 히드록시에틸메틸셀룰로오스, 및 이들의 혼합물 중에서 선택된 1종 이상; 검류는 구아검, 로커스트 콩 검, 트라가칸타, 카라기난, 아카시아검, 아라비아검, 젤란검, 잔탄검, 및 이들의 혼합물 중에서 선택된 1종 이상; 단백질류는 젤라틴, 카제인, 제인, 및 이들의 혼합물 중에서 선택된 1종 이상; 폴리비닐 유도체는 폴리비닐 알코올, 폴리비닐 피롤리돈, 폴리비닐아세탈디에틸아미노아세테이트, 및 이들의 혼합물 중에서 선택된 1종 이상; 친수성 폴리메타크릴레이트 공중합체는 폴리(부틸 메타크릴레이트, (2-디메틸아미노에틸)메타크릴레이트-메틸메타크릴레이트) 공중합체(예컨대, 유드라짓 E, 에보닉, 독일), 폴리(메타크릴산-메틸메타크릴레이트) 공중합체, 폴리(메타크릴산-에틸아크릴레이트) 공중합체 및 이들의 혼합물 중에서 선택된 1종 이상; 폴리에틸렌 유도체는 폴리에틸렌 글리콜, 폴리에틸렌 옥사이드 및 이들의 혼합물 중에서 선택된 1종 이상이며; 카르복시비닐공중합체는 카보머인 것이 바람직하다. 상기 친수성 고분자의 더욱 바람직한 예는 폴리비닐 피롤리돈, 히드록시프로필셀룰로오스, 히프로멜로오스, 폴리(에틸 아크릴레이트-메틸 메타크릴레이드-트리에틸아미노에틸- 메타크릴레이트 클로라이드) 공중합체 중에서 선택된 1종 이상이다. The hydrophilic polymer refers to a polymeric material that is dissolved in pharmaceutically acceptable water that controls the release of the drug. Examples of hydrophilic polymers usable in the present invention include 1 selected from the group consisting of sugars, cellulose derivatives, gums, proteins, polyvinyl derivatives, hydrophilic polymethacrylate copolymers, polyethylene derivatives, carboxyvinyl copolymers, and mixtures thereof. More than species. Wherein the sugars are dextrins, polydextrins, dextran, pectin and pectin derivatives, alginates, polygalacturonic acids, xylans, arabinoxylans, arabinogalactan, starch, hydroxypropylstarches, amylose, amylopectin, and their At least one selected from mixtures; Cellulose derivatives include hydroxypropylmethylcellulose (or hypromellose), hydroxypropylcellulose, hydroxymethylcellulose, hydroxyethylcellulose, methylcellulose, carboxymethylcellulose sodium, hydroxypropyl methylcellulose acetate succinate, At least one selected from hydroxyethyl methyl cellulose, and mixtures thereof; The gum is at least one selected from guar gum, locust bean gum, tragacanta, carrageenan, acacia gum, arabic gum, gellan gum, xanthan gum, and mixtures thereof; Proteins may be at least one selected from gelatin, casein, zein, and mixtures thereof; The polyvinyl derivative is at least one selected from polyvinyl alcohol, polyvinyl pyrrolidone, polyvinyl acetal diethylamino acetate, and mixtures thereof; Hydrophilic polymethacrylate copolymers include poly (butyl methacrylate, (2-dimethylaminoethyl) methacrylate-methylmethacrylate) copolymers (eg Eudragit E, Evonik, Germany), poly (meth At least one selected from methacrylic acid-methylmethacrylate) copolymer, poly (methacrylic acid-ethylacrylate) copolymer, and mixtures thereof; The polyethylene derivative is at least one selected from polyethylene glycol, polyethylene oxide and mixtures thereof; The carboxyvinyl copolymer is preferably a carbomer. More preferred examples of the hydrophilic polymer are selected from polyvinyl pyrrolidone, hydroxypropyl cellulose, hypromellose, and poly (ethyl acrylate-methyl methacrylate-triethylaminoethyl- methacrylate chloride) copolymer. More than species.
여기서, 친수성 고분자는 활성성분 1 중량부에 대해서 0.05 중량부 ~ 30 중량부, 바람직하게는 0.5~20 중량부로 포함될 수 있으며, 0.05 중량부 미만인 경우에는 방출속도가 조절되지 않을 염려가 있고, 30 중량부 초과인 경우에는 과도하게 용출이 지연될 염려가 있다. Here, the hydrophilic polymer may be included in an amount of 0.05 parts by weight to 30 parts by weight, preferably 0.5 to 20 parts by weight with respect to 1 part by weight of the active ingredient, and when it is less than 0.05 parts by weight, the release rate may not be controlled. If the excess is excessive, there is a fear that excessive dissolution is delayed.
(2-2) 삼투압 조절제 및 반투과성막 코팅기제 (2-2) Osmotic pressure regulator and semipermeable membrane coating base
본 발명의 지연방출성 구획은 삼투압 조절제를 포함하며, 반투과성막 코팅기제로 코팅된 구획일 수 있다. The delayed-release compartment of the present invention includes an osmotic pressure regulator and may be a compartment coated with a semipermeable membrane coating base.
상기 삼투압조절제는 황산마그네슘, 염화마그네슘, 염화나트륨, 염화리튬, 황산칼륨, 황산리튬, 황산나트륨 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상이 바람직하다. The osmotic pressure regulator is preferably at least one selected from the group consisting of magnesium sulfate, magnesium chloride, sodium chloride, lithium chloride, potassium sulfate, lithium sulfate, sodium sulfate and mixtures thereof.
여기서, 삼투압 조절제는 활성성분 1 중량부에 대해서 0.01 중량부 ~ 10 중량부, 바람직하게는 0.05 중량부~ 0.5 중량부로 포함될 수 있으며, 0.01 중량부 미만에서 충분한 시간차 방출성을 얻기 어려울 염려가 있고, 10 중량부 초과에서 약물방출이 지연되어 유의성 있는 임상적 효과를 얻기 어려울 염려가 있다. Here, the osmotic pressure control agent may be included in an amount of 0.01 parts by weight to 10 parts by weight, preferably 0.05 parts by weight to 0.5 parts by weight with respect to 1 part by weight of the active ingredient, and there is a concern that it is difficult to obtain sufficient time difference release property at less than 0.01 parts by weight, Drug release may be delayed at more than 10 parts by weight, making it difficult to obtain a significant clinical effect.
상기 반투과성막 코팅기제는 약제학적 제제의 코팅층에 배합되는 물질로서, 일부 성분은 통과시키지만 다른 성분은 통과시키지 않는 막을 형성하는데 사용되는 물질을 말한다. 본 발명에서 반투과성막 코팅기제는 상기 언급된 수불용성 중합체를 사용할 수 있다. The semi-permeable membrane coating base is a substance to be blended into the coating layer of the pharmaceutical formulation, and refers to a substance used to form a membrane that allows some components to pass but does not pass other components. In the present invention, the semipermeable membrane coating base may use the above-mentioned water-insoluble polymer.
본 발명에서 반투과성막 코팅기제는 예컨대 폴리비닐 아세테이트, 폴리메타크릴레이트 공중합체, 폴리(에틸아크릴레이트, 메틸 메타크릴레이트) 공중합체, 폴리(에틸아크릴레이트, 메틸 메타크릴레이트, 트리메틸아미노에틸메타크릴레이트 클로라이드)공중합체, 에틸셀룰로오스, 셀룰로오스 에스테르, 셀룰로오스 에테르, 셀룰로오스 아실레이트, 셀룰로오스 디아실레이트, 셀룰로오스 트리아실레이트, 셀룰로오스 아세테이트, 셀룰로오스 디아세테이트, 셀룰로오스 트리아세테이트 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상을 들 수 있다. The semipermeable membrane coating base in the present invention is, for example, polyvinyl acetate, polymethacrylate copolymer, poly (ethylacrylate, methyl methacrylate) copolymer, poly (ethylacrylate, methyl methacrylate, trimethylaminoethylmethacrylate Late chloride) copolymer, ethyl cellulose, cellulose ester, cellulose ether, cellulose acylate, cellulose dicylate, cellulose triacylate, cellulose acetate, cellulose diacetate, cellulose triacetate and mixtures thereof The above is mentioned.
여기서, 반투과성막 코팅기제는 활성성분 1 중량부에 대해서 0.01 중량부 ~ 10 중량부, 바람직하게는 0.05 중량부 ~ 1.25 중량부로 포함될 수 있으며, 0.01 중량부 미만인 경우에는 충분한 지연 시간을 갖기 어려울 염려가 있고, 10 중량부 초과인 경우에는 약물의 방출이 일어나지 않거나 지연시간이 길어지는 문제점이 있다. Here, the semi-permeable membrane coating base may be included in an amount of 0.01 parts by weight to 10 parts by weight, preferably 0.05 parts by weight to 1.25 parts by weight, with respect to 1 part by weight of the active ingredient, and when it is less than 0.01 parts by weight, it may be difficult to have a sufficient delay time. And, if more than 10 parts by weight there is a problem that the release of the drug does not occur or the delay time is long.
(3) 약제학적으로 허용가능한 첨가제 (3) pharmaceutically acceptable additives
본 발명의 제제는 본 발명의 효과를 해치지 않는 범위 안에서 약학적으로 허용 가능한 (2-1) 방출제어물질 및 (2-2) 삼투압 조절제 및 반투과성막 코팅기제로 언급한 것 이외의 희석제, 결합제, 붕해제, 윤활제, pH 조절제, 소포제, 용해보조제 등의 통상적으로 사용되는 첨가제를 지연방출성의 성격을 벗어나지 않는 범위 내에서 추가로 사용하여 제제화할 수 있다. The formulations of the present invention are diluents, binders, and borates other than those mentioned as pharmaceutically acceptable (2-1) release controlling substances and (2-2) osmotic pressure regulators and semipermeable membrane coating agents within the scope of not impairing the effects of the present invention. Commonly used additives such as release, lubricants, pH adjusters, antifoams, dissolution aids and the like can be formulated further using within a range not departing from the nature of delayed release.
상기 첨가제에 관한 상세한 내용은 상기 선방출성 구획에서 상술한 내용과 동일하다. Details of the additives are the same as those described above in the above-release compartment.
본 발명의 약제학적 제제는 다양한 제형으로 제조할 수 있으며, 예를 들어 나정, 코팅정, 다층정, 또는 유핵정 등의 정제, 분말제, 과립제, 또는 캡슐제 등으로 제형화할 수 있다. The pharmaceutical preparations of the present invention can be prepared in a variety of formulations and can be formulated, for example, in tablets, powders, granules, capsules, and the like, such as uncoated tablets, coated tablets, multilayer tablets, or nucleated tablets.
본 발명의 제제는 선방출성 구획을 이루는 과립 등과 지연방출성 구획을 이루는 과립 등에 선택적으로 첨가제를 후혼합하여 타정한 것으로 단일 정제 내에 선방출성 구획과 지연방출성 구획을 갖게 되어 각각의 구획의 활성성분이 별도로 용출하게 되어 각각의 약효를 나타내게 되는 나정 형태일 수 있다. The formulation of the present invention is a tabletting by selectively mixing additives such as granules constituting the pre-release compartment and granules constituting the delayed-release compartment and the like to have a pre-release compartment and a delayed-release compartment in a single tablet, and thus the active ingredient of each compartment. This may be in the form of uncoated tablets will be eluted separately to show the respective effects.
본 발명의 제제는 지연방출성 구획 및 선방출성 구획이 균일하게 혼합된 후 타정하여 얻어지는 2상의 매트릭스 정제 형태일 수 있다. The formulation of the present invention may be in the form of a two-phase matrix tablet obtained by tableting after the delayed-release compartment and the prior-release compartment are uniformly mixed.
또한, 본 발명의 약제학적 제제는 지연방출성 구획으로 이루어진 정제와 상기 정제의 외부를 둘러싸는 선방출성 구획으로 이루어진 필름코팅층으로 구성된 필름코팅정 형태일 수 있으며, 필름코팅층이 용해됨에 따라 필름코팅층의 활성성분이 먼저 용출되게 된다. In addition, the pharmaceutical formulation of the present invention may be in the form of a film coated tablet consisting of a tablet consisting of a delayed-release compartment and a film coating layer consisting of a pre-release compartment surrounding the outside of the tablet, the film coating layer of the film coating layer as it is dissolved The active ingredient is eluted first.
또한, 본 발명의 약제학적 제제는 지연방출성 구획과 선방출성 구획을 구성하는 과립물에 약제학적인 첨가제를 혼합하고, 다중 타정기를 사용하여 2중정 혹은 3중정으로 타정하여 얻어진, 지연방출성 구획과 선방출성 구획이 층을 이루는 다층구조의 다층정 형태일 수 있다. 이 제제는 층별로 선방출과 지연방출이 가능하도록 제제화된 경구 투여용 정제이다. In addition, the pharmaceutical formulation of the present invention is a delayed-release compartment, obtained by mixing the pharmaceutical additives in the granules constituting the delayed-release compartment and the prior-release compartment, and tableting in a double or triple tablet using a multiple tableting machine and The pre-emitting compartments may be in the form of a multi-layered tablet with a layered structure. This formulation is a tablet for oral administration which is formulated to enable pre-release and delayed release in layers.
또한, 본 발명의 약제학적제제는 지연방출성 구획으로 이루어진 내핵정과 상기 내핵정의 외면을 둘러싸고 있는 선방출성 구획으로 이루어진 외층으로 구성된 유핵정 형태일 수 있다. 상기 유핵정은 삼투성 유핵정일 수 있으며, 상기 삼투성 유핵정은 지연방출을 위해 삼투압조절제를 정제의 내부에 함유하게 하여 타정한 후, 반투과성막 코팅기제로 정제의 표면을 코팅하여 이를 내핵으로 하고, 선방출성 구획을 구성하는 과립물을 약제학적인 첨가제와 혼합한 뒤 외층으로 하여 타정함으로써 지연방출성의 내핵을 갖고 상기 내핵의 표면을 선방출층이 둘러싼 형태의 제형이다. In addition, the pharmaceutical formulation of the present invention may be in the form of a nucleus tablet consisting of an inner layer consisting of a delayed-release compartment and an outer layer consisting of a prior-release compartment surrounding the outer surface of the inner core tablet. The nucleated tablet may be an osmotic nucleated tablet, and the osmotic nucleated tablet contains an osmotic pressure-controlling agent inside the tablet for delayed release, followed by compression, and then coated the surface of the tablet with a semipermeable membrane coating agent to form an inner core. It is a dosage form in which the granules constituting the pre-release compartment are mixed with pharmaceutical additives and compressed into an outer layer to have a delayed-release inner core and the surface of the inner core is surrounded by a pre-release layer.
본 발명의 약제학적 제제는 지연방출성 구획으로 이루어진 입자, 과립, 펠렛, 또는 정제와, 선방출성 구획으로 이루어진 입자, 과립, 펠렛, 또는 정제를 포함하는 캡슐제 형태일 수 있다. Pharmaceutical formulations of the present invention may be in the form of capsules comprising particles, granules, pellets, or tablets consisting of delayed-release compartments, and particles, granules, pellets, or tablets consisting of prior release compartments.
상기 캡슐제의 지연방출성 구획으로 이루어진 정제는 삼투압 조절제를 정제 내부에 포함하고, 정제의 표면에 반투과성막 코팅기제를 갖는 삼투성 코팅정일 수 있다. The tablet consisting of the delayed-release compartment of the capsule may include an osmotic pressure-controlling agent within the tablet and an osmotic coated tablet having a semipermeable membrane coating base on the surface of the tablet.
상기 캡슐제의 기제는 젤라틴, 숙시네이트 젤라틴, 또는 히드록시프로필메틸셀룰로오스, 그 혼합물 중에서 선택된 하나일 수 있다. The base of the capsule may be one selected from gelatin, succinate gelatin, or hydroxypropylmethylcellulose, or a mixture thereof.
또한 본 발명의 약제학적 제제는 지연방출성 구획, 및 선방출성 구획을 포함하는 키트 형태일 수 있으며, 구체적으로, 상기 키트는 (a) 선방출성 구획 (b) 지연방출성 구획 및 (c)상기 선방출성 구획 및 지연방출성 구획을 충진하기 위한 용기로 이루어진 것일 수 있다. 상기 키트는 선방출성 구획을 구성하는 입자, 과립물, 펠렛, 또는 정제를 제조하고, 지연방출성 구획을 구성하는 과립물, 펠렛 또는 정제를 별도로 제조하여, 호일, 블리스터, 병 등에 같이 충전하여 동시에 복용이 가능한 형태로 제조할 수 있다. In addition, the pharmaceutical formulation of the present invention may be in the form of a kit comprising a delayed-release compartment, and a prior-release compartment, specifically, the kit comprises (a) a prior-release compartment (b) a delayed-release compartment and (c) the It may consist of a container for filling the pre-release compartment and the delayed-release compartment. The kit prepares the particles, granules, pellets, or tablets constituting the prerelease compartment, and separately prepares the granules, pellets, or tablets constituting the delayed release compartment, and fills them together with foil, blisters, bottles, and the like. It can be prepared in a form that can be taken at the same time.
본 발명의 제제는 지연방출성 구획 및/또는 선방출성 구획의 외부에 코팅층을 추가로 형성할 수 있다. 즉 지연방출성 구획 및/또는 선방출성 구획으로 이루어진 입자, 과립, 펠렛, 또는 정제 등의 표면에 방출제어 또는 제제 안정을 위한 목적으로 코팅을 할 수 있다. The formulations of the present invention may further form a coating layer on the exterior of the delayed release compartment and / or the prior release compartment. In other words, the surface of the particles, granules, pellets, or tablets consisting of delayed-release compartments and / or pre-release compartments may be coated for the purpose of release control or formulation stability.
또한, 본 발명에 따른 제제는, 추가의 코팅이 없는 나정 등의 상태로도 제공되지만, 필요에 따라 상기 제제의 외부에 코팅층을 형성시켜, 코팅층을 추가로 포함하는 코팅정 형태의 제제일 수 있다.  코팅층을 형성함으로써, 활성성분의 안정성을 더욱 확보할 수 있다.In addition, the formulation according to the present invention is also provided in a state such as uncoated tablets without additional coating, but may be in the form of a coated tablet containing a coating layer further by forming a coating layer on the outside of the formulation, if necessary. . By forming the coating layer, it is possible to further ensure the stability of the active ingredient.
코팅층을 형성하는 방법은 정제층의 표면에 필름상의 코팅층을 형성할 수 있는 방법 중에서 당업자의 선택에 의하여 적절히 선택할 수 있으며, 유동층 코팅법, 팬 코팅법 등의 방법을 적용할 수 있으며, 바람직하게는 팬 코팅법을 적용할 수 있다. The method of forming the coating layer may be appropriately selected by a person skilled in the art from the method of forming a film-like coating layer on the surface of the tablet layer, a method such as a fluidized bed coating method, a fan coating method may be applied, and preferably Fan coating can be applied.
코팅층은 피막제, 피막 보조제 또는 이들의 혼합물을 사용하여 형성할 수 있으며, 예를 들어, 피막제는 히드록시프로필메틸셀룰로오스, 히드록시프로필셀룰로오스 등과 같은 셀룰로오스 유도체, 당 유도체, 폴리비닐 유도체, 왁스류, 지방류, 젤라틴, 또는 이들의 혼합물 등을 사용할 수 있고, 피막 보조제는 폴리에틸렌글리콜, 에틸셀룰로오스, 글리세라이드류, 산화티탄, 탈크, 디에틸프탈레이트, 트리에틸 시트레이트 또는 이들의 혼합물 등을 사용할 수 있다. The coating layer may be formed by using a coating agent, a coating aid, or a mixture thereof. For example, the coating agent may be a cellulose derivative such as hydroxypropylmethylcellulose, hydroxypropylcellulose, sugar derivatives, polyvinyl derivatives, waxes, or fatty acids. , Gelatin, or a mixture thereof, and the like, and a coating aid may be polyethylene glycol, ethyl cellulose, glycerides, titanium oxide, talc, diethyl phthalate, triethyl citrate or a mixture thereof.
코팅층은 정제 총 중량에 대하여 0.5~ 15 중량퍼센트 (% w/w) 범위로 포함될 수 있다. The coating layer may be included in the range of 0.5 to 15% by weight (% w / w) based on the total weight of the tablet.
0.5중량% 미만인 경우 제품의 보호와 제형에 따라 안정성의 확보가 어려울 수 있으며, 15중량% 초과하는 경우에는 활성성분의 방출양상에 영향을 미칠 우려가 있다.If it is less than 0.5% by weight, it may be difficult to secure stability depending on the protection and formulation of the product, and when it is more than 15% by weight, it may affect the release pattern of the active ingredient.
본 발명의 제제는 저녁투여용으로, 1일 1회 저녁 5시 내지 11시 사이에 투여하여 24시간 균등하게 항압작용과 합병증 예방 작용을 발휘할 수 있다.The formulation of the present invention can be administered once a day between 5 to 11 o'clock in the evening, and can exert anti-pressure action and complication prevention evenly for 24 hours.
베타 아드레날린 차단제는 새벽 이후 항압 작용이 강한 것으로 알려져 있고[N EnglJ Med 2003; 348; 2377-8], [Chronology International 2006; 23(4): 813-829], 안지오텐신-2 수용체 길항제는 한밤중 이후 새벽까지의 혈압 강하 작용이 강한 것으로 알려져 있으므로[J. Hypertens, 2005; 23: 1913-1922],[Hypertension, 2003; 42: 283-290], [Chronobiol. Int. 2005; 22: 755-776], 본 발명의 제제를 저녁시간대에 한번 복용하여도, 선방출 되는 안지오텐신-2 수용체 길항제에 의하여 새벽까지 혈압을 효과적으로 강하하고, 지연방출되는 베타 아드레날린 차단제에 의해 새벽 이후의 혈압을 효과적으로 강하하게 되어 24시간 균등한 항압작용과 합병증 예방 작용을 발휘하게 된다.Beta adrenergic blockers are known to have strong antitumor activity after dawn [N EnglJ Med 2003; 348; 2377-8, Chronology International 2006; 23 (4): 813-829], angiotensin-2 receptor antagonists are known to have strong blood pressure lowering effects from midnight to dawn [J. Hypertens, 2005; 23: 1913-1922, Hypertension, 2003; 42: 283-290, Chronobiol. Int. 2005; 22: 755-776], even when the preparation of the present invention is taken once in the evening, the blood pressure is effectively lowered until dawn by an angiotensin-2 receptor antagonist, which is released, and the blood pressure after dawn by a beta-adrenergic blocker delayed release. It effectively lowers the 24 hours even anti-pressure action and prevent complications.
본 발명의 제제를 저녁시간대에 투여함으로써, 특히 합병증 발생 위험시간(새벽부터 오전 시간) 대에 혈압을 균등하게 유지시켜 줄 수 있으므로, 심장의 교감계 과잉 흥분 상태를 24시간 균등하게 억제해 주어야 하는 합병증을 지닌 고혈압환자에게 유용하다. 또한, 일반 고혈압 환자와 달리 수면 중 혈압이 저하되지 않는 유형의 고혈압인 Non-dipper형 고혈압의 억제에 본 발명의 제제는 유용하다. 상기 Non-dipper형 고혈압은 노인, 당뇨병자, 심장비대자 등에서 나타나며, 뇌졸중과 같은 합병증 위험이 높은 고혈압으로, 본 발명의 제제를 저녁시간대에 복용하여 수면 중 혈압억제에 유용하다. By administering the preparation of the present invention in the evening time, blood pressure can be maintained evenly in the time of risk of complications (from dawn to morning time). Therefore, the sympathetic hyper-excited state of the heart should be suppressed evenly for 24 hours. It is useful for patients with high blood pressure with complications. In addition, unlike the general hypertension patients, the formulation of the present invention is useful for suppressing non-dipper type hypertension, a type of hypertension in which blood pressure does not decrease during sleep. The non-dipper type hypertension is present in the elderly, diabetics, cardiac hypertrophy, and high blood pressure with a high risk of complications such as stroke, and is useful for suppressing blood pressure during sleep by taking the preparation of the present invention in the evening.
또한, 본 발명의 제제는 안지오텐신-2 수용체 길항체가 먼저 간에 유입되어 간에서 사이토크롬 P450 효소에 의해 활성화되고 난 후, 베타 아드레날린 차단제가 간을 통과하게 되므로 베타 아드레날린 차단제와 안지오텐신-2 수용체 길항제 간에 발생할 수 있는 대사적인 상호작용을 차단하게 되어 약물간 상호작용에 따른 부작용을 감소시켜 준다. In addition, the agent of the present invention is an angiotensin-2 receptor antagonist is first introduced into the liver is activated by the cytochrome P450 enzyme in the liver, after the beta adrenergic blocker passes through the liver between the beta adrenergic blocker and the angiotensin-2 receptor antagonist Blocks metabolic interactions that may occur, reducing side effects of drug interactions.
또한, 본 발명은 본 발명의 제제를 인간을 포함하는 포유류에게 투여하는 단계를 포함하는 심혈관계 질환 치료방법을 제공한다. The present invention also provides a method for treating cardiovascular disease, comprising administering the agent of the present invention to a mammal including a human.
본 발명의 제제를 하루에 한번 특히 저녁시간대(17 ~ 23시)에 투여하는 치료방법에 의해, 24시간 균등하게 항압작용과 합병증 예방 작용을 발휘할 수 있으므로, 심혈관계 질환을 치료할 수 있다. By the treatment method of administering the preparation of the present invention once a day, especially in the evening (17 to 23 o'clock), since it can exert an anti-pressure action and a complication prevention effect evenly for 24 hours, cardiovascular diseases can be treated.
상기 심혈관계 질환은 고혈압, 또는 고혈압 합병증 등을 포함하는 의미이다. The cardiovascular disease is meant to include hypertension, or high blood pressure complications.
본 발명의 약제학적 제제는 당 분야의 적절한 방법으로, 예를 들어 Chronotherapeutics(2003, Peter Redfern, PhP), Remington's Pharmaceutical Science(최근판), Mack Publishing Company, Easton PA 등에 개시되어 있는 방법을 참조하여 각 질환에 따라 또는 성분에 따라 바람직하게 제제화 할 수 있으며, 구체적으로 이하의 단계를 포함하는 방법에 의해 제조될 수 있다. Pharmaceutical formulations of the present invention may be prepared by any suitable method in the art, for example, with reference to methods disclosed in Chronotherapeutics (2003, Peter Redfern, PhP), Remington's Pharmaceutical Science (Recent Edition), Mack Publishing Company, Easton PA, and the like. It can be formulated preferably according to a disease or a component, and can be manufactured by the method containing the following steps specifically ,.
제 1 단계는 지연방출성 구획의 활성성분을 장용성 고분자, 수불용성 중합체, 소수성 화합물, 친수성 고분자 중에서 선택된 방출제어물질 1종 또는 2종과 약제학적으로 사용되는 통상의 첨가제를 투여하여 혼합, 연합, 건조, 제립 또는 코팅, 및 타정을 통해 지연방출성 과립 또는 정제를 얻거나, 상기 활성성분을 삼투압조절제와 약제학적으로 사용되는 통상의 첨가제를 투여하여 혼합, 연합, 건조, 제립 또는 타정한 후 반투과성막 코팅기제로 코팅 하여 지연방출성 과립 또는 정제를 얻는 단계이다. In the first step, the active ingredient of the delayed-release compartment is mixed with, or combined with, one or two release controlling substances selected from an enteric polymer, a water insoluble polymer, a hydrophobic compound, and a hydrophilic polymer, and a conventional additive used in pharmaceuticals. Delayed-release granules or tablets are obtained through drying, granulation or coating, and tableting, or the active ingredient is semipermeable after mixing, associating, drying, granulating or tableting by administering an osmotic agent and a conventional additive which is used pharmaceutically. It is a step of obtaining delayed-release granules or tablets by coating with a membrane coating base.
제 2 단계는 선방출성 구획의 활성성분과 약제학적으로 허용되는 통상의 첨가제를 투여하여 혼합, 연합, 건조, 제립 혹은 코팅, 및 타정을 통해 경구 고형제를 생산하기 위한 통상의 과정을 통하여 얻어진 선방출성 과립 또는 정제를 얻는 단계이다. The second step involves the administration of the active ingredient of the prior-release compartment and the conventionally acceptable pharmaceutically acceptable additives to produce the oral solids through mixing, coalescing, drying, granulating or coating, and tableting to produce oral solids. Obtaining extruded granules or tablets.
제 3 단계는 상기 제 1 단계 및 제 2 단계에서 얻어진 각각의 과립 혹은 정제를 약제학적인 부형제와 혼합하여 타정 또는 충전하여 경구 투여용 제제를 얻는 단계이다. In the third step, the granules or tablets obtained in the first step and the second step are mixed with pharmaceutical excipients, tableted or filled to obtain a preparation for oral administration.
상기 제 1 단계와 상기 제 2 단계는 순서를 바꾸거나, 동시에 실시할 수 있다. The first step and the second step may be reversed or executed simultaneously.
상기 과정에 의하여 본 발명의 약제학적 제제가 제조될 수 있으며, 제3단계의 제제화 방법을 보다 상세하게 설명하면 다음과 같으나, 이에 한정되는 것은 아니다. The pharmaceutical formulation of the present invention may be prepared by the above process, and the formulation method of the third step is described in more detail as follows, but is not limited thereto.
[가] 2상의 매트릭스 정제의 제조 [A] Preparation of two-phase matrix tablet
제 1 단계에서 얻어진 입자 또는 과립을 그대로 또는 방출제어물질로 추가 코팅한 후, 제 2 단계에서 제조한 과립과 혼합하여 일정량의 무게로 타정하여 정제를 제조한다. 얻어진 정제를 안정성 또는 성상 개선의 목적으로 필요에 따라 필름 코팅을 할 수 있다. The particles or granules obtained in the first step are further coated as they are or with a release controlling material, and then mixed with the granules prepared in the second step and compressed into a certain amount of weight to prepare a tablet. The obtained tablet can be film coated as necessary for the purpose of improving stability or property.
[나] 활성성분을 함유한 필름코팅정의 제조 [B] Preparation of Film-Coated Tablets Containing Active Ingredients
제 1 단계에서 얻어진 코팅정 또는 과립을 그대로 또는 방출제어물질로 추가 코팅하고 건조한 후 일정량으로 타정하여 그대로 혹은 추가로 코팅하여 정제를 제조한 후, 별도로 선방출성 구획의 활성성분을 수용성의 필름코팅용액에 용해, 분산시켜 제 1 단계에서 얻은 정제 외층에 코팅함으로써 필름코팅에 활성성분을 함유한 경구투여형 필름코팅정제를 제조할 수 있다. The coated tablets or granules obtained in the first step are additionally coated as they are or with a release control material, dried, and then compressed into a predetermined amount to prepare tablets as they are or additionally coated, and then the active ingredients of the pre-release compartments are separately coated with a water-soluble film coating solution. By dissolving and dispersing in a coating, the tablet outer layer obtained in step 1 can be used to prepare an orally administered film coating tablet containing the active ingredient in a film coating.
[다] 다층정의 제조 Preparation of multi-layered tablets
제 1 단계에서 얻어진 과립을 그대로 또는 방출제어물질로 추가 코팅하고 건조하여 얻은 과립과 제 2 단계에서 얻어진 과립을 타정기를 이용하여 2중정으로 제조할 수 있다. 제형설계 또는 필요에 따라 방출 보조층을 추가하여3중 또는 그 이상의 다층정을 제조하거나 코팅하여 코팅 다층정을 제조할 수 있다. The granules obtained in the first step as they are or are additionally coated and dried with a release controlling substance and the granules obtained in the second step can be prepared in double tablets using a tablet press. Coated multi-layered tablets can be prepared by formulating or coating triple or more multi-layered tablets by adding a release aid layer as needed, or by formulation.
[라] 유핵정의 제조 [D] Preparation of Nucleated Tablets
제 1 단계에서 얻어진 코팅정 또는 과립을 그대로 또는 방출제어물질로 추가 코팅하고 건조한 후 일정량으로 타정하여 그대로 혹은 추가로 코팅을 하여 내핵으로 한 후, 제 2 단계에서 얻은 과립과 함께 유핵정타정기로 타정하여 내핵의 표면을 선방출층이 둘러싼 형태의 유핵정을 제조하거나 코팅하여 코팅 유핵정을 제조할 수 있다. The coated tablet or granules obtained in the first step are additionally coated as it is or with a release control material, dried, and then compressed into a predetermined amount to be coated as it is or additionally to the inner core, followed by a nucleated tableting machine together with the granules obtained in the second step. The coated nucleated tablet may be prepared by preparing or coating a nucleated tablet having a form in which a pre-release layer is enclosed on the surface of the inner core.
[마] 캡슐제(과립 또는 정제 함유)의 제조 [E] Preparation of Capsules (Granules or Tablets)
제 1 단계에서 얻어진 과립을 그대로 또는 방출제어물질로 추가 코팅하고 건조한 과립 또는 정제와, 제 2 단계에서 얻은 과립 또는 정제를 캡슐충전기에 넣고 일정 크기의 캡슐에 각 주성분 유효량 해당 량만큼 충전하여 캡슐제를 제조할 수 있다. The granules obtained in the first step are additionally coated as is or with a release controlling substance, and the dried granules or tablets and the granules or tablets obtained in the second step are placed in a capsule charger and filled into capsules of a predetermined size by an effective amount of each active ingredient in an appropriate amount. Can be prepared.
[바] 캡슐제(펠렛)의 제조 [Bar] Preparation of capsules (pellets)
(1) 지연방출성 구획의 활성성분과 방출제어물질, 필요에 따라 약제학적으로 허용 가능한 첨가제를 물, 유기용매, 또는 혼합용매에 용해시키거나 현탁시켜 설탕 구형과립에 코팅, 건조 후 필요에 따라 방출제어물질 단독 또는 2종 이상을 사용하여 물, 유기용매, 또는 혼합용매에 용해시킨 후 코팅, 건조한 후 제 2 단계에서 얻은 과립 또는 제 3 단계에서 얻은 정제와 혼합 후 캡슐충진기로 캡슐에 충전하여 캡슐제를 제조할 수 있다. (1) Dissolve or suspend the active ingredients in the delayed-release compartments, controlled release substances and, if necessary, pharmaceutically acceptable additives in water, organic solvents or mixed solvents, coating them on sugar granules, drying them as necessary. After dissolving in water, organic solvent or mixed solvent using release control material alone or two or more, coating, drying, mixing with granules obtained in the second step or tablets obtained in the third step, and then filling the capsule with a capsule filler Capsules can be prepared.
(2) 선방출성 구획의 활성성분과 제제학적으로 허용 가능한 첨가제를 물, 유기용매, 또는 혼합용매에 용해시키거나 현탁시켜 설탕 구형과립에 코팅, 건조 후 상기(1)의 지연방출성 구획의 활성성분을 함유한 방출제어 펠렛과 혼합 후 캡슐충진기로 캡슐에 충전하여 캡슐제를 제조할 수 있다. (2) Activity of delayed-release compartment as described in (1) above after coating and drying the sugar spherical granules by dissolving or suspending the active ingredient and pharmaceutically acceptable additive in the pre-release compartment in water, organic solvent or mixed solvent. Capsules may be prepared by mixing the release control pellets containing the ingredients and filling the capsules with a capsule filling machine.
[사] 키트의 제조 [Product] Kit Preparation
제 1 단계에서 얻어진 지연방출성 구획의 제제(베타 아드레날린 차단제 함유 제제)와, 제 2 단계에서 얻은 선방출성 구획의 제제(안지오텐신-2 수용체 길항제 함유 제제)를 호일, 블리스터, 병 등에 같이 충전하여 동시에 복용이 가능한 키트로 제조할 수 있다. The delayed-release compartment preparation (beta-adrenergic blocker-containing formulation) obtained in the first stage and the prior-release compartment preparation (angiotensin-2 receptor antagonist-containing formulation) obtained in the second stage are filled together with a foil, blister, bottle, etc. It can be made into a kit that can be taken at the same time.
본 발명 제제의 인체 투여량은 체내에서 활성성분의 흡수도, 불활성화율 및 배설속도, 환자의 연령, 성별 및 상태 등에 따라 적절히 선택되나, 일반적으로는 성인에게 베타 아드레날린 차단제와 안지오텐신-2 수용체 길항제의 합계량으로, 1일 2.0 ~ 2200 mg 투여하며, 바람직하게는 1일 42 ~ 1900mg을 투여하여 항압작용과 합병증 예방 작용을 발휘토록 할 수 있다. The human dosage of the formulation of the present invention is appropriately selected depending on the absorption rate, inactivation rate and excretion rate of the active ingredient in the body, the age, sex and condition of the patient, but in general, the beta adrenergic blocker and angiotensin-2 receptor antagonist in adults In a total amount, 2.0 to 2200 mg per day, preferably 42 to 1900 mg per day can be exerted anti-pressure action and prevent complications.
본 발명의 제제는 안지오텐신-2 수용체 길항제와 베타 아드레날린 차단제 두 성분 간의 시간차 용출을 가능하게 함으로써, 약물 대사 효소 상호 작용 이론과 시간차 투약 원리에 적합하도록 제제화한 것으로, 두 성분을 동시에 복용하여, 약물이 동시에 방출되는 경우에 비해 약효를 증가시키고 부작용을 감소시킬 수 있다. The formulation of the present invention is formulated to be compatible with the theory of drug metabolic enzyme interactions and the principle of time difference administration by allowing time difference dissolution between two components of angiotensin-2 receptor antagonist and beta adrenergic blocker. It can increase the efficacy and reduce the side effects compared with the simultaneous release.
즉, 본 발명의 제제는 복합 성분 각각의 체내 약리 작용 발현에 시간차 투약(Chronotherapy)원리와 약물의 체내 대사(Xenobiotics)원리를 적용하여 특정 속도로 체내에서 제어 방출하여 체내 흡수시 가장 이상적인 효과를 나타낼 수 있도록 설계된 약물 송달 시스템이다. In other words, the formulation of the present invention applies the principle of chronotherapy and the principle of Xenobiotics of the drug to the expression of pharmacological action of each of the complex components to control release in the body at a specific rate to achieve the most ideal effect upon absorption in the body. Is a drug delivery system designed to help.
본 발명의 약제학적 제제는 균등한 항압작용과 합병증 예방 작용을 나타내며, 특히 합병증 발생 위험시간대에 혈압을 균등하게 유지시켜 줄 수 있어 합병증을 지닌 고혈압 환자, 수면 중 혈압억제 등에 유용하고, 약물간 상호작용에 따른 부작용을 감소시켜 준다.The pharmaceutical preparation of the present invention exhibits an equal anti-pressure action and prevents complications, and is particularly useful for hypertension patients with complications, suppressing blood pressure during sleep, and the like. Reduces the side effects of action.
도 1은 실시예 2의 발사르탄-염산 네비볼롤 함유 제제에서 발사르탄과 염산 네비볼롤의 용출률을 대조약과 비교하여 나타낸 그래프이다. 1 is a graph showing the dissolution rate of valsartan and nebivolol in the valsartan-hydrochloride nebivolol-containing formulation of Example 2 compared with the reference drug.
도 2는 실시예 2, 3 4, 5의 제제에서 염산 네비볼롤의 용출률을 나타낸 그래프이다. 2 is a graph showing the dissolution rate of nebivolol hydrochloride in the formulation of Examples 2, 3 4, 5.
도 3은 실시예 14의 로사르탄-염산 네비볼롤 함유 제제에서 로사르탄과 염산 네비볼롤의 용출률을 대조약과 비교하여 나타낸 그래프이다. FIG. 3 is a graph showing the dissolution rate of losartan and nebivolol hydrochloride in the losartan-hydrochloride nebivolol-containing preparation of Example 14 compared with the reference drug.
도 4는 실시예 21, 22, 23, 24의 제제에서 염산 네비볼롤의 용출률을 대조약과 비교하여 나타낸 그래프이다. 4 is a graph showing the dissolution rate of nebivolol hydrochloride in the formulations of Examples 21, 22, 23, and 24 compared with the reference drug.
도 5는 실시예 21, 22, 23, 24의 제제에서 로사르탄의 용출률을 대조약과 비교하여 나타낸 그래프이다. 5 is a graph showing the dissolution rate of losartan in the formulations of Examples 21, 22, 23, and 24 compared with the reference drug.
도 6은 실시예 34의 칸데사르탄 실렉세틸-아테놀롤 함유 제제에서 칸데사르탄 실렉세틸, 아테놀롤의 용출률을 대조약과 비교하여 나타낸 그래프이다. 6 is a graph showing the dissolution rate of candesartan cilexetil and atenolol in the candesartan cilexetil-atenolol-containing formulation of Example 34 in comparison with the reference drug.
도 7은 실시예 36의 에프로사르탄-카르베딜롤 함유 제제에서 에프로사르탄, 카르베딜롤의 용출률을 대조약과 비교하여 나타낸 그래프이다. 7 is a graph showing the dissolution rate of eprosartan and carvedilol in the eprosartan-carvedilol-containing formulation of Example 36 in comparison with the reference drug.
도 8은 실시예 37의 텔미사르탄-카르베딜롤 함유 제제에서 텔미사르탄, 카르베딜롤의 용출률을 대조약과 비교하여 나타낸 그래프이다. 8 is a graph showing the dissolution rate of telmisartan and carvedilol in the telmisartan-carvedilol-containing formulation of Example 37 compared with the reference drug.
도 9는 실시예 39의 로사르탄-메토프롤롤 함유 제제에서 로사르탄, 메토프롤롤의 용출률을 대조약과 비교하여 나타낸 그래프이다. 9 is a graph showing the dissolution rate of losartan and metoprolol in the losartan-metoprolol-containing formulation of Example 39, compared with the reference drug.
본 발명의 이해를 돕기 위하여 실시예를 제시한다. 하기의 실시예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐, 실시예에 의해 본 발명의 내용이 한정되는 것은 아니다. Examples are provided to help understand the present invention. The following examples are merely provided to more easily understand the present invention, but the contents of the present invention are not limited by the examples.
<실시예 1>  염산 네비볼롤 - 발사르탄2상 매트릭스 정제 제조 Example 1 Preparation of Navivolol Hydrochloride-Valsartan Two-Phase Matrix Tablets
(1) 발사르탄 선방출성 과립 제조 (1) Preparation of Valsartan Pre-Release Granules
표 1에 나타난 성분 및 함량으로 발사르탄(Ranbaxy, India), 유당(Parmatose, DMV Pharma, Netherlands), 미결정셀룰로오스(AvicelPH, FMC Biopolymer, USA), 폴록사머188(Lutrol-F68, BASF, Germany), 전분글리콘산나트륨(Primojel, DMV Pharma, 네덜란드)을 달아 35 호체로 사과하였다. 별도로 히드록시프로필셀룰로오스(HPC-L, Nippon soda, Japan)를 정제수에 녹여 결합액을 제조하였다. 상기 혼합물을 유동층과립기 GPCG-1(Glatt, Germany)에 넣고 결합액을 가하여 조립하였다. 조립이 완료된 후 유동층 건조기 조립물을 건조시켰다. Valsartan (Ranbaxy, India), lactose (Parmatose, DMV Pharma, Netherlands), microcrystalline cellulose (AvicelPH, FMC Biopolymer, USA), poloxamer 188 (Lutrol-F68, BASF, Germany), starch Sodium glyconate (Primojel, DMV Pharma, The Netherlands) was weighed and apologized as No. 35. Separately, hydroxypropyl cellulose (HPC-L, Nippon soda, Japan) was dissolved in purified water to prepare a binding solution. The mixture was put into a fluid bed granulator GPCG-1 (Glatt, Germany) and granulated by the addition of a binder solution. The fluid bed dryer assembly was dried after assembly was complete.
건조가 완료되면 건조물을 20호체가 장착된 F형 정립기(KYK-60, 코리아메디, 한국)를 사용하여 정립하여, 발사르탄 선방출 과립을 제조하였다. When the drying was completed, the dried product was sized using an F-type sizer (KYK-60, Korea Medi, Korea) equipped with a No. 20 body, thereby preparing Valsartan pre-release granules.
(2) 염산 네비볼롤의 지연방출층 과립 제조 (2) Preparation of delayed-release layer granules of nebivolol hydrochloride
표 1에 나타난 성분 및 함량으로, 염산 네비볼롤(Cadila, India), 미결정셀룰로오스, 라우릴 황산나트륨(Texapon K12P, Cognis Corp, USA), 가교 폴리비닐피롤리돈(Crospovidone, BASF, Germany)을 35호체로 사과하고, 별도로 히드록시프로필셀룰로오스(HPC-L, Nippon soda, Japan)를 정제수에 녹여 결합액으로 하였다. 유동층 과립기 및 유동층 건조 등은 발사르탄 선방출 과립의 공정과 동일하다. 건조물을 유동층 코팅기에 넣고 별도로 셀룰로오스아세테이트(아세탈기 32%)(Eastman Chemical Company, USA), 셀룰로오스아세테이트(아세탈기 39.8%)(Eastman Chemical Company, USA), 히드록시프로필메틸셀룰로오스를 에탄올과 염화메틸렌에 녹인 액을 조제하여 상기 조립물을 코팅하였다. As the ingredients and contents shown in Table 1, hydrochloric acid nebivolol (Cadila, India), microcrystalline cellulose, sodium lauryl sulfate (Texapon K12P, Cognis Corp, USA), crosslinked polyvinylpyrrolidone (Crospovidone, BASF, Germany) No. 35 The apples were sieved, and hydroxypropyl cellulose (HPC-L, Nippon soda, Japan) was separately dissolved in purified water to obtain a binding solution. Fluidized bed granulators and fluidized bed drying are the same as those of valsartan pre-release granules. The dried material was placed in a fluidized bed coater and separately cellulose acetate (acetal group 32%) (Eastman Chemical Company, USA), cellulose acetate (acetal group 39.8%) (Eastman Chemical Company, USA), and hydroxypropylmethylcellulose in ethanol and methylene chloride The melted solution was prepared to coat the granulated product.
공정이 진행되는 동안에는 제품온도를 34 ~ 38 ℃ 유지시키고, 코팅이 완료되면 제품온도를 40 ℃ 유지하면서 약 1시간 정도 건조 및 표면 작업을 하여, 염산 네비볼롤 지연방출성 과립을 제조하였다. While the process is in progress, the product temperature is maintained at 34 ~ 38 ℃, when the coating is complete, while drying and surface work for about 1 hour while maintaining the product temperature 40 ℃, to produce a nebivolol delayed-release granules hydrochloric acid.
(3) 타정 및 코팅 (3) tableting and coating
상기 (1), (2)의 두 과립물을 혼합한 후, 스테아르산 마그네슘을 투입 후, 4분간 혼합하여 직경 10.0 mm 펀치가 장착된 로타리 타정기(MRC-33: 세종파마텍, 한국)에서 타정하였다. 타정이 완료된 정제를 히드록시프로필메틸셀룰로오스 2910(Shin-etsu, Japan), 폴리에틸렌글리콜 6000(BASF, Germany), 산화티탄(Tioside Americas, USA)을 에탄올과 정제수에 용해, 분산하여 제조한 코팅액을 사용하여 코팅하였다. After mixing the two granules of (1) and (2), magnesium stearate was added thereto, and then mixed for 4 minutes and compressed into tablets using a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 10.0 mm diameter punch. . Tablets that have been tableted are prepared by dissolving and dispersing hydroxypropylmethylcellulose 2910 (Shin-etsu, Japan), polyethylene glycol 6000 (BASF, Germany), and titanium oxide (Tioside Americas, USA) in ethanol and purified water. By coating.
<실시예 2> 염산 네비볼롤 - 발사르탄 이중정제 제조Example 2 Preparation of Navivolol Hydrochloride-Valsartan Double Tablet
표 1에 나타난 성분과 함량으로, 상기 실시예 1의 (2) 염산 네비볼롤의 지연방출층 과립에 스테아르산마그네슘을 투입 후, 최종 혼합하여 염산 네비볼롤 지연방출층을 제조한다.In the ingredients and contents shown in Table 1, magnesium stearate was added to the delayed-release layer granules of (2) nebivolol hydrochloride of Example 1, and finally mixed to prepare a delayed-release layer of nebivolol hydrochloride.
표 1에 나타난 성분과 함량으로, 상기 실시예 1의 (1) 발사르탄 선방출성 과립에 스테아르산마그네슘을 투입하고, 최종혼합하여 발사르탄 선방출층을 제조한다. 제조된 네비볼롤 지연방출층과 발사르탄 선방출층을 직경 10 mm 펀치가 장착된 로타리 다층정 타정기(MRC-37T: 세종파마텍, 한국)의 다른 과립 주입구에 각각 넣고 타정한 후, 타정이 완료된 정제를 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 산화티탄을 에탄올, 정제수에 용해 및 분산시켜 제조한 코팅액을 사용하여 코팅하였다. With the ingredients and contents shown in Table 1, magnesium stearate was added to the (1) valsartan pre-release granules of Example 1, and finally mixed to prepare a valsartan pre-release layer. The prepared Navivolol delayed-release layer and valsartan pre-release layer were put into different granule inlet holes of a rotary multi-layer tablet tablet press (MRC-37T: Sejong Pharmatech, Korea) equipped with a 10 mm diameter puncher, and then tableted. Hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were coated using a coating solution prepared by dissolving and dispersing in ethanol and purified water.
<실시예 3> 염산 네비볼롤 - 발사르탄 다층정제 제조 Example 3 Preparation of Navivolol Hydrochloride-Valsartan Multilayer Tablet
표 1에 나타난 성분과 함량으로 상기 실시예 1의 (2) 염산 네비볼롤의 지연방출층 과립에 스테아르산마그네슘을 투입 후, 최종 혼합한 혼합물을 중간층(2번째층)으로 쌓고, 실시예 1의 (1) 발사르탄 선방출성 과립에 스테아르산마그네슘을 투입하고, 최종혼합한 혼합물을 1층 및 3층으로 분할하여 직경 10 mm 펀치가 장착된 로타리 다층정 타정기(MRC-37T: 세종파마텍, 한국)의 다른 과립 주입구에 각각 넣고 타정한 후, 타정이 완료된 정제를 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 산화티탄을 에탄올, 정제수에 용해 및 분산시켜 제조한 코팅액으로 코팅하여 코팅정제를 제조하였다. After adding magnesium stearate to the delayed-release layer granules of (2) nebivolol hydrochloride of Example 1 according to the ingredients and contents shown in Table 1, the final mixed mixture was stacked as an intermediate layer (second layer), and (1) Magnesium stearate was injected into the valsartan pre-release granules, and the final mixed mixture was divided into one and three layers, and the rotary multi-layer tablet press machine (MRC-37T: Sejong Pharmatech, Korea) equipped with a 10 mm diameter punch was installed. After the tablets were put into different granules, and tableted, the tablets were tableted with hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide in a coating solution prepared by dissolving and dispersing ethanol in purified water.
<실시예 4> 염산 네비볼롤 - 발사르탄 유핵정제 제조 Example 4 Preparation of Navivolol Hydrochloride-Valsartan Nucleated Tablets
표 1에 나타난 성분 및 함량으로, 상기 실시예 1의 (2) 방법으로 제조한 염산 네비볼롤의 지연방출층 과립에 스테아르산마그네슘을 투입 후, 최종 혼합한 혼합물을 직경 6 mm 펀치가 장착된 로타리 타정기(MRC-33 : 세종파마텍, 한국)로 타정을 하여 내핵을 제조한 후 12 mm 펀치가 장착된 유핵정타정기(RUD-Ⅰ Killian, 독일)에서 상기 실시예 1(1)의 발사르탄 속방형 과립에 스테아르산마그네슘을 투입하여 혼합한 혼합물과 함께 타정하였다. 타정이 완료된 정제를 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 산화티탄을 에탄올, 정제수에 용해 및 분산시켜 제조한 코팅액으로 코팅하여 코팅정제를 제조하였다. After the magnesium stearate was added to the granules of delayed-release layer of nebivolol hydrochloride prepared by the method of Example 1 (2), the final mixture was rotary with a 6 mm diameter punch. The valsartan immediate release granules of Example 1 (1) in a nucleus tableting machine (RUD-I Killian, Germany) equipped with a 12 mm punch after tableting with a tableting machine (MRC-33: Sejong Pharmatech, Korea) Magnesium stearate was added to the mixture, and the mixture was compressed into tablets. The tablets having been tableted are coated with a coating solution prepared by dissolving and dispersing hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide in ethanol and purified water to prepare a coated tablet.
<실시예 5> 염산 네비볼롤 - 발사르탄 캡슐제(정제 + 정제)의 제조 Example 5 Preparation of Navivolol Hydrochloride-Valsartan Capsule (Tablet + Tablet)
(1) 발사르탄 선방출 과립 제조(1) valsartan pre-release granule manufacture
표 1에 나타난 성분 및 함량으로, 유당을 포함하지 않는 것을 제외하고는 상기 실시예 2의 방법에 따라 발사르탄 선방출 과립을 제조하였다.Valsartan pre-release granules were prepared according to the method of Example 2, except that the components and contents shown in Table 1 do not include lactose.
(2) 염산 네비볼롤 지연방출 과립 제조(2) Preparation of Nebivolol Delayed-Release Granules Hydrochloride
표 1에 나타난 성분 및 함량으로, 셀룰로오스 아세테이트 대신 프탈산히드록시프로필메틸셀룰로오스와 카보머를 사용하고, 가교폴리비닐피롤리돈을 사용하지 않은 것을 제외하고는 상기 실시예 2의 염산 네비볼롤 지연방출층 제조와 동일한 방법으로 제조하였다.As the components and contents shown in Table 1, except for using phthalic acid hydroxypropylmethyl cellulose and carbomer, and not crosslinked polyvinylpyrrolidone, the nebivolol delayed-release layer of Example 2 above It prepared by the same method as the preparation.
(3) 타정 및 캡슐 충진(3) tableting and capsule filling
발사르탄 선방출 과립을 직경 6mm 펀치가 장착된 로타리 타정기에서 타정하고, 염산 네비볼롤 지연방출 과립은 직경 5 mm 펀치가 장착된 로타리 타정기(MRC-33: 세종파마텍, 한국)에서 타정하였다. 타정이 완료된 두 정제를 캡슐충진기(SF-40N, 세종파마텍, 한국)로 1호 캡슐(HPMC 경질 캡슐, 서흥캅셀, 한국)에 충전하여 캡슐제를 제조하였다. The valsartan pre-release granules were compressed in a rotary tablet press equipped with a 6 mm diameter punch, and the hydrochloride nebivolol delayed-release granules were compressed in a rotary tablet press equipped with a 5 mm diameter punch (MRC-33: Sejong Pharmatech, Korea). Tableting was completed two tablets were filled capsule No. 1 capsule (SF-40N, Sejong Pharmatech, Korea) into No. 1 capsule (HPMC hard capsule, Seoheung capsule, Korea) to prepare a capsule.
<실시예 6> 염산 네비볼롤 - 발사르탄 캡슐제(정제 + 과립)의 제조 Example 6 Preparation of Nebivolol Hydrochloride-Chamvalsartan Capsule (Tablet + Granules)
표 1에 나타난 성분 및 함량으로, 실시예 5 중 염산 네비볼롤 지연방출형 정제 제조방법에 따라 염산 네비볼롤 지연방출형 정제를 제조하고, 실시예 2의 (1) 제조방법에 따라 발사르탄 선방출성 과립을 제조한 후, 각각의 정제와 과립을 캡슐충전기로 동일한 캡슐(0호 캡슐)에 충전하여 염산 네비볼롤 발사르탄 캡슐제(정제 + 과립)를 제조하였다. According to the ingredients and contents shown in Table 1, the nebivolol delayed-release tablet hydrochloride was prepared according to the preparation method of delayed-release tablet hydrochloride in Example 5, and the valsartan pre-release granules were prepared according to the preparation method of Example 2 (1). After the preparation, each tablet and granules were filled in the same capsule (No. 0 capsule) with a capsule charger to prepare a nebivolol valsartan hydrochloride (tablet + granules).
<실시예 7> 염산 네비볼롤 발사르탄 캡슐제(과립 + 과립)의 제조 Example 7 Preparation of Navivolol Chevalvalsartan Capsule (Granule + Granule) Hydrochloride
표 2에 나타난 성분 및 함량으로, 상기 실시예 2와 동일한 방법으로 제조한 염산 네비볼롤 지연방출 과립과 발사르탄 과립을 캡슐충전기로 0호 캡슐에 충전하여 캡슐제를 제조하였다. In the ingredients and contents shown in Table 2, the capsules were prepared by filling the No. 0 capsules with Navivolol delayed-release granules and valsartan granules prepared in the same manner as in Example 2 with a capsule charger.
<실시예 8> 염산 네비볼롤 - 발사르탄 캡슐제(펠렛+펠렛)의 제조 Example 8 Preparation of Navivolol Hydrochloride-Valsartan Capsule (Pellets + Pellets)
(1) 발사르탄 선방출 펠렛의 제조 (1) Preparation of Valsartan Prerelease Pellets
표 2에 나타난 성분 및 함량으로, 슈가 시드(Sugar sphere)를 유동층 과립기(GPCG 1: Glatt)에 투입한 뒤, 따로 정제수와 에탄올 혼합용매에 폴록사머, 히드록시프로필셀룰로오스(HPC-L, Nippon soda, Japan)와 발사르탄을 용해 또는 현탁시킨 결합액을 분무하여 발사르탄을 함유하는 펠렛을 형성, 건조하여 발사르탄 선방출성 펠렛을 제조하였다. As the ingredients and contents shown in Table 2, sugar seeds were added to a fluidized bed granulator (GPCG 1: Glatt), and then poloxamer and hydroxypropyl cellulose (HPC-L, Nippon) were mixed separately with purified water and ethanol. soda, Japan) and valsartan were sprayed to form a pellet containing valsartan by spraying a binder solution in which valsartan was dissolved or suspended to prepare valsartan pre-release pellets.
(2) 염산 네비볼롤 지연 방출 펠렛의 제조 (2) Preparation of Nebivolol Delayed-Release Pellets
표 2에 나타난 성분 및 함량으로, 슈가 시드(Sugar sphere)를 유동층 과립기(GPCG 1: Glatt)에 투입한 뒤, 따로 정제수와 에탄올 혼합용매에 히드록시프로필메틸셀룰로오스, 라우릴 황산나트륨과 염산 네비볼롤을 용해 또는 현탁시킨 결합액을 분무하여 염산 네비볼롤 함유 펠렛을 형성하고, 건조하였다. 다시 상기의 과립에 프탈산히드록시프로필메틸셀룰로오스를 에탄올과 염화메틸렌 혼합액에 녹인 액을 분무하여 염산 네비볼롤 지연 방출 펠렛을 제조하였다. In the ingredients and contents shown in Table 2, sugar seeds were added to a fluidized bed granulator (GPCG 1: Glatt), and then hydroxypropylmethylcellulose, sodium lauryl sulfate, and nebivolol hydrochloric acid were separately mixed with purified water and ethanol. The binder solution dissolved or suspended was sprayed to form a nebivolol-containing pellet, and dried. Again, the granules were sprayed with phthalic hydroxypropylmethylcellulose in a mixture of ethanol and methylene chloride to prepare nebivolol delayed-release pellets.
(3) 캡슐 충진(3) capsule filling
상기 (1), (2) 에서 제조한 두 종의 펠렛을 혼합하여 캡슐충전기로 0호 캡슐에 충전하여 캡슐제를 제조하였다. Capsules were prepared by mixing two pellets prepared in the above (1) and (2) and filling the capsule No. 0 with a capsule charger.
<실시예 9> 염산 네비볼롤 - 발사르탄 캡슐제 제조 (정제-펠렛) Example 9 Preparation of Navivolol Hydrochloride-Valsartan Capsule (Tablet-Pellets)
표 2에 나타난 성분 및 함량으로, 상기 실시예 8의 (1)과 동일한 방식으로 발사르탄 선방출 펠렛을 제조하고, 염산 네비볼롤 지연방출 정제는 상기 실시예 1의 (2) 방법으로 제조한 염산 네비볼롤의 지연방출층 과립에 스테아르산마그네슘을 투입 후, 최종 혼합한 혼합물을 직경 6 mm 펀치가 장착된 로타리 타정기(MRC-33 : 세종파마텍, 한국)로 타정을 하여 핵정을 제조하여, 정제와 펠렛을 캡슐충전기로 0호 캡슐에 충전하여 캡슐제를 제조하였다. With the ingredients and contents shown in Table 2, valsartan pre-release pellets were prepared in the same manner as in Example (1), and the nebivolol delayed-release tablet of hydrochloric acid was prepared by the method of Example 2 (2). Magnesium stearate was added to the delayed-release layer granules of the ball roll, and the final mixture was compressed into tablets and pellets by tableting with a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 6 mm diameter punch. The capsules were filled in capsule No. 0 with a capsule charger to prepare a capsule.
<실시예 10>  염산 네비볼롤-발사르탄 2상 매트릭스 정제의 제조 Example 10 Preparation of Nebivolol Hydrochloride-Valsartan Biphasic Matrix Tablets
(1) 발사르탄 선방출 과립 제조 (1) valsartan pre-release granule manufacture
표 2에 나타난 성분 및 함량으로, 발사르탄, 유당, 폴록사머, 미결정셀룰로오스, 전분글리콘산나트륨을 칭량하여 35 호체로 사과하고 더블콘믹서에서 20분간 혼합하여 혼합물을 제조하였다. With the ingredients and contents shown in Table 2, valsartan, lactose, poloxamer, microcrystalline cellulose, sodium starch glycolate were weighed and appled into No. 35 sieve and mixed for 20 minutes in a double cone mixer to prepare a mixture.
(2) 염산 네비볼롤의 지연방출 과립 제조 (2) Preparation of delayed-release granules of nebivolol hydrochloride
표 2에 나타난 성분 및 함량으로, 염산 네비볼롤, 미결정셀룰로오스, 라우릴 황산 나트륨을 35호체로 사과하고 더블콘믹서로 5분간 혼합하여 혼합물을 제조하였다. 별도로 히드록시프로필셀룰로오스를 정제수에 녹여 결합액으로 하였다. 위의 혼합물을 유동층 과립기에 투여한 후, 결합액을 분무하며 과립을 제조하였다. 완료된 과립을 유동층 건조기에서 건조한 후, 다시 상기의 과립에 카보머 71G(카복시비닐폴리머, 루브리졸)를 분말상태로 투입하여 10분간 혼합한 후, 일정한 크기로 체과하였다. 별도로 히드록시프로필메틸셀룰로오스를 80% 에탄올에 녹이고, 아크릴이즈(Acryl-eze, Colorcon, USA)를 정제수에 분산시켜 코팅액을 제조하였다. 코팅액 제조가 완료된 후, 상기 과립을 유동층 코팅기에 투여하고 1차 코팅(히드록시프로필메칠셀룰로오스 코팅액)을 한 후, 2차 코팅(아크릴이즈 코팅액)을 하였다. With the ingredients and contents shown in Table 2, a mixture of nebivolol hydrochloride, microcrystalline cellulose, and sodium lauryl sulfate as an apple No. 35 was mixed for 5 minutes with a double cone mixer. Separately, hydroxypropyl cellulose was dissolved in purified water to obtain a binding solution. After the above mixture was administered to the fluidized bed granulator, the binder was sprayed to prepare granules. The granules were dried in a fluidized bed dryer, and then carbomer 71G (carboxyvinyl polymer, lubricazole) was added to the granules in a powder state, mixed for 10 minutes, and sieved to a constant size. Separately, hydroxypropyl methyl cellulose was dissolved in 80% ethanol, and acrylic acid (Acryl-eze, Colorcon, USA) was dispersed in purified water to prepare a coating solution. After the preparation of the coating solution was completed, the granules were administered to the fluidized bed coater and subjected to the first coating (hydroxypropylmethylcellulose coating solution), followed by a second coating (acrylic coating solution).
(3) 타정 및 코팅 (3) tableting and coating
상기의 두 과립물을 혼합한 후, 스테아르산 마그네슘을 투입하여 4분간 혼합하여 직경 12.0 mm 펀치가 장착된 로타리 타정기(MRC-33: 세종파마텍, 한국)로 타정하였다. 타정이 완료된 정제를 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 및 산화티탄을 에탄올 및 정제수에 용해 및 분산시킨 코팅액으로 코팅하였다. After the two granules were mixed, magnesium stearate was added thereto, mixed for 4 minutes, and compressed using a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 12.0 mm diameter punch. Tablet-finished tablets were coated with a coating solution in which hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were dissolved and dispersed in ethanol and purified water.
<실시예 11> 염산 네비볼롤 - 발사르탄 블리스터 포장 키트의 제조 Example 11 Preparation of Navivolol Hydrochloride-Valsartan Blister Packaging Kit
(1) 발사르탄 선방출 과립 제조(1) valsartan pre-release granule manufacture
표 2에 나타난 성분 및 함량으로, 발사르탄, 유당, 폴록사머188, 미결정셀룰로오스, 전분글리콘산나트륨, 전젤라틴화전분(Starch 1500G, Colorcon, USA)을 칭량하여 35호체로 사과하고, 더블콘믹서에서 20분간 혼합하여 혼합물을 제조하였다. 혼합 완료 후, 스테아르산 마그네슘을 투입 후 4분간 혼합하여 발사르탄 선방출 과립을 제조하였다. Weigh valsartan, lactose, poloxamer 188, microcrystalline cellulose, sodium starch glycolate, starch gelatinized starch (Starch 1500G, Colorcon, USA) and weigh apples in No. 35 with the ingredients and contents shown in Table 2. The mixture was prepared by mixing for 20 minutes at. After completion of mixing, magnesium stearate was added and mixed for 4 minutes to prepare valsartan pre-release granules.
(2) 염산 네비볼롤의 지연방출 과립 제조 (2) Preparation of delayed-release granules of nebivolol hydrochloride
표 2에 나타난 성분 및 함량으로, 염산 네비볼롤, 미결정셀룰로오스, 라우릴 황산 나트륨을 35호체로 사과하고, 더블콘믹서로 5분간 혼합하여 혼합물을 제조하였다. 별도로 히드록시프로필셀룰로오스를 정제수에 녹여 결합액으로 하였다. 위의 혼합물을 유동층 과립기(GPCG-1, Glatt, Germany)에 투여한 후, 결합액을 분무하여 과립을 제조하였다. 완료된 과립을 유동층 건조기(GPCC-1, Glatt, Germany)에서 건조한 후, 다시 상기의 과립에 카보머 71G(카복시비닐폴리머, 루브리졸)를 분말상태로 투입하여 10분간 혼합한 후, 일정한 크기로 체과하였다. 체과 후 스테아르산 마그네슘을 투입하고 4분간 혼합하여 염산 네비볼롤 지연방출 과립을 제조하였다. With the ingredients and contents shown in Table 2, nebivolol hydrochloride, microcrystalline cellulose, sodium lauryl sulfate was appled with No. 35 sieve, and mixed for 5 minutes with a double cone mixer to prepare a mixture. Separately, hydroxypropyl cellulose was dissolved in purified water to obtain a binding solution. The above mixture was administered to a fluidized bed granulator (GPCG-1, Glatt, Germany), followed by spraying the binding solution to prepare granules. The granules were dried in a fluidized bed dryer (GPCC-1, Glatt, Germany), and then carbomer 71G (carboxyvinyl polymer, lubrizol) was added to the granules in a powder state and mixed for 10 minutes. I passed. After sieving, magnesium stearate was added and mixed for 4 minutes to prepare nebivolol delayed-release granules.
(3) 타정 및 코팅 후 포장 키트 제조 (3) Manufacture of packaging kits after tableting and coating
발사르탄 선방출 과립은 직경 6mm 펀치가 장착된 로타리 타정기(MRC-33: 세종파마텍, 한국)으로 타정하고, 염산 네비볼롤 지연방출 과립은 직경 5mm 펀치가 장착된 로타리 타정기(RC-30: 세종파마텍, 한국)로 타정하였다. 타정이 완료된 정제를 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 및 산화티탄을 에탄올 및 정제수에 용해 및 분산시킨 코팅액으로 코팅하였다. 코팅이 완료된 각각의 정제는 하나의 PTP(Press Through Pack)포장용기에 포장기(WiderVII, 부천기계, 한국)로 포장하여 동시복용이 가능한 포장키트를 제조하였다. Valsartan pre-release granules are compressed into a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 6 mm diameter punch.Navivolol delayed-release granules hydrochloric acid rotary tablets (RC-30: Sejong Pharmatech, equipped with a 5 mm diameter punch) Korea). Tablet-finished tablets were coated with a coating solution in which hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were dissolved and dispersed in ethanol and purified water. Each coated tablet was packaged in a single press through pack (PTP) packaging container with a packaging machine (WiderVII, Bucheon machine, Korea) to prepare a packaging kit for simultaneous use.
<실시예 12> 염산 네비볼롤 - 발사르탄 함유 필름코팅정의 제조 Example 12 Preparation of Navivolol Hydrochloride-Valsartan-Containing Film-Coated Tablets
(1) 염산 네비볼롤 나정의 제조 (1) Preparation of Navivolol Hydrochloride
표 2에 나타난 성분 및 함량으로, 염산 네비볼롤과 미결정 셀룰로오스, 라우릴황산나트륨을 35호체로 사과하고 더블콘믹서로 혼합한 후 유동층과립기(GPCG 1: Glatt)에 투입하고, 따로 히드록시프로필셀룰로오스를 물에 녹여 만든 결합액을 분무하여 과립을 형성, 건조완료 하였다. 다시 상기의 과립에 카보머 71G를 분말상태로 투입하여 10분간 혼합한 후, 여기에 스테아르산 마그네슘을 넣어 최종 더블콘믹서로 혼합한 후 상기 최종 혼합물을 로타리 타정기(MRC-33 : 세종파마텍, 한국)를 사용하여 지름 7 mm 펀치로 타정하여 나정을 제조하였다. In the ingredients and contents shown in Table 2, nebivolol hydrochloride, microcrystalline cellulose and sodium lauryl sulfate were appled in a No. 35 sieve, mixed in a double cone mixer, and then poured into a fluidized bed granulator (GPCG 1: Glatt), and separately hydroxypropyl cellulose. After spraying the binder solution dissolved in water to form granules, drying was completed. Carbomer 71G was added to the granules in a powder state and mixed for 10 minutes. Magnesium stearate was added thereto and mixed with a final double cone mixer, and the final mixture was rotary tablet press (MRC-33: Sejong Pharmatech, Korea). Uncoated tablet was prepared by tableting with a 7 mm diameter punch.
(2) 발사르탄 코팅액의 제조 (2) Preparation of Cheval Valsartan Coating Liquid
표 2에 나타난 성분 및 함량으로, 발사르탄, 콜로이드성이산화규소, 폴록사머, 및 히드록시프로필메틸셀룰로오스를 에탄올, 및 염화메칠렌 혼합액에 녹여 발사르탄 코팅액을 제조하였다. The valsartan coating solution was prepared by dissolving valsartan, colloidal silicon oxide, poloxamer, and hydroxypropylmethylcellulose in a mixture of ethanol and methylene chloride with the components and contents shown in Table 2.
(3) 코팅 (3) heat coating
염산 네비볼롤 나정을 하이코터(SFC-30F 세종 기계, 한국)에 투여하고 발사르탄 코팅액으로 코팅하였다. 약물 코팅완료 후, 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 및 산화티탄을 에탄올, 및 정제수에 용해 및 분산시킨 코팅액을 이용하여 코팅하였다. Hydrochloric acid nebivolol uncoated tablets were administered to a high coater (SFC-30F Sejong Machinery, Korea) and coated with valsartan coating solution. After the drug coating was completed, hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were coated using a coating solution dissolved and dispersed in ethanol and purified water.
<실시예 13> 염산 네비볼롤 발사르탄 삼투성 유핵정의 제조 Example 13 Preparation of Hydrochloric Acid Nebivolol Valsartan Osmotic Nucleated Tablets
(1) 염산 네비볼롤 지연방출 내핵의 제조(1) Preparation of delayed-release inner core of nebivolol hydrochloride
표 2의 성분 및 함량으로, 염산 네비볼롤과 미결정 셀룰로오스 및 염화나트륨, 라우릴황산나트륨을 35호체로 사과하고 더블콘믹서로 혼합한 후 여기에 스테아르산 마그네슘을 넣어 최종적으로 더블콘믹서로 혼합하고, 상기 최종 혼합물을 로타리 타정기(세종파마텍-30: 세종)를 사용하여 지름 5 mm 펀치로 타정하였다. 타정 후 삼투성 코팅기제로서 에틸셀룰로오스를 90% 에탄올에 분산시킨 후 하이코터(SFC-30F, 세종 기계, 한국)를 이용하여 내핵에 코팅하여 삼투성 핵정을 제조하였다. In the ingredients and contents of Table 2, nebivolol hydrochloride, microcrystalline cellulose and sodium chloride, sodium lauryl sulfate were apologized with a No. 35 sieve, mixed with a double cone mixer, and magnesium stearate was added thereto, and finally mixed with a double cone mixer, The final mixture was compressed into a 5 mm diameter punch using a rotary tablet press (Sejong Pharmatech-30: Sejong). After tableting, the osmotic coating was prepared by dispersing ethyl cellulose in 90% ethanol as an osmotic coating base and coating the inner core with a high coater (SFC-30F, Sejong Machinery, Korea).
(2) 발사르탄 선방출 과립의 제조 (2) Preparation of Valsartan Pre-Release Granules
표 2의 성분 및 함량으로, 실시예 11(1)의 방법과 동일한 방법으로 발사르탄 선방출 과립을 제조하였다. Valsartan pre-release granules were prepared in the same manner as in Example 11 (1), with the components and contents shown in Table 2.
(3) 타정 및 코팅 (3) tableting and coating
유핵정 타정기(RUD-1: Kilian)를 사용하여 염산 네비볼롤 삼투성 핵정을 내핵으로 하고 발사르탄 선방출성 과립을 외층으로 하여 지름 12 mm펀치로 타정한 다음 하이코터(SFC-30F, 세종 기계, 한국)로서 필름 코팅층을 형성하여 유핵정을 제조하였다. 타정이 완료된 정제를 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 산화티탄을 에탄올, 정제수에 용해 및 분산시킨 코팅액으로 코팅하였다. Using a nucleated tablet tableting machine (RUD-1: Kilian) as the inner core of hydrochloric acid nebivolol osmotic nuclear tablets and valsartan pre-release granules as outer layers, tablets were made with a 12 mm diameter punch and then a high coater ) To form a film coating layer to prepare a nucleated tablet. Tablets having been tableted were coated with a coating solution in which hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were dissolved and dispersed in ethanol and purified water.
<실시예 14> 염산 네비볼롤 - 로사르탄 칼륨염 2상 매트릭스 정제 제조 Example 14 Preparation of Navivolol Hydrochloride-Losartan Potassium Salt Two-Phase Matrix Tablets
(1) 로사르탄 칼륨염 선방출 과립의 제조(1) Preparation of Losartan Potassium Salt Pre-Release Granule
표 3에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄(Cadila, India)을 사용하는 것을 제외하고는 상기 실시예 1의 (1)과 동일한 방법으로 제조하였다. The ingredients and contents shown in Table 3 were prepared in the same manner as in Example 1 (1), except that losartan (Cadila, India) was used instead of mbalvaltan.
(2) 염산 네비볼롤 지연방출 과립의 제조(2) Preparation of Nebivolol Delayed-Release Granules Hydrochloride
표 3에 나타난 성분 및 함량으로, 셀룰로오스 아세테이트 (아세탈기 32%)와 셀룰로오스아세테이트(아세탈기 39.8%) 대신 유드라짓RL을 사용한 것을 제외하고는 상기 실시예 1의 (2)와 동일한 방법으로 제조하였다.Prepared in the same manner as in Example 2 (2), except that Eudragit RL was used instead of cellulose acetate (acetal group 32%) and cellulose acetate (39.8%) with the ingredients and contents shown in Table 3. It was.
(3) 타정 및 코팅 (3) tableting and coating
표 3에 나타난 성분 및 함량으로, 상기 (1)과 (2)에서 제조한 과립을 사용한 것을 제외하고, 실시예 1의 (3)과 동일한 방법으로 제조하였다.As the ingredients and contents shown in Table 3, except that the granules prepared in (1) and (2) were used, it was prepared in the same manner as in (3) of Example 1.
<실시예 15> 염산 네비볼롤 - 로사르탄 칼륨염 이중정 제조 Example 15 Preparation of Nebivolol Hydrochloride-Losartan Potassium Salt Double Tablets
표 3에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 2의 방법과 동일한 방법으로 제조하였다. The ingredients and contents shown in Table 3 were prepared in the same manner as in Example 2, except that losartan was used instead of mbalsartan.
<실시예 16> 염산 네비볼롤 - 로사르탄 칼륨염 다층정 제조 Example 16 Preparation of Navivolol Hydrochloride-Losartan Potassium Salt Multilayer Tablet
표 3에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 3과 동일한 방법으로 제조하였다. The ingredients and contents shown in Table 3 were prepared in the same manner as in Example 3, except that losartan was used instead of mbalsartan.
<실시예 17> 염산 네비볼롤 - 로사르탄 칼륨염유핵정의 제조 Example 17 Preparation of Nebivolol Hydrochloride-Losartan Potassium Salt Nucleated Tablets
(1) 염산 네비볼롤 지연방출 내핵의 제조 (1) Preparation of Navivolol delayed-release inner core
표 3에 나타난 성분 및 함량으로, 염산 네비볼롤과 미결정 셀룰로오스, 라우릴황산나트륨을 35호체로 사과하고 더블콘믹서로 혼합한 후 유동층과립기(GPCG 1: Glatt)에 투입하고, 따로 히드록시프로필셀룰로오스를 정제수에 녹여 만든 결합액을 분무하여 과립을 형성, 건조완료 하였다. 다시 상기의 과립에 카보머 71G를 분말상태로 투입하여 10분간 혼합한 후, 여기에 스테아르산 마그네슘을 넣어 최종 더블콘믹서로 혼합한 후 상기 최종 혼합물을 로타리 타정기(MRC-33: 세종파마텍)를 사용하여 지름 5 mm펀치로 타정하였다. 이렇게 제조한 나정을 염화메틸렌과 에탄올 혼합용매에 프탈산히드록시프로필메틸셀룰로오스(Shin-etsu, Japan)를 녹인 코팅액으로 코팅하여 핵정을 제조하였다. As the ingredients and contents shown in Table 3, apple nebivolol hydrochloride, microcrystalline cellulose and sodium lauryl sulfate were appled in a No. 35 sieve, mixed in a double cone mixer, and then put into a fluidized bed granulator (GPCG 1: Glatt), and separately hydroxypropyl cellulose. To prepare a granule by spraying the binder solution dissolved in purified water and dried. Carbomer 71G was added to the granules in a powder state and mixed for 10 minutes. Magnesium stearate was added thereto and mixed in a final double cone mixer, and the final mixture was mixed with a rotary tablet press (MRC-33: Sejong Pharmatech). It was compressed into a 5 mm diameter punch. The uncoated tablet thus prepared was coated with a coating solution in which phthalic acid hydroxypropylmethylcellulose (Shin-etsu, Japan) was dissolved in a mixed solvent of methylene chloride and ethanol to prepare a core tablet.
(2) 로사르탄 칼륨염선방출 과립의 제조 (2) Preparation of Pherosartan Potassium Salt-Released Granule
표 3에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄(Cadila, India)을 사용하는 것을 제외하고는 상기 실시예 2의 (1)과 동일한 방법으로 제조하였다. The ingredients and contents shown in Table 3 were prepared in the same manner as in Example 1, except that losartan (Cadila, India) was used instead of mbalvaltan.
(3) 후혼합, 타정 및 코팅 (3) post mixing, tableting and coating
유핵정 타정기(RUD-1: Kilian)를 사용하여 염산 네비볼롤 핵정을 내핵으로 하고 로사르탄 칼륨염 선방출성 과립을 외층으로 하여 타정하여 유핵정 제조를 완료한 후, 상기 실시예 1 (3)의 코팅방법과 동일한 방법으로 코팅하였다.After the preparation of the nucleated tablet by the tablet using a nucleated hydrochloric acid tablet as the inner core using a nucleated tablet tableting machine (RUD-1: Kilian) and an outer layer of losartan potassium salt as an outer layer, Coating was carried out in the same manner as the coating method.
<실시예 18> 염산 네비볼롤 - 로사르탄 칼륨염 캡슐제(정제 + 정제)의 제조 Example 18 Preparation of Nebivolol Hydrochloride-Losartan Potassium Salt Capsules (Tablets + Tablets)
표 3에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 5의 방법과 동일한 방법으로 제조하였다. The ingredients and contents shown in Table 3 were prepared in the same manner as in Example 5, except that losartan was used instead of zavalsartan.
<실시예 19> 염산 네비볼롤 - 로사르탄 칼륨염 캡슐제(정제 + 과립)의 제조 Example 19 Preparation of Nebivolol Hydrochloride-Losartan Potassium Salt Capsules (Tablets + Granules)
표 3에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 6의 방법과 동일한 방법으로 제조하였다. The ingredients and contents shown in Table 3 were prepared in the same manner as in Example 6, except that losartan was used instead of mbalsartan.
<실시예 20> 염산 네비볼롤 - 로사르탄 칼륨염 캡슐제(과립 + 과립)의 제조 Example 20 Preparation of Navivolol Hydrochloride-Losartan Potassium Salt Capsules (Granules + Granules)
다음 표 3에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 7의 방법과 동일한 방법으로 제조하였다. To the ingredients and contents shown in Table 3 below, was prepared in the same manner as in Example 7, except that losartan was used instead of mbalsartan.
<실시예 21> 염산 네비볼롤 - 로사르탄 칼륨염 캡슐제(펠렛 + 펠렛)의 제조 Example 21 Preparation of Navivolol Hydrochloride-Losartan Potassium Salt Capsule (Pellets + Pellets)
표 4에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 8의 방법과 동일한 방법으로 제조하였다. The ingredients and contents shown in Table 4 were prepared in the same manner as in Example 8, except that losartan was used instead of mbalsartan.
<실시예 22> 염산 네비볼롤 - 로사르탄 칼륨염 캡슐제(정제 + 펠렛)의 제조 Example 22 Preparation of Nebivolol Hydrochloride-Losartan Potassium Salt Capsules (Tablets + Pellets)
표 4에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 9의 방법과 동일한 방법으로 제조하였다. The ingredients and contents shown in Table 4 were prepared in the same manner as in Example 9, except that losartan was used instead of mbalsartan.
<실시예 23> 염산 네비볼롤 - 로사르탄 칼륨염 2상 매트릭스 정제의 제조 Example 23 Preparation of Nebivolol Hydrochloride-Losartan Potassium Salt Two-Phase Matrix Tablets
표 4에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 10의 방법에 따라 제조하였다. The ingredients and contents shown in Table 4 were prepared according to the method of Example 10, except that losartan was used instead of mbalvaltan.
<실시예 24> 염산 네비볼롤 - 키트제의 제조 Example 24 Preparation of Nebivolol Hydrochloride-Kit
표 4에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 11의 방법에 따라 제조하였다. The ingredients and contents shown in Table 4 were prepared according to the method of Example 11, except that losartan was used instead of mbalvaltan.
<실시예 25> 염산 네비볼롤 - 로사르탄 칼륨염 필름코팅정의 제조 Example 25 Preparation of Navivolol Hydrochloride-Losartan Potassium Salt Film-Coated Tablet
표 4에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 12의 방법에 따라 제조하였다. The ingredients and contents shown in Table 4 were prepared according to the method of Example 12, except that losartan was used instead of mbalsartan.
<실시예 26> 염산 네비볼롤 - 로사르탄 칼륨염 삼투성 유핵정의 제조 Example 26 Preparation of Nebivolol Hydrochloride-Losartan Potassium Salt Osmotic Nucleated Tablets
표 4에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄을 사용하는 것을 제외하고는 상기 실시예 13의 방법에 따라 제조하였다. The ingredients and contents shown in Table 4 were prepared according to the method of Example 13, except that losartan was used instead of mbalvaltan.
<실시예 27> 염산 네비볼롤 - 텔미사르탄 정제의 제조Example 27 Preparation of Nebivolol Hydrochloride -Telmisartan Tablets
(1) 텔미사르탄 선방출과립의 제조(1) Preparation of Telmisartan prior-release granules
표 5에 나타난 성분 및 함량으로, 발사르탄 대신 텔미사르탄(Ranbaxy, India)을 사용하는 것을 제외하고는 상기 실시예 1(1)의 방법에 따라 제조하였다. The ingredients and contents shown in Table 5 were prepared according to the method of Example 1 (1), except that telmisartan (Ranbaxy, India) was used instead of mbvalsartan.
(2) 염산 네비볼롤 지연방출과립의 제조(2) Preparation of delayed-release granules of nebivolol hydrochloride
표 5에 나타난 성분 및 함량으로, 셀룰로오스 아세테이트(아세탈기 32%)와 셀룰로오스아세테이트(아세탈기 39.8%) 대신 유드라짓RS을 사용한 것을 제외하고는 상기 실시예 1(2)의 방법에 따라 제조하였다. The ingredients and contents shown in Table 5 were prepared according to the method of Example 1 (2), except that Eudragit RS was used instead of cellulose acetate (acetal group 32%) and cellulose acetate (39.8% acetal group). .
(3) 타정 및 코팅 (3) tableting and coating
표 5에 나타난 성분 및 함량으로, 상기 실시예 1 (3)의 방법에 따라 제조하였다. To the ingredients and contents shown in Table 5, were prepared according to the method of Example 1 (3).
<실시예 28> 염산 네비볼롤 - 에프로사르탄 이중정 제조 Example 28 Preparation of Navivolol Hydrochloride-Eprosartan Double Tablet
표 5에 나타난 성분 및 함량으로, 발사르탄 대신 에프로사르탄(MSN, India)을 사용하는 것을 제외하고는, 상기 실시예 2의 방법에 따라 제조하였다. The ingredients and contents shown in Table 5 were prepared according to the method of Example 2, except that eprosartan (MSN, India) was used instead of valsartan.
<실시예 29> 염산 네비볼롤 - 칸데사르탄 실렉세틸 다층정 제조 Example 29 Preparation of Navivolol Hydrochloride-Candesartan Silexetil Multilayer Tablet
표 5에 나타난 성분 및 함량으로, 발사르탄 대신 칸데사르탄실렉세틸(Ranbaxy, India)을 사용하는 것을 제외하고는, 상기 실시예 3의 방법에 따라 제조하였다.The ingredients and contents shown in Table 5 were prepared according to the method of Example 3, except for using candesartan cilexetil (Ranbaxy, India) instead of valsartan.
<실시예 30> 염산 네비볼롤 - 이르베사르탄 유핵정 제조 Example 30 Preparation of Navivolol Hydrochloride-Irbesartan Nucleated Tablets
표 5에 나타난 성분 및 함량으로, 발사르탄 대신 이르베사르탄(Ranbaxy, India)을 사용하는 것을 제외하고는 상기 실시예 4의 방법에 따라 제조하였다. The ingredients and contents shown in Table 5 were prepared according to the method of Example 4, except for using Irbesartan (Ranbaxy, India) instead of valsartan.
<실시예 31> 염산 네비볼롤 올메사르탄 메독소밀 캡슐제(정제 + 과립) 제조 Example 31 Preparation of Hydrochloride Navivolol Olmesartan Medozomil Mill Capsule (Tablet + Granules)
표 5에 나타난 성분 및 함량으로, 발사르탄 대신 올메사르탄 메독소밀(Cadila, India)을 사용하는 것을 제외하고는 상기 실시예 6의 방법에 따라 제조하였다. The ingredients and contents shown in Table 5 were prepared according to the method of Example 6, except that Olmesartan Medoxomil (Cadila, India) was used instead of valsartan.
<실시예 32> 아테놀롤 칸데사르탄 실렉세틸 캡슐제(정제 + 펠렛) 제조 Example 32 Preparation of Athenolol Candesartan Silecetyl Capsule (Tablet + Pellets)
(1) 칸데사르탄 실렉세틸 선방출성 펠렛의 제조 (1) Preparation of candesartan cilexetil prior-release pellet
표 5에 나타난 성분 및 함량으로, 슈가 시드(Sugar sphere)를 유동층 과립기(GPCG 1: Glatt)에 투입한 뒤, 따로 정제수와 에탄올 혼합용매에 폴록사머, 수산화나트륨, 히드록시프로필메틸셀룰로오스와 칸데사르탄 실렉세틸을 용해 또는 현탁시킨 결합액을 분무하여 발사르탄을 함유하는 펠렛을 형성, 건조하여 칸데사르탄 실렉세틸 선방출성 펠렛을 제조하였다. With the ingredients and contents shown in Table 5, sugar seeds were added to a fluidized bed granulator (GPCG 1: Glatt), and then poloxamer, sodium hydroxide, hydroxypropylmethylcellulose and candee were mixed with purified water and ethanol. By spraying the binder solution in which the satan cilexetil was dissolved or suspended, pellets containing valsartan were formed and dried to prepare candesartan cilexetil pre-release pellets.
(2) 아테놀롤 지연 방출 정제의 제조 (2) Preparation of Atenolol Delayed-Release Tablets
표 5의 성분 및 함량으로, 아테놀롤과 미결정 셀룰로오스, 히드록시프로필셀룰로오스, 염화나트륨, 라우릴황산나트륨, 가교폴리비닐피롤리돈을 35호체로 사과하고 더블콘믹서로 혼합한 후 여기에 스테아르산 마그네슘을 넣어 최종적으로 더블콘믹서로 혼합하고, 상기 최종 혼합물을 로타리 타정기(세종파마텍-30: 세종)를 사용하여 지름 6 mm 펀치로 타정하였다. 타정 후 반투과 막 코팅기제로서, 셀룰로오스아세테이트(아세탈기32%)(Eastman Chemical Company, USA), 셀룰로오스아세테이트(아세탈기 39.8%)(Eastman Chemical Company, USA)를 에탄올과 염화메틸렌 혼합용매에 녹여 코팅액을 조제한 후, 하이코터(SFC-30F, 세종 기계, 한국)를 이용하여 상기 정제를 코팅하여 삼투성 정제를 제조하였다.In the ingredients and contents of Table 5, atenolol and microcrystalline cellulose, hydroxypropyl cellulose, sodium chloride, sodium lauryl sulfate, crosslinked polyvinylpyrrolidone were mixed with a No. 35 sieve, mixed with a double cone mixer, and magnesium stearate was added thereto. The mixture was finally mixed with a double cone mixer, and the final mixture was compressed into a 6 mm diameter punch using a rotary tablet press (Sejong Pharmatech-30: Sejong). As a semipermeable membrane coating base after tableting, cellulose acetate (acetal group 32%) (Eastman Chemical Company, USA) and cellulose acetate (acetal group 39.8%) (Eastman Chemical Company, USA) are dissolved in a mixed solvent of ethanol and methylene chloride After the preparation, the tablet was coated with a high coater (SFC-30F, Sejong Machinery, Korea) to prepare an osmotic tablet.
(3) 캡슐 충진(3) capsule filling
상기 칸데사르탄 실렉세틸 선방출성 펠렛과 아테놀롤 지연 방출 정제를 캡슐충전기로 0호 캡슐에 충전하여 캡슐제를 제조하였다The candesartan cilexetil pre-release pellet and atenolol delayed-release tablets were filled into capsule 0 using a capsule charger to prepare a capsule.
<실시예 33> 메토프롤롤 칸데사르탄 실렉세틸 캡슐제(펠렛 + 펠렛) 제조 Example 33 Preparation of Metoprolol Candesartan Silecetyl Capsule (Pellets + Pellets)
표 5에 나타난 성분 및 함량으로, 발사르탄 대신 칸데사르탄 실렉세틸, 염산 네비볼롤 대신 메토프롤롤(Dr Reddy, India)을 사용하는 것을 제외하고는 상기 실시예 8의 방법에 따라 제조하였다. The ingredients and contents shown in Table 5 were prepared according to the method of Example 8, except for using candesartan cilexetil instead of valsartan and metoprolol (Dr Reddy, India) instead of nebivolol hydrochloride.
<실시예 34> 아테놀롤 칸데사르탄 실렉세틸 필름코팅정의 제조 Example 34 Preparation of Athenolol Candesartan Silecetyl Film-Coated Tablets
(1) 아테놀롤 지연방출정의 제조 (1) Preparation of Atenolol Delayed-Release Tablets
표 6에 나타난 성분 및 함량으로, 아테놀롤, 미결정셀룰로오스(AvicelPH, FMC Biopolymer, USA), 라우릴황산나트륨, 히드록시프로필셀룰로오스, 카보머 71G, 가교비닐피롤리돈을 칭량하여 35 호체로 사과 후 더블콘믹서에서 20분간 혼합하여 혼합물을 제조하였다. 혼합물에 스테아르산 마그네슘을 투입하여 최종혼합한 후, 로타리 타정기(세종파마텍-30: 세종)를 사용하여 지름 5 mm 펀치로 타정하였다.Athenolol, microcrystalline cellulose (AvicelPH, FMC Biopolymer, USA), sodium lauryl sulfate, hydroxypropyl cellulose, carbomer 71G, and crosslinked vinylpyrrolidone were weighed into the ingredients and contents shown in Table 6. The mixture was prepared by mixing for 20 minutes in a cone mixer. Magnesium stearate was added to the mixture for final mixing, followed by compression with a 5 mm diameter punch using a rotary tablet press (Sejong Pharmatech-30: Sejong).
정제수에 아크릴이즈 (Acryl-EZE, Colorcon, USA)를 분산시켜 코팅액을 제조한 후, 하이코터(SFC-30F, 세종 기계, 한국)를 이용하여 상기 정제를 코팅하여 아테놀롤 지연방출정을 제조하였다.After dispersing acrylamide (Acryl-EZE, Colorcon, USA) in purified water to prepare a coating solution, using a high coater (SFC-30F, Sejong Machinery, South Korea) was coated with the tablet to prepare a delayed release of atenolol. .
(2) 칸데사르탄 실렉세틸 코팅액의 제조 (2) Preparation of candesartan cilexetil coating solution
표 6에 나타난 성분 및 함량으로, 칸데사르탄 실렉세틸, 콜로이드성이산화규소, 폴록사머, 및 히드록시프로필셀룰로오스를 에탄올, 및 염화메틸렌 혼합액에 녹여 칸데사르탄 실렉세틸 코팅액을 제조하였다. With the ingredients and contents shown in Table 6, candesartan cilexetil, colloidal silicon oxide, poloxamer, and hydroxypropyl cellulose were dissolved in a mixture of ethanol and methylene chloride to prepare a candesartan cilexetil coating solution.
(3) 코팅 (3) heat coating
아테놀롤 지연방출정을 하이코터(SFC-30F 세종 기계, 한국)에 투여하고 상기 (2)의 칸데사르탄 실렉세틸 코팅액으로 코팅하였다. 약물 코팅완료 후, 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 및 산화티탄을 에탄올, 및 정제수에 용해 및 분산시킨 코팅액으로 코팅하였다. Atenolol delayed-release tablets were administered to a high coater (SFC-30F Sejong Machinery, Korea) and coated with the candesartan cilexetil coating solution of (2). After the drug coating was completed, hydroxypropylmethylcellulose 2910, polyethylene glycol 6000, and titanium oxide were coated with a coating solution dissolved and dispersed in ethanol and purified water.
<실시예 35> 카르베딜롤 - 이르베사르탄 삼투성 유핵정 제조 Example 35 Preparation of Carvedilol-Irversartan Osmotic Nucleated Tablets
표 6에 나타난 성분 및 함량으로, 발사르탄 대신 이르베사르탄(Ranbaxy, India), 염산 네비볼롤 대신 카르베딜롤(Cadila, India)을 사용한 것을 제외하고는, 상기 실시예 13의 방법에 따라 제조하였다. The ingredients and contents shown in Table 6 were prepared according to the method of Example 13, except that irbesartan (Ranbaxy, India) instead of valsartan and carvedilol (Cadila, India) instead of nebivolol hydrochloride were used. .
<실시예 36> 카르베딜롤 - 에프로사르탄 유핵정 제조 Example 36 Preparation of Carvedilol-Eprosartan Nucleated Tablets
(1) 에프로사르탄 속방출과립의 제조(1) Preparation of Eprosartan Quick Release Granules
표 6에 나타난 성분 및 함량으로, 에프로사르탄(MSN), 유당(Parmatose, DMV Pharma, Netherlands), 미결정셀룰로오스(AvicelPH, FMC Biopolymer, USA), 폴록사머188(Lutrol-F68, BASF, Germany), 전젤라틴화전분을 칭량하여 35 호체로 사과하였다. 별도로 히드록시프로필셀룰로오스(HPC-L, Nippon soda, Japan)를 정제수에 녹여 결합액을 제조하였다. 상기 혼합물에 결합액을 가하여 조립한 후, 건조하였다. 건조물을20호체가 장착된 F형 정립기를 사용하여 정립한 후, 콜로이드성이산화규소와 스테아르산 마그네슘을 넣고 최종혼합하여 에프로사르탄 속방출 과립을 제조하였다.By ingredients and contents shown in Table 6, eprosartan (MSN), lactose (Parmatose, DMV Pharma, Netherlands), microcrystalline cellulose (AvicelPH, FMC Biopolymer, USA), poloxamer 188 (Lutrol-F68, BASF, Germany) The whole gelatinized starch was weighed and appled into No. 35 sieve. Separately, hydroxypropyl cellulose (HPC-L, Nippon soda, Japan) was dissolved in purified water to prepare a binding solution. A binder was added to the mixture to assemble it, and then dried. The dried product was stipulated using an F-type sizer equipped with a No. 20 sieve, and then colloidal silicon oxide and magnesium stearate were added and finally mixed to prepare eprosartan-release granules.
(2) 카르베딜롤 지연방출 과립의 제조(2) Preparation of Carvedilol Delayed-Release Granules
표 6에 나타난 성분 및 함량으로 염산 네비볼롤 대신 카르베딜롤을 사용한 것을 제외하고, 상기 실시예 1의 (2) 염산네비볼롤의 지연방출층 과립 제조방법과 동일한 방법으로 카르베딜롤 지연방출과립을 제조하였다.Carvedilol delayed-release granules were prepared in the same manner as the method of preparing granules of delayed-release layer granules of (2) nebivolol hydrochloride of Example 1, except that carvedilol was used instead of nebivolol hydrochloride in the ingredients and contents shown in Table 6. Prepared.
(3) 타정 및 코팅(3) tableting and coating
상기 (2)에서 제조된 카르베딜롤 지연방출층 과립을 직경 5 mm 펀치가 장착된 로타리 타정기(MRC-33 : 세종파마텍, 한국)로 타정을 하여 내핵을 제조한 후 12 mm 펀치가 장착된 유핵정타정기(RUD-Ⅰ Killian, 독일)에서 상기 (1)의 에프로사르탄 속방출 과립과 함께 타정하였다. 타정이 완료된 정제를 히드록시프로필메틸셀룰로오스 2910, 폴리에틸렌글리콜 6000, 산화티탄을 에탄올 및 정제수에 용해 및 분산시켜 제조한 코팅액으로 정제를 코팅하여 유핵정을 제조하였다. The carvedilol delayed-release layer granules prepared in (2) were compressed into tablets with a rotary tablet press (MRC-33: Sejong Pharmatech, Korea) equipped with a 5 mm diameter punch to prepare an inner core, followed by a 12 mm punch equipped oil. It was compressed with the eprosartan fast-release granules of (1) in a nuclear tablet press (RUD-I Killian, Germany). Tablets are finished tablets hydroxypropyl methyl cellulose 2910, polyethylene glycol 6000, titanium oxide was dissolved and dispersed in ethanol and purified water coating the tablet with a coating solution to prepare a nucleated tablet.
<실시예 37> 카르베딜롤 - 텔미사르탄 캡슐제(과립 + 과립) 제조 Example 37 Preparation of Carvedilol-Telmisartan Capsule (Granule + Granule)
표 6에 나타난 성분 및 함량으로, 발사르탄 대신 텔미사르탄(Ranbaxy, India), 염산 네비볼롤 대신 카르베딜롤을 사용하는 것을 제외하고는 상기 실시예 7의 방법에 따라 제조하였다. The ingredients and contents shown in Table 6 were prepared according to the method of Example 7, except for using telmisartan (Ranbaxy, India) instead of valsartan and carvedilol instead of nebivolol hydrochloride.
<실시예 38> 카르베딜롤 칸데사르탄 실렉세틸 삼투성 유핵정 제조 <Example 38> Carvedilol candesartan cilexetil osmotic nucleated tablet preparation
(1) 칸데사르탄 실렉세틸 속방출 과립의 제조(1) Preparation of candesartan cilexetil rapid release granules
표 6에 나타난 성분 및 함량으로, 에프로사르탄 대신 칸데사르탄 실렉세틸(Ranbaxy, India)을 사용한 것을 제외하고는 상기 실시예 36 (1)의 방법에 따라 제조하였다.The ingredients and contents shown in Table 6 were prepared according to the method of Example 36 (1), except that candesartan cilexetil (Ranbaxy, India) was used instead of eprosartan.
(2) 카르베딜롤 지연방출 과립의 제조(2) Preparation of Carvedilol Delayed-Release Granules
표 6에 나타난 성분 및 함량으로, 상기 실시예 13 (2)의 방법에 따라 제조하였다.To the ingredients and contents shown in Table 6, were prepared according to the method of Example 13 (2).
(3) 타정 및 코팅(3) tableting and coating
표 6에 나타난 성분 및 함량으로, 상기 실시예 36 (3)과 동일한 방법으로 타정 및 코팅하였다.The ingredients and the contents shown in Table 6 were tableted and coated in the same manner as in Example 36 (3).
<실시예 39> 메토프롤롤 - 로사르탄 칼륨염 블리스터 키트 제조 Example 39 Preparation of Metoprolol-Losartan Potassium Salt Blister Kit
표 6에 나타난 성분 및 함량으로, 발사르탄 대신 로사르탄, 염산 네비볼롤 대신 메토프롤롤(Dr Reddy, India)을 사용하는 것을 제외하고는 상기 실시예 11의 방법에 따라 제조하였다. The ingredients and contents shown in Table 6 were prepared according to the method of Example 11 except for using losartan instead of valsartan and metoprolol (Dr Reddy, India) instead of nebivolol hydrochloride.
[표 1]TABLE 1
Figure PCTKR2009003969-appb-I000001
Figure PCTKR2009003969-appb-I000001
[표 2]TABLE 2
Figure PCTKR2009003969-appb-I000002
Figure PCTKR2009003969-appb-I000002
[표 3]TABLE 3
Figure PCTKR2009003969-appb-I000003
Figure PCTKR2009003969-appb-I000003
[표 4]TABLE 4
Figure PCTKR2009003969-appb-I000004
Figure PCTKR2009003969-appb-I000004
[표 5]TABLE 5
Figure PCTKR2009003969-appb-I000005
Figure PCTKR2009003969-appb-I000005
[표 6]TABLE 6
Figure PCTKR2009003969-appb-I000006
Figure PCTKR2009003969-appb-I000006
<실험예 1> 용출 양상 시험 (dissolution profile test) Experimental Example 1 Dissolution Profile Test
상기 실시 예 2에서 얻은 정제와 대조약으로 발사르탄(Novartis : Diovan, 발사르탄 단일제)과 네비볼롤(Forest labs: Bystolic, 네비볼롤 단일제)을 사용하여 비교 용출시험을 실시하였다. 네비볼롤 성분 제제의 경우 2시간을 기점으로 용출액을0.1 N-염산용액(산성환경)에서 pH 6.8(인공장액)완충액으로 변경하여 용출시험을 진행하였다. 발사르탄의 경우, 인산염완충액을 사용하였다. 각 성분별 용출시험 방법은 아래와 같으며, 그 결과를 도 1에 나타내었다(시험 검체수는 각각 12개). Comparative dissolution tests were performed using valsartan (Novartis: Diovan, Valsartan monolith) and nebivolol (Forest labs: Bystolic, nebivolol monolith) as the tablets and the control agent obtained in Example 2. In the case of the nebivolol component preparation, the dissolution test was performed by changing the elution solution from 0.1 N-hydrochloric acid solution (acidic environment) to pH 6.8 (phosphate solution). In the case of valsartan, phosphate buffer was used. The dissolution test method for each component is as follows, and the results are shown in FIG. 1 (the number of test samples is 12 pieces each).
도 1에 의하면 실시예 2의 정제는 하기 조건에서 용출 시험시 발사르탄 성분은 대조 제제인 디오반(Diovan)과 비교하여 거의 동등한 용출특성을 나타내는 것으로 확인되었으며, 네비볼롤 성분은 대조 제제인 Bystolic과 비교할 때 매우 늦어진 용출 속도를 확인할 수 있다. 실시예 2의 네비볼롤/발사르탄 이중정제는 2 시간까지의 네비볼롤 성분의 용출률이 모두 10% 이내로서 대조 제제의 용출률(약 99%) 보다 훨씬 느리다. According to FIG. 1, the tablet of Example 2 was found to have almost the same elution characteristics as the valsartan component in the dissolution test under the following conditions, compared to the control agent Diovan, and the nebivolol component was compared with the control agent Bystolic. Very slow elution rates can be seen. The nebivolol / valsartan double tablets of Example 2 had a dissolution rate of all of the nebivolol components up to 2 hours within 10%, much slower than the dissolution rate of the control formulation (about 99%).
이처럼 본 발명의 네비볼롤/발사르탄의 정제는 대조약인 네비볼롤 단일제와 달리 네비볼롤의 초기 방출이 발사르탄보다 매우 느리기 때문에 발사르탄보다 먼저 간에서 대사를 받을 확률이 낮아지게 된다. As such, the tablets of the nebivolol / valsartan of the present invention are unlikely to be metabolized in the liver before the valsartan because the initial release of the nebivolol is much slower than the valsartan, in contrast to the control drug nevivolol single agent.
[네비볼롤 시험방법] [Navivolol test method]
용출시험 근거 : 대한약전 제 8 개정 중 일반시험법의 용출시험법 Dissolution test basis: Dissolution test method of General Test Method
시험 방법 : 패들법(Paddle method), 50 회전/분 Test method: Paddle method, 50 revolutions / minute
시험액 : 0.1 N 염산용액, 500 mL (0~2시간), Test solution: 0.1 N hydrochloric acid solution, 500 mL (0-2 hours),
pH 6.8 인공장액, 1,000 mL (2시간 이후)          pH 6.8 artificial intestine, 1,000 mL (after 2 hours)
분석방법 : 자외가시부흡광광도법 (검출파장= 최대 280 nm) Analysis method: UV-visible absorption spectroscopy (detection wavelength = maximum 280 nm)
[발사르탄 시험방법] [Valsartan Test Method]
용출시험 근거 : 미국약전(USP 31)중의 'Valsartan and Hydrochlorothiazide tablet'항 Dissolution test basis: 'Valsartan and Hydrochlorothiazide tablet' in USP 31
시험 방법 : 패들법(Paddle method), 50회전/분 Test method: Paddle method, 50 revolutions / minute
시험액 : pH=6.8 완충액(인산염 용액), 1000 mL Test solution: pH = 6.8 buffer (phosphate solution), 1000 mL
분석방법 : 자외가시부흡광광도법 (검출파장= 최대 270, 최소 250 nm) Analysis method: UV-visible absorption spectroscopy (detection wavelength = maximum 270, minimum 250 nm)
<실험예 2> 용출 양상 시험 (dissolution profile test) Experimental Example 2 Dissolution Profile Test
실시예 2, 3, 4, 5에서 얻은 여러 제형의 제제를 실험예 1의 네비볼롤 비교용출방법에 따라 실시하여, 그 결과를 도 2에 나타내었다(시험 검체수는 각각 12개임). Formulations of various formulations obtained in Examples 2, 3, 4, and 5 were carried out according to the nebivolol comparative elution method of Experimental Example 1, and the results are shown in FIG. 2 (the number of test samples was 12 each).
도 2의 결과에서 보는 바와 같이 일정 시간 경과 후 네비볼롤이 용출되는 것을 확인할 수 있다. 따라서 본 발명의 목적대로 실시예 2 내지 5에서 제조된 제제는 저녁시간 경구 투여 시 지연용출되는 네비볼롤이 낮 시간 동안의 혈압을 조절할 수 있음을 알 수 있다. 또한, 경구투여 형태가 달라지더라도 네비볼롤의 용출률의 편차가 크지 않으므로 실시예의 모든 제형으로 본 발명의 제제를 개발할 수 있음을 확인할 수 있다. As shown in the results of Figure 2 it can be seen that after a certain time elapsed navigolol. Therefore, it can be seen that the preparation prepared in Examples 2 to 5 for the purpose of the present invention can control the blood pressure during the day time of nebivolol delayed eluting during oral administration during the evening. In addition, even if the oral dosage form is different because the deviation of the dissolution rate of nebivolol can be confirmed that the formulation of the present invention can be developed in all formulations of the examples.
<실험예 3> 용출 양상 시험 (dissolution profile test) Experimental Example 3 Dissolution Profile Test
실시예 14에서 제조한 정제와 대조약 로사르탄(Merck: Cozaar, 로사르탄 단일제)과 네비볼롤(Forest labs: Bystolic, 네비볼롤 단일제)을 사용하여 실험예 1의 네비볼롤 비교 용출시험방법에 따라 실시하였고, 로사르탄 비교용출시험방법은 아래와 같이 실시하여, 각각의 결과를 도 3에 나타내었다(시험 검체수는 각각 12개임). Using the tablets prepared in Example 14 and the control agent Losartan (Merck: Cozaar, Losartan single agent) and nebivolol (Forest labs: Bystolic, Nebivolol single agent) according to the comparative test method of Navivolol of Experimental Example 1 Losartan comparative dissolution test method was carried out as follows, each result is shown in Figure 3 (the number of test samples each 12).
도 3은 안지오텐신-2 수용체 길항제 약물인 로사르탄과 네비볼롤의 복합제인 실시예 14의 정제를 로사르탄과 네비볼롤 각각의 대조약과 비교 용출시험한 결과로, 네비볼롤은 본 발명이 의도한 용출시험개시 2시간 후에 약물이 용출되기 시작하여 1 시간 이내에 80% 이상이 용출되는 것을 확인할 수 있었다. FIG. 3 is a result of comparative dissolution test of the tablet of Example 14, a combination agent of losartan and nebivolol, which is an angiotensin-2 receptor antagonist drug, and a control drug of losartan and nebivolol. The drug began to elute 2 hours after the start and it was confirmed that more than 80% eluted within 1 hour.
[로사르탄 시험방법] [Losartan Test Method]
용출시험 근거 : Dissolution Methods for Drug Products(FDA) 중의 'Losartan Potassium tablet'항 Dissolution test basis: 'Losartan Potassium tablet' in Dissolution Methods for Drug Products (FDA)
시험 방법 : 패들법(Paddle method), 50회전/분 Test method: Paddle method, 50 revolutions / minute
시험액 : 물(탈기), 900 mL Test liquid: water (degassing), 900 mL
분석방법 : 자외가시부흡광광도법 (검출파장= 254 nm) Analytical Method: Ultraviolet-visible Spectrophotometry (Detect Wavelength = 254 nm)
<실험예 4> 용출 양상 시험 (dissolution profile test) Experimental Example 4 Dissolution Profile Test
실시예 21, 22, 23, 24에서 얻은 여러 제형의 제제와 대조약 네비볼롤(Forest labs: Bystolic, 네비볼롤 단일제)을 사용하여 실험예 1의 네비볼롤 비교 용출시험방법에 따라 실시하였고, 그 결과를 도 4에 나타내었다(시험 개체수는 각각 12개임). Using the formulations of the various formulations obtained in Examples 21, 22, 23, 24 and the control drug nebivolol (Forest labs: Bystolic, nebivolol single agent) was carried out according to the nebivolol comparative dissolution test method of Experimental Example 1 Is shown in Figure 4 (test population of 12 each).
도 4는 상기 실시 예 21, 22, 23, 24의 제제와 네비볼롤 대조약과의 비교용출시험 결과를 나타내며, 네비볼롤은 본 발명이 의도한 용출시험개시 2시간 후에 약물이 용출되기 시작하여 1 시간 이내에 80% 이상이 용출되는 것을 확인할 수 있었다. Figure 4 shows the comparative dissolution test results of the formulations of Examples 21, 22, 23, 24 and the nebivolol control, the nebivolol is 1 hour after starting the drug dissolution 2 hours after the start of the dissolution test intended by the present invention It was confirmed that more than 80% eluted within.
<실험예 5> 용출 양상 시험 (dissolution profile test) Experimental Example 5 Dissolution Profile Test
실시 예 21, 22, 23, 24에서 얻은 여러 제형의 제제와 대조약 로사르탄(Merck:Cozaar, 로사르탄 단일제)을 사용하여 실험예 3의 로사르탄 비교 용출시험방법에 따라 실시하였고, 그 결과를 도 5에 나타내었다(시험 개체수는 각각 12개임). Using the formulation of the various formulations obtained in Examples 21, 22, 23, 24 and the control agent Losartan (Merck: Cozaar, Losartan single agent) according to the Losartan comparative dissolution test method of Experimental Example 3, the results It is shown in FIG. 5 (the test population was 12 each).
도 5 는 실시 예 21, 22, 23, 24의 제제와 로사르탄 대조약과의 비교용출시험결과를 나타내며, 본 발명의 실시예에 따른 로사르탄 용출률은 대조약과 동등하거나, 유사한 용출양상을 나타내었다. 5 shows comparative dissolution test results of the formulations of Examples 21, 22, 23, and 24 and the Losartan reference drug, and the Losartan dissolution rate according to the Examples of the present invention showed the same or similar dissolution pattern as the reference drug.
<실험예 6> 용출 양상 시험 (dissolution profile test) Experimental Example 6 Dissolution Profile Test
실시예 34에서 제조한 정제와 대조약 칸데사르탄(Astrazeneca : Atacand, 칸데사르탄 단일제)과 아테놀롤(현대약품 : 현대테놀민정, 아테놀롤 단일제)을 사용하여 아래와 같은 방법으로 비교 용출시험을 실시하였다. A comparative dissolution test was conducted using the tablet prepared in Example 34 and the control agent candesartan (Astrazeneca: Atacand, candesartan mono) and atenolol (modern drugs: Hyundai tenolmin tablet, atenolol mono). It was.
아테놀롤 성분 제제의 경우 2시간을 기점으로 용출액을 0.1 N-염산용액(산성환경)에서 pH 6.8(인공장액)완충액으로 변경하여 용출시험을 진행하였다. 각 성분별 용출시험 방법은 아래와 같으며, 그 결과를 도 6에 나타내었다(시험 검체수는 각각 12개임).For the atenolol component preparation, the dissolution test was performed by changing the eluate from 0.1 N hydrochloric acid solution (acidic environment) to pH 6.8 (phosphate) solution. The dissolution test method for each component is as follows, and the results are shown in FIG. 6 (the number of test samples is 12 each).
도 6에 의하면 실시예 34의 정제는 하기 조건에서 용출 시험시 칸데사르탄 성분은 대조 제제인 아타칸(아스트라제네카 : Atacand, 칸데사르탄 단일제)과 비교하여 거의 동등한 용출특성을 나타내는 것으로 확인되었으나, 아테놀롤 성분은 대조 제제인 현대테놀민과 용출결과를 비교할 때 초기 약물방출이 지연된 것을 확인할 수 있다. According to Figure 6, the tablet of Example 34 was confirmed that the candesartan component in the dissolution test under the following conditions compared to the control agent atacane (Astrageneca: Atacand, Candesartan single agent), but almost the same elution characteristics, The norol component can be seen that the initial drug release is delayed when comparing the dissolution results with the control agent Hyundaitenolmin.
이처럼 본 발명의 아테놀롤/칸데사르탄 정제는 대조약인 아테놀롤 단일제와 달리 아테놀롤의 초기 방출이 칸데사르탄보다 매우 느리기 때문에 칸데사르탄보다 먼저 간에서 대사를 받을 확률이 낮아지게 된다. As described above, the atenolol / candesartan tablet of the present invention is unlikely to be metabolized in the liver prior to candesartan because the initial release of atenolol is much slower than candesartan, unlike the atenolol monotherapy, which is a reference drug.
도 6 의 결과로부터 네비볼롤 이외의 아드레날린 베타 수용체 차단제 계열 약물인 아테놀롤을 칸데사르탄과 함께 사용하여 제조한 제제에 있어서도 본 발명이 의도한 효과를 얻을 수 있음을 확인하였다. From the results of FIG. 6, it was confirmed that the present invention can achieve the intended effect even in preparations prepared by using athenolol, which is an adrenaline beta receptor blocker-based drug other than nebivolol, in combination with candesartan.
[아테놀롤 시험방법] [Athenolol Test Method]
용출시험 근거 : 대한약전 제 8 개정 중 일반시험법의 용출시험법 Dissolution test basis: Dissolution test method of General Test Method
시험 방법 : 패들법(Paddle method), 50 회전/분 Test method: Paddle method, 50 revolutions / minute
시험액 : 0.1 N 염산용액, 500 mL (0~2시간), Test solution: 0.1 N hydrochloric acid solution, 500 mL (0-2 hours),
pH 6.8 인공장액, 1,000 mL (2시간 이후)         pH 6.8 artificial intestine, 1,000 mL (after 2 hours)
분석방법 : 고속액체크로마토그래프법 Analysis method: high performance liquid chromatograph method
검출 파장 : 226 nm Detection wavelength: 226 nm
이동상 : 메탄올:pH 3.0 완충액(헵탄설폰산나트륨:무수인산수소나트륨:디부틸아민=1.1 g:0.71 g:2 mL, 0.8 M 인산으로pH 조정) = 300:700 Mobile phase: Methanol: pH 3.0 buffer (Sodium heptane sulfonate: Sodium hydrogen phosphate anhydrous: Dibutylamine = 1.1 g: 0.71 g: 2 mL, pH adjusted with 0.8 M phosphoric acid) = 300: 700
컬럼 : 안지름 3.9 mm, 길이 300 mm의 스테인레스관에 옥타데실실릴화한 겔 Column: octadecylsilylated gel in stainless steel tube with inner diameter of 3.9 mm and length of 300 mm
유속 : 0.6 mL/분 Flow rate: 0.6 mL / min
[칸데사르탄 시험방법] [Candesartan Test Method]
용출시험 근거 : Dissolution Methods for Drug Products(FDA) 중의'Candesartan Cilexetil tablet'항 Dissolution test basis: 'Candesartan Cilexetil tablet' in Dissolution Methods for Drug Products (FDA)
시험 방법 : 패들법(Paddle method), 50 회전/분 Test method: Paddle method, 50 revolutions / minute
시험액 : pH=6.5 완충액(조성= 0.35% 폴리소르베이트의 0.05 몰 인산염 용액), 900 mL Test solution: pH = 6.5 buffer (composition = 0.05 molar phosphate solution of 0.35% polysorbate), 900 mL
분석방법 : 고속액체크로마토그래프법 Analysis method: high performance liquid chromatograph method
검출 파장 : 215 nm Detection wavelength: 215 nm
이동상 : 물(0.1% 트리플루오로아세트산 ):아세토나이트릴(0.1% 트리플루오로아세트산 ) = 30:70 Mobile phase: water (0.1% trifluoroacetic acid): acetonitrile (0.1% trifluoroacetic acid) = 30:70
컬럼 : 안지름 4.6 mm, 길이 250 mm의 스테인레스관에 옥타데실실릴화한 겔 Column: octadecylsilylated gel in stainless steel pipe with inner diameter of 4.6 mm and length 250 mm
유속 : 1 mL/분 Flow rate: 1 mL / min
<실험예 7> 용출 양상 시험 (dissolution profile test) Experimental Example 7 Dissolution Profile Test
실시 예 36, 37, 39에서 얻은 여러 제형의 제제와 대조약 에프로사르탄(Abbott : Teveten, 에프로사르탄 단일정), 카르베딜롤(종근당: 딜라트렌,카르베딜롤 단일정), 텔미사르탄(Boehringer ingelheim: Micardis, 텔미사르탄 단일제), 메토프롤롤(Astrazeneca: Toprol-XL, 메토프롤롤 서방출성 정제 단일제)을 사용하여 비교용출시험을 실시하였다. Formulations of the various formulations obtained in Examples 36, 37 and 39 and the control agent eprosartan (Abbott: Teveten, eprosartan monolith), carvedilol (common root: dilatrene, carvedilol monolith), telme Comparative dissolution tests were carried out using Sartan (Boehringer ingelheim: Micardis, Telmisartan mono), Metoprolol (Astrazeneca: Toprol-XL, Metoprolol sustained-release tablet mono).
카르베딜롤 및 메토프롤롤 성분 제제의 경우 2시간을 기점으로 용출액을 0.1 N-염산용액(산성환경)에서 pH 6.8(인공장액)완충액으로 변경하여 용출시험을 진행하였다. 각 성분별 용출시험 방법은 아래와 같으며, 그 결과를 도 7, 8, 9에 나타내었다(시험 개체수는 각각 12개임).   For the preparation of carvedilol and metoprolol, the dissolution test was performed by changing the eluate from 0.1 N hydrochloric acid solution (acid environment) to pH 6.8 (phosphate solution). The dissolution test method for each component is as follows, and the results are shown in FIGS. 7, 8, and 9 (the test population is 12 each).
도 7, 8, 9의 결과로부터 네비볼롤 이외의 아드레날린 베타 수용체 차단제 계열 약물인 카르베딜롤, 메토프롤롤도 의도한 방출패턴을 가짐을 확인하였다. 7, 8, and 9 it was confirmed that carvedilol and metoprolol, which are adrenergic beta receptor blocker-based drugs other than nebivolol, also have an intended release pattern.
[카르베딜롤 시험방법] [Carvedilol Test Method]
용출시험 근거 : 대한약전 제 8 개정 중 일반시험법의 용출시험법 Dissolution test basis: Dissolution test method of General Test Method
시험 방법 : 패들법(Paddle method), 50 회전/분 Test method: Paddle method, 50 revolutions / minute
시험액 : 0.1 N 염산용액, 500 mL (0~2시간), Test solution: 0.1 N hydrochloric acid solution, 500 mL (0-2 hours),
pH 6.8 인공장액, 1,000 mL (2시간 이후)         pH 6.8 artificial intestine, 1,000 mL (after 2 hours)
분석방법 : 자외가시부흡광광도법 (검출파장= 최대 240 nm) Analysis method: UV-visible absorption spectroscopy (detection wavelength = maximum 240 nm)
[메토프롤롤 시험방법] [Methoprolol test method]
용출시험 근거 : 미국약전(USP 29) 중의 ‘Metoprolol Succinate Extended-Release Tablets’항 Dissolution test basis: Metoprolol Succinate Extended-Release Tablets in the US Pharmacopeia (USP 29)
시험 방법 : 패들법(Paddle method), 50 회전/분 Test method: Paddle method, 50 revolutions / minute
시험액 : 0.1 N 염산용액, 500 mL (0~2시간),Test solution: 0.1 N hydrochloric acid solution, 500 mL (0-2 hours),
pH 6.8 인산완충액, 500 mL          pH 6.8 phosphate buffer, 500 mL
분석방법 : 고속액체크로마토그래프법 Analysis method: high performance liquid chromatograph method
검출 파장 : 280 nm Detection wavelength: 280 nm
이동상 : 아세토니트릴:pH 3.0 인산완충액 = 125:375 Mobile phase: Acetonitrile: pH 3.0 phosphate buffer = 125: 375
컬럼 : 안지름 4.0 mm, 길이 125 mm의 옥틸실란 컬럼 Column: 4.0 mm inner diameter, 125 mm long octylsilane column
유속 : 1.0 mL/분 Flow rate: 1.0 mL / min
[에프로사르탄 시험방법] [Eprosartan Test Method]
용출시험 근거 : Dissolution Methods for Drug Products(FDA) 중의 'Eprosartan Mesylate/Hydrochlorothiazide tablet'항 Dissolution test basis: 'Eprosartan Mesylate / Hydrochlorothiazide tablet' in Dissolution Methods for Drug Products (FDA)
시험 방법 : 패들법(Paddle method), 75 회전/분 Test method: Paddle method, 75 revolutions / minute
시험액 : pH=7.5 완충액(인산염 용액), 1000 mL Test solution: pH = 7.5 buffer (phosphate solution), 1000 mL
분석방법 : 고속액체크로마토그래프법 Analysis method: high performance liquid chromatograph method
검출 파장 : 235 nm Detection wavelength: 235 nm
이동상 : 물(0.1몰 아세트산 암모늄):아세토나이트릴 = 50:50 Mobile phase: water (0.1 mol ammonium acetate): acetonitrile = 50:50
컬럼 : 안지름 4.6 mm, 길이 750 mm의 스테인레스관에 옥타데실실릴화한 겔 Column: octadecylsilylated gel in stainless steel tube with inner diameter of 4.6 mm and length of 750 mm
유속 : 2 mL/분 Flow rate: 2 mL / min
[텔미사르탄 시험방법] [Telmisartan Test Method]
용출시험 근거 : Dissolution Methods for Drug Products(FDA) 중의'Telmisartan tablet'항 Dissolution test basis: 'Telmisartan tablet' in Dissolution Methods for Drug Products (FDA)
시험 방법 : 패들법(Paddle method), 75 회전/분 Test method: Paddle method, 75 revolutions / minute
시험액 : pH=7.5 완충액(인산염 용액), 900 mL Test solution: pH = 7.5 buffer (phosphate solution), 900 mL
분석방법 : 고속액체크로마토그래프법 Analysis method: high performance liquid chromatograph method
검출 파장 : 271 nm Detection wavelength: 271 nm
이동상 : 물(0.05몰 인산이수소칼륨 ):아세토나이트릴 = 60:40 Mobile phase: Water (0.05 mol potassium dihydrogen phosphate): acetonitrile = 60:40
컬럼 : 안지름 4.6 mm, 길이 250 mm의 스테인레스관에 옥타데실실릴화한 겔 Column: octadecylsilylated gel in stainless steel pipe with inner diameter of 4.6 mm and length 250 mm
유속 : 1 mL/분 Flow rate: 1 mL / min
본 발명의 제제는 균등한 항압작용과 합병증 예방 작용을 나타내며, 특히 합병증 발생 위험시간대에 혈압을 균등하게 유지시켜 줄 수 있어 합병증을 지닌 고혈압 환자, 수면 중 혈압억제 등에 유용하고, 약물간 상호작용에 따른 부작용을 감소시켜 준다.The formulation of the present invention exhibits an equal anti-pressure action and prevents complications, and is particularly useful for hypertension patients with complications, suppressing blood pressure during sleep, and for drug interactions. Reduce side effects.

Claims (38)

  1. 약리학적 활성성분으로 안지오텐신-2 수용체 길항제를 포함하는 선방출성 구획, 및 약리학적 활성성분으로 베타 아드레날린 차단제를 포함하는 지연방출성 구획을 포함하는 약제학적 제제. A pharmaceutical formulation comprising a prior release compartment comprising an angiotensin-2 receptor antagonist as a pharmacologically active ingredient and a delayed release compartment comprising a beta adrenergic blocker as a pharmacologically active ingredient.
  2. 제1항에 있어서, 상기 베타아드레날린 차단제는 안지오텐신-2 수용체 길항제 방출 후 1시간 내지 8시간 이후에 방출되는 것인 약제학적 제제. The pharmaceutical formulation of claim 1, wherein the beta adrenergic blocker is released 1 hour to 8 hours after release of the angiotensin-2 receptor antagonist.
  3. 제1항에 있어서, 상기 선방출성 구획에 포함되는 활성성분은 방출개시 후 1시간 이내에 85중량% 이상이 방출되는 것인 약제학적 제제. The pharmaceutical formulation of claim 1, wherein the active ingredient included in the pre-release compartment releases at least 85% by weight within 1 hour after the start of release.
  4. 제1항에 있어서, 상기 제제는 안지오텐신-2 수용체 길항제 방출개시 후 2시간 이내에 방출되는 베타 아드레날린 차단제의 방출량이 베타아드레날린 차단제 총량의 0~10% 이내인 약제학적 제제. The pharmaceutical formulation of claim 1, wherein the amount of the beta adrenergic blocker released within 2 hours after the release of the angiotensin-2 receptor antagonist is within 0-10% of the total amount of the beta adrenergic blocker.
  5. 제1항 내지 제4항 중 어느 한 항에 있어서, 상기 안지오텐신-2 수용체 길항제는 로사르탄, 발사르탄, 텔미사르탄, 이르베사르탄, 칸데사르탄, 올메사르탄, 에프로사르탄, 이들의 이성질체 및 이들의 약학적으로 허용 가능한 염 중에서 선택된 하나 이상인 약제학적 제제.The method according to any one of claims 1 to 4, wherein the angiotensin-2 receptor antagonist is losartan, valsartan, telmisartan, irbesartan, candesartan, olmesartan, eprosartan, A pharmaceutical formulation that is one or more selected from isomers and pharmaceutically acceptable salts thereof.
  6. 제1항 내지 제4항 중 어느 한 항에 있어서, 상기 베타 아드레날린 차단제는 알프레놀롤, 아세부토롤, 아모술랄롤, 아로티놀롤, 아테놀롤, 베푸놀롤, 베탁솔롤, 베반톨롤, 비소프롤롤, 보핀돌롤, 부쿠몰롤, 뷔페톨롤, 부푸랄롤, 부니트롤롤, 부프란돌롤, 부토필롤롤, 카라졸롤, 카테올롤, 카르베딜롤, 셀릴프롤롤, 세타몰롤, 클로라놀롤, 딜레발롤, 에파놀롤, 인데놀롤, 라베탈롤, 레보부놀롤, 메핀돌롤, 메티프라놀롤, 메토프롤롤, 모프롤롤, 나돌롤, 나독솔롤, 염산 네비볼롤, 니프라딜롤, 옥스프레놀롤, 퍼부톨롤, 핀돌롤, 프랙톨롤, 프로네탈롤, 프로프라놀롤, 소탈롤, 수피날롤, 탈린돌, 테르타톨롤, 틸리솔롤, 티몰롤, 톨리프롤롤, 지베놀롤, 이들의 이성질체, 및 이들의 약학적으로 허용 가능한 염 중에서 선택된 하나 이상인 약제학적 제제. The method of claim 1, wherein the beta adrenergic blocker is alprenolol, acebutorol, amosulolol, arotinol, atenolol, befunolol, betaxolol, bevantolol, bisoprolol , Bopindolol, bucumolol, buffet tolol, bufuralol, bunitrolol, bufrandolol, butopylolol, carazolol, cateolol, carvedilol, selylprolol, cetamolol, chloranol, dilevalol, epa Nolol, Innolol, Labetalol, Levobunol, Mepindolol, Methipralol, Metoprolol, Moprolol, Nadolol, Nadoxolol, Navivolol, Nipradilol, Oxprenolol, Perbutolol, Pindolol, Fracol One selected from tollol, pronetolol, propranolol, sotalol, supinalol, thaldolol, tertatolol, tilisolol, timolol, toliprolol, givenolol, isomers thereof, and pharmaceutically acceptable salts thereof Pharmaceutical formulations.
  7. 제 1항 내지 제4항 중 어느 한 항에 있어서, 상기 지연방출성 구획은 활성성분 외에 장용성 고분자, 수불용성 중합체, 소수성 화합물, 친수성 고분자 및 이들의 혼합물 중에서 선택된 1종 이상의 방출제어물질을 추가로 포함하는 약제학적 제제. The method according to any one of claims 1 to 4, wherein the delayed-release compartment further comprises at least one release controlling substance selected from an enteric polymer, a water insoluble polymer, a hydrophobic compound, a hydrophilic polymer, and a mixture thereof in addition to the active ingredient. Pharmaceutical formulations comprising.
  8. 제 7 항에 있어서, 상기 방출제어물질은 베타아드레날린 차단제 1중량부에 대하여 0.1 ~ 100 중량부로 포함되는 것인 약제학적 제제. The pharmaceutical formulation of claim 7, wherein the release controlling substance is included in an amount of 0.1 to 100 parts by weight based on 1 part by weight of the beta adrenergic blocker.
  9. 제 7 항에 있어서, 상기 장용성 고분자는 장용성 셀룰로오스 유도체, 장용성 아크릴산계 공중합체, 장용성 폴리메타크릴레이트 공중합체, 장용성 말레인산계 공중합체, 장용성 폴리비닐 유도체, 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상인 약제학적 제제. The method of claim 7, wherein the enteric polymer is one selected from the group consisting of an enteric cellulose derivative, an enteric acrylic acid copolymer, an enteric polymethacrylate copolymer, an enteric maleic acid copolymer, an enteric polyvinyl derivative, and a mixture thereof. Pharmaceutical formulations.
  10. 제9 항에 있어서, 상기 장용성 셀룰로오스 유도체는 히드록시프로필메틸셀룰로오스아세테이트숙시네이트, 히드록시프로필메틸셀룰로오스프탈레이트, 히드록시메틸에틸셀룰로오스프탈레이트, 셀룰로오스아세테이트프탈레이트, 셀룰로오스아세테이트숙시네이트, 셀룰로오스아세테이트말레이트, 셀룰로오스벤조에이트프탈레이트, 셀룰로오스프로피오네이트프탈레이트, 메틸셀룰로오스프탈레이트, 카르복시메틸에틸셀룰로오스, 에틸히드록시에틸셀룰로오스프탈레이트, 메틸히드록시에틸셀룰로오스 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 장용성 아크릴산계 공중합체는 스티렌-아크릴산 공중합체, 아크릴산메틸-아크릴산 공중합체, 아크릴산메틸메타크릴산 공중합체, 아크릴산부틸-스티렌-아크릴산 공중합체, 아크릴산메틸-메타크릴산-아크릴산옥틸공중합체 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 장용성 폴리메타크릴레이트 공중합체는 폴리(메타크릴산-메틸메타크릴레이트) 공중합체, 폴리(메타크릴산ㆍ에틸아크릴레이트)공중합체 및 이들의 혼합물 중에서 선택된 1종이상이고; 상기 장용성 말레인산계 공중합체는 아세트산비닐-말레인산 무수물 공중합체, 스티렌-말레인산 무수물 공중합체, 스티렌-말레인산모노에스테르 공중합체, 비닐메틸에테르-말레인산 무수물 공중합체, 에틸렌-말레인산 무수물 공중합체, 비닐부틸에테르-말레인산 무수물 공중합체, 아크릴로니트릴-아크릴산메틸ㆍ말레인산 무수물 공중합체, 아크릴산부틸-스티렌-말레인산 무수물 공중합체 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 장용성 폴리비닐 유도체는 폴리비닐알콜프탈레이트, 폴리비닐아세테이트프탈레이트, 폴리비닐부티레이트프탈레이트, 폴리비닐아세트아세탈프탈레이트 및 이들의 혼합물 중에서 선택된 1종 이상인 약제학적 제제. The method of claim 9, wherein the enteric cellulose derivative is hydroxypropylmethylcellulose acetate succinate, hydroxypropylmethylcellulose phthalate, hydroxymethylethyl cellulose phthalate, cellulose acetate phthalate, cellulose acetate succinate, cellulose acetate maleate, cellulose benzo One or more selected from eight phthalate, cellulose propionate phthalate, methyl cellulose phthalate, carboxymethylethyl cellulose, ethyl hydroxyethyl cellulose phthalate, methyl hydroxyethyl cellulose and mixtures thereof; The enteric acrylic acid copolymers include styrene-acrylic acid copolymers, methyl acrylate-acrylic acid copolymers, methyl methacrylate acrylates, butyl acrylate-styrene-acrylic acid copolymers, methyl acrylate-methacrylic acid-octyl acrylate copolymers, and these At least one selected from a mixture of; The enteric polymethacrylate copolymer is at least one selected from poly (methacrylic acid-methylmethacrylate) copolymer, poly (methacrylic acid ethyl acrylate) copolymer, and mixtures thereof; The enteric maleic acid-based copolymers include vinyl acetate-maleic anhydride copolymer, styrene-maleic anhydride copolymer, styrene-maleic acid monoester copolymer, vinyl methyl ether-maleic anhydride copolymer, ethylene-maleic anhydride copolymer, vinyl butyl ether- At least one selected from maleic anhydride copolymer, acrylonitrile-methyl acrylate maleic anhydride copolymer, butyl styrene-maleic-maleic anhydride copolymer and mixtures thereof; The enteric polyvinyl derivative is at least one selected from polyvinyl alcohol phthalate, polyvinylacetate phthalate, polyvinyl butyrate phthalate, polyvinylacetacetal phthalate, and mixtures thereof.
  11. 제7항에 있어서, 상기 장용성 고분자가 활성성분1 중량부에 대해서 0.1 중량부 내지 20 중량부인 약제학적 제제. The pharmaceutical preparation according to claim 7, wherein the enteric polymer is 0.1 to 20 parts by weight based on 1 part by weight of the active ingredient.
  12. 제 7 항에 있어서, 상기 수불용성 중합체는 폴리비닐 아세테이트, 수불용성 폴리메타크릴레이트 공중합체, 에틸셀룰로오스, 셀룰로오스 에스테르, 셀룰로오스 에테르, 셀룰로오스 아실레이트, 셀룰로오스 디아실레이트, 셀룰로오스 트리아실레이트, 셀룰로오스 아세테이트, 셀룰로오스 디아세테이트, 셀룰로오스 트리아세테이트 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상인 약제학적 제제. The method of claim 7, wherein the water insoluble polymer is polyvinyl acetate, water insoluble polymethacrylate copolymer, ethyl cellulose, cellulose ester, cellulose ether, cellulose acylate, cellulose dicylate, cellulose triacylate, cellulose acetate, A pharmaceutical formulation which is at least one selected from the group consisting of cellulose diacetate, cellulose triacetate and mixtures thereof.
  13. 제 7항에 있어서, 상기 수불용성 중합체가 활성성분1 중량부에 대해서 0.1 중량부 내지 30 중량부인 약제학적 제제. The pharmaceutical formulation according to claim 7, wherein the water-insoluble polymer is 0.1 to 30 parts by weight based on 1 part by weight of the active ingredient.
  14. 제 12 항에 있어서, 상기 수불용성 중합체가 폴리비닐 아세테이트, 에틸셀룰로오스, 폴리(에틸아크릴레이트, 메틸메타크릴레이트, 트리메틸아미노에틸메타크릴레이트 클로라이드)공중합체, 및 셀룰로오스 아세테이트 중에서 선택된 1종 이상인 약제학적 제제. The pharmaceutical composition of claim 12, wherein the water-insoluble polymer is at least one selected from polyvinyl acetate, ethyl cellulose, poly (ethyl acrylate, methyl methacrylate, trimethylaminoethyl methacrylate chloride) copolymer, and cellulose acetate. Formulation.
  15. 제 7 항에 있어서, 상기 방출제어물질은 수불용성 중합체 및 장용성 고분자 중에서 선택된 1종 이상인 약제학적 제제. 8. The pharmaceutical formulation of claim 7, wherein the release controlling substance is at least one selected from a water insoluble polymer and an enteric polymer.
  16. 제15항에 있어서, 상기 수불용성중합체는 폴리비닐 아세테이트, 에틸셀룰로오스, 폴리(에틸아크릴레이트, 메틸메타크릴레이트, 트리메틸아미노에틸메타크릴레이트 클로라이드)공중합체, 및 셀룰로오스 아세테이트 중에서 선택된 1종 이상이고, 상기 장용성 고분자는 히드록시프로필메틸셀룰로오스프탈레이트 및 아크릴산메틸메타크릴산 공중합체 중에서 선택된 1종 이상인 약제학적 제제. The method of claim 15, wherein the water-insoluble polymer is at least one selected from polyvinyl acetate, ethyl cellulose, poly (ethyl acrylate, methyl methacrylate, trimethylaminoethyl methacrylate chloride) copolymer, and cellulose acetate, The enteric polymer is at least one selected from hydroxypropylmethylcellulose phthalate and methyl methacrylate copolymer.
  17. 제 7 항에 있어서, 상기 소수성 화합물은 지방산 및 지방산 에스테르류, 지방산 알코올류,왁스류, 무기물질 및 이들의 혼합물 중에서 선택된 1종 이상인 약제학적 제제. The pharmaceutical formulation of claim 7, wherein the hydrophobic compound is at least one selected from fatty acids and fatty acid esters, fatty alcohols, waxes, inorganic substances and mixtures thereof.
  18. 제 17 항에 있어서, 상기 지방산 및 지방산 에스테르류로는 글리세릴 팔미토스테아레이트, 글리세릴 스테아레이트, 글리세릴 디스테아레이트, 글리세릴 비헤네이트, 세틸 팔미테이트, 글리세릴 모노 올레이트, 스테아르산 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 지방산 알코올류는 세토스테아릴 알코올, 세틸알코올, 스테아릴알코올 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 왁스류는 카르나우바왁스, 밀납, 미결정왁스 및 이들의 혼합물 중에서 선택된 1종 이상이고 ;상기 무기물질은 탈크, 침강탄산칼슘, 인산일수소칼슘, 산화아연, 산화티탄, 카올린, 벤토나이트, 몬모릴로나이트, 비검 및 이들의 혼합물 중에서 선택된 1종 이상인 약제학적 제제. 18. The method of claim 17, wherein the fatty acids and fatty acid esters are glyceryl palmitostearate, glyceryl stearate, glyceryl distearate, glyceryl bihenate, cetyl palmitate, glyceryl monooleate, stearic acid and At least one selected from a mixture thereof; The fatty acid alcohols are at least one selected from cetostearyl alcohol, cetyl alcohol, stearyl alcohol, and mixtures thereof; The waxes are at least one selected from carnauba wax, beeswax, microcrystalline wax, and mixtures thereof; the inorganic material is talc, precipitated calcium carbonate, calcium dihydrogen phosphate, zinc oxide, titanium oxide, kaolin, bentonite, montmorillonite At least one pharmaceutical agent selected from the group consisting of non-gum and mixtures thereof.
  19. 제7항에 있어서, 소수성 화합물이 활성성분 1 중량부에 대해서 0.1 중량부 ~ 20 중량부인 약제학적 제제. The pharmaceutical formulation according to claim 7, wherein the hydrophobic compound is 0.1 to 20 parts by weight based on 1 part by weight of the active ingredient.
  20. 제 7 항에 있어서, 상기 친수성 고분자는 당류, 셀룰로오스 유도체, 검류, 단백질류, 폴리비닐 유도체, 친수성 폴리메타크릴레이트 공중합체, 폴리에틸렌 유도체, 카르복시비닐공중합체 및 이들의 혼합물 중에서 선택된 1종 이상인 약제학적 제제. The pharmaceutical composition of claim 7, wherein the hydrophilic polymer is at least one selected from sugars, cellulose derivatives, gums, proteins, polyvinyl derivatives, hydrophilic polymethacrylate copolymers, polyethylene derivatives, carboxyvinyl copolymers, and mixtures thereof. Formulation.
  21. 제 20 항에 있어서, 상기 당류는 덱스트린, 폴리덱스트린, 덱스트란, 펙틴 및 펙틴 유도체, 알긴산염, 폴리갈락투론산, 자일란, 아라비노자일란, 아라비노갈락탄, 전분, 히드록시프로필스타치, 아밀로오스, 아밀로펙틴 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 셀룰로오스 유도체는 히드록시프로필메틸셀룰로오스, 히드록시프로필셀룰로오스, 히드록시메틸셀룰로오스, 히드록시에틸셀룰로오스, 메틸셀룰로오스, 카르복시메틸셀룰로오스 나트륨, 히드록시프로필 메틸셀룰로오스 아세테이트 숙시네이트, 히드록시에틸메틸셀룰로오스 및 이들의 혼합물 중에서 선택된 1종 이상이며; 상기 검류는 구아검, 로커스트 콩검, 트라가칸타, 카라기난, 아카시아검, 아라비아검, 젤란검, 잔탄검 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 단백질류는 젤라틴, 카제인, 제인 및 이들의 혼합물 중에서 선택 1종 이상이며; 상기 폴리비닐 유도체는 폴리비닐 알코올, 폴리비닐 피롤리돈, 폴리비닐아세탈디에틸아미노아세테이트 및 이들의 혼합물 중에서 선택된 1종 이상이며; 상기 친수성 폴리메타크릴레이트 공중합체는 폴리(부틸 메타크릴레이트,(2-디메틸아미노에틸)메타크릴레이트-메틸메타크릴레이트) 공중합체, 폴리(메타크릴산-메틸메타크릴레이트) 공중합체 및 폴리(메타크릴산-에틸아크릴레이트) 공중합체, 및 이들의 혼합물 중에서 선택된 1종 이상이고; 상기 폴리에틸렌 유도체는 폴리에틸렌글리콜, 폴리에틸렌 옥사이드 및 이들의 혼합물 중에서 선택된 1종 이상이며; 상기 카르복시비닐 공중합체는 카보머인 것인 약제학적 제제. The method of claim 20, wherein the saccharide is dextrin, polydextrin, dextran, pectin and pectin derivatives, alginate, polygalacturonic acid, xylan, arabinoxylan, arabinogalactan, starch, hydroxypropylstarch, amylose At least one selected from amylopectin and mixtures thereof; The cellulose derivative is hydroxypropyl methyl cellulose, hydroxypropyl cellulose, hydroxymethyl cellulose, hydroxyethyl cellulose, methyl cellulose, carboxymethyl cellulose sodium, hydroxypropyl methyl cellulose acetate succinate, hydroxyethyl methyl cellulose and their At least one selected from a mixture; The gum is at least one selected from guar gum, locust bean gum, tragacanta, carrageenan, acacia gum, arabic gum, gellan gum, xanthan gum and mixtures thereof; The protein is at least one selected from gelatin, casein, zein and mixtures thereof; The polyvinyl derivative is at least one selected from polyvinyl alcohol, polyvinyl pyrrolidone, polyvinyl acetal diethylamino acetate and mixtures thereof; The hydrophilic polymethacrylate copolymers include poly (butyl methacrylate, (2-dimethylaminoethyl) methacrylate-methylmethacrylate) copolymer, poly (methacrylate-methylmethacrylate) copolymer and poly (Methacrylic acid-ethylacrylate) copolymer, and mixtures thereof; The polyethylene derivative is at least one selected from polyethylene glycol, polyethylene oxide and mixtures thereof; The carboxyvinyl copolymer is a carbomer.
  22. 제 7 항에 있어서, 친수성 고분자가 활성성분 1 중량부에 대해서 0.05 중량부 ~ 30 중량부인 약제학적 제제. The pharmaceutical formulation according to claim 7, wherein the hydrophilic polymer is 0.05 part by weight to 30 parts by weight with respect to 1 part by weight of the active ingredient.
  23. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 약제학적 제제는 지연방출성 구획 및 선방출성 구획이 균일하게 혼합된 후 타정하여 얻어지는 2상의 매트릭스 정제 형태인 약제학적 제제. The pharmaceutical formulation according to any one of claims 1 to 4, wherein the pharmaceutical formulation is in the form of a two-phase matrix tablet obtained by tableting after the delayed-release compartment and the prior-release compartment are uniformly mixed.
  24. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 약제학적 제제는 지연방출성 구획으로 이루어진 정제와 상기 정제의 외부를 둘러싸는 선방출성 구획으로 이루어진 필름코팅층으로 구성된 필름코팅정 형태인 약제학적 제제. The pharmaceutical formulation according to any one of claims 1 to 4, wherein the pharmaceutical formulation is in the form of a film-coated tablet consisting of a tablet consisting of a delayed-release compartment and a film-coating layer consisting of a prior-release compartment surrounding the outside of the tablet. Formulation.
  25. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 약제학적 제제는 상기 지연방출성 구획과 상기 선방출성 구획이 층을 이루는 다층정 형태인 약제학적 제제. The pharmaceutical formulation according to any one of claims 1 to 4, wherein the pharmaceutical formulation is in the form of a multilayer tablet in which the delayed-release compartment and the prior-release compartment are layered.
  26. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 제제는 지연방출성 구획으로 이루어진 내핵정과, 상기 내핵정의 외면을 둘러싸고 있는 선방출성 구획으로 이루어진 외층으로 구성된 유핵정 형태인 약제학적 제제. The pharmaceutical preparation according to any one of claims 1 to 4, wherein the preparation is in the form of a nucleated tablet consisting of an inner core consisting of a delayed-release compartment and an outer layer consisting of a prior-release compartment surrounding the outer surface of the inner core tablet.
  27. 제 26 항에 있어서, 상기 유핵정은 삼투성 유핵정인 약제학적 제제. The pharmaceutical formulation of claim 26 wherein the nucleated tablet is an osmotic nucleated tablet.
  28. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 약제학적 제제는 지연방출성 구획으로 이루어진 입자, 과립, 펠렛, 또는 정제와, 선방출성 구획으로 이루어진 입자, 과립, 펠렛, 또는 정제를 포함하는 캡슐제 형태인 약제학적 제제. The pharmaceutical formulation of claim 1, wherein the pharmaceutical formulation comprises particles, granules, pellets, or tablets consisting of delayed-release compartments, and particles, granules, pellets, or tablets consisting of prior-release compartments. A pharmaceutical formulation in the form of a capsule.
  29. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 지연방출성 구획, 또는 상기 선방출성 구획 중 하나 이상의 외부에 코팅층을 추가로 포함하는 약제학적 제제. The pharmaceutical formulation according to any one of claims 1 to 4, further comprising a coating layer on the outside of at least one of the delayed-release compartment or the prior-release compartment.
  30. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 지연방출성 구획은 삼투압 조절제를 포함하며 반투과성막 코팅기제로 코팅된 것인 약제학적 제제. The pharmaceutical formulation according to any one of claims 1 to 4, wherein the delayed-release compartment comprises an osmotic pressure control agent and is coated with a semipermeable membrane coating base.
  31. 제 30 항에 있어서, 상기 삼투압 조절제는 황산마그네슘, 염화마그네슘, 염화나트륨, 염화리튬, 황산칼륨, 황산리튬, 황산나트륨, 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상인 것인 약제학적 제제. 31. The pharmaceutical formulation of claim 30, wherein the osmotic pressure regulator is at least one selected from the group consisting of magnesium sulfate, magnesium chloride, sodium chloride, lithium chloride, potassium sulfate, lithium sulfate, sodium sulfate, and mixtures thereof.
  32. 제 30 항에 있어서, 상기 반투과성막 코팅기제는 폴리비닐 아세테이트, 폴리메타크릴레이트 공중합체, 폴리(에틸아크릴레이트, 메틸 메타크릴레이트) 공중합체, 폴리(에틸아크릴레이트, 메틸 메타크릴레이트, 트리메틸아미노에틸메타크릴레이트 클로라이드)공중합체, 에틸셀룰로오스, 셀룰로오스 에스테르, 셀룰로오스 에테르, 셀룰로오스 아실레이트, 셀룰로오스 디아실레이트, 셀룰로오스 트리아실레이트, 셀룰로오스 아세테이트, 셀룰로오스 디아세테이트, 셀룰로오스 트리아세테이트, 및 이들의 혼합물로 이루어진 군에서 선택된 1종 이상인 것인 약제학적 제제. 31. The method of claim 30, wherein the semipermeable membrane coating base is polyvinyl acetate, polymethacrylate copolymer, poly (ethylacrylate, methyl methacrylate) copolymer, poly (ethylacrylate, methyl methacrylate, trimethylamino Ethyl methacrylate chloride) copolymer, ethyl cellulose, cellulose ester, cellulose ether, cellulose acylate, cellulose dicylate, cellulose triacylate, cellulose acetate, cellulose diacetate, cellulose triacetate, and mixtures thereof At least one pharmaceutical agent selected from.
  33. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 외부에 코팅층을 추가로 포함하는 코팅정 형태인 약제학적 제제. The pharmaceutical formulation according to any one of claims 1 to 4, which is in the form of a coated tablet further comprising a coating layer on the outside.
  34. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 제제는 지연방출성 구획, 및 선방출성 구획을 포함하는 키트 형태인 약제학적 제제. The pharmaceutical formulation according to any one of claims 1 to 4, wherein the formulation is in the form of a kit comprising a delayed release compartment and a prior release compartment.
  35. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 베타아드레날린 차단제는 제제중 1~1400mg포함하는 것인 약제학적 제제.The pharmaceutical formulation according to any one of claims 1 to 4, wherein the beta adrenergic blocker comprises 1 to 1400 mg of the formulation.
  36. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 안지오텐신-2 수용체 길항제는 제제중 1~800mg포함하는 것인 약제학적 제제.The pharmaceutical formulation according to any one of claims 1 to 4, wherein the angiotensin-2 receptor antagonist comprises 1 to 800 mg of the formulation.
  37. 제 1 항 내지 제4항 중 어느 한 항에 있어서, 상기 제제는 저녁투여용인 약제학적 제제.The pharmaceutical formulation of claim 1, wherein the formulation is for evening administration.
  38. 제 1 항 내지 제4항 중 어느 한 항의 제제를 포유류에게 투여하는 단계를 포함하는 심혈관계 질환 치료방법.A method of treating cardiovascular disease, comprising administering the agent of any one of claims 1 to 4 to a mammal.
PCT/KR2009/003969 2008-07-18 2009-07-17 Pharmaceutical preparation WO2010008244A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20080069793 2008-07-18
KR10-2008-0069793 2008-07-18

Publications (2)

Publication Number Publication Date
WO2010008244A2 true WO2010008244A2 (en) 2010-01-21
WO2010008244A3 WO2010008244A3 (en) 2010-05-14

Family

ID=41550875

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2009/003969 WO2010008244A2 (en) 2008-07-18 2009-07-17 Pharmaceutical preparation

Country Status (1)

Country Link
WO (1) WO2010008244A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105012246A (en) * 2015-08-19 2015-11-04 湖北博华农牧科技有限公司 Nano-zinc oxide coating process
CN105030718A (en) * 2015-08-18 2015-11-11 石家庄格瑞药业有限公司 Arotinolol hydrochloride preparation and preparation method thereof
WO2017048033A1 (en) * 2015-09-15 2017-03-23 Chong Kun Dang Pharmaceutical Corp. Pharmaceutical combination preparation
US12031128B2 (en) 2022-04-07 2024-07-09 Battelle Memorial Institute Rapid design, build, test, and learn technologies for identifying and using non-viral carriers

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20050000556A (en) * 2002-05-17 2005-01-05 노파르티스 아게 Combination of angiotensin ii receptor blocker and beta-blocker for secondary prevention of myocardial infarction
KR100582347B1 (en) * 2004-12-30 2006-05-22 한미약품 주식회사 Complex composition of 3-hydroxy-3-methyl glutaryl coa reductase inhibitor and antihypertensive agent, and process for preparing same
KR20070078625A (en) * 2006-01-27 2007-08-01 씨제이 주식회사 Multiple unit type sustained release oral formulation and process for the preparation thereof
KR20080032616A (en) * 2006-10-10 2008-04-15 한올제약주식회사 Combination preparation for cardiovascular disease therapy by chronotherapy theory

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20050000556A (en) * 2002-05-17 2005-01-05 노파르티스 아게 Combination of angiotensin ii receptor blocker and beta-blocker for secondary prevention of myocardial infarction
KR100582347B1 (en) * 2004-12-30 2006-05-22 한미약품 주식회사 Complex composition of 3-hydroxy-3-methyl glutaryl coa reductase inhibitor and antihypertensive agent, and process for preparing same
KR20070078625A (en) * 2006-01-27 2007-08-01 씨제이 주식회사 Multiple unit type sustained release oral formulation and process for the preparation thereof
KR20080032616A (en) * 2006-10-10 2008-04-15 한올제약주식회사 Combination preparation for cardiovascular disease therapy by chronotherapy theory

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105030718A (en) * 2015-08-18 2015-11-11 石家庄格瑞药业有限公司 Arotinolol hydrochloride preparation and preparation method thereof
CN105012246A (en) * 2015-08-19 2015-11-04 湖北博华农牧科技有限公司 Nano-zinc oxide coating process
WO2017048033A1 (en) * 2015-09-15 2017-03-23 Chong Kun Dang Pharmaceutical Corp. Pharmaceutical combination preparation
US12031128B2 (en) 2022-04-07 2024-07-09 Battelle Memorial Institute Rapid design, build, test, and learn technologies for identifying and using non-viral carriers

Also Published As

Publication number Publication date
WO2010008244A3 (en) 2010-05-14

Similar Documents

Publication Publication Date Title
WO2009134057A2 (en) Pharmaceutical formulation containing angiotensin-ii receptor blocker
WO2009125981A9 (en) Pharmaceutical formulation
WO2009104932A2 (en) Composite preparation
WO2009134086A9 (en) Pharmaceutical formulation for treatment of cardiovascular disease
WO2012124973A2 (en) Combined formulation with improved stability
WO2010008203A2 (en) Pharmaceutical formulation containing a calcium channel blocker
CN101005829A (en) Quetiapine formulations
KR20240091095A (en) PHARMACEUTICAL COMBINATION, COMPOSITOIN AND FORMULATION CONTAINING GLUCOKINASE ACTIVATOR AND α-GLUCOSIDASE INHIBITOR, PREPARATION METHODS THEREFOR AND USES THEREOF
WO2011081493A2 (en) Pharmaceutical composition comprising metformin and rosuvastatin
WO2009104939A2 (en) Pharmaceutical preparation
WO2010008244A2 (en) Pharmaceutical preparation
WO2019031898A2 (en) Pharmaceutical composition and method for preparing same
WO2018097629A1 (en) Varenicline sustained-release preparation and production method thereof
WO2009125944A9 (en) Pharmaceutical preparation containing non-dihydropyridine calcium channel blocker and angiotensin-2 receptor blocker
WO2020009352A1 (en) Pharmaceutical preparation containing amorphous dapagliflozin l-proline, and preparation method therefor
WO2009104916A2 (en) Pharmaceutical formulations for the treatment of cardiovascular diseases
WO2020256309A1 (en) Controlled-release composition for oral administration comprising complex of alpha adrenergic blocker compound and clay mineral
WO2015012633A1 (en) Complex formulation containing sustained release metformin and immediate release hmg-coa reductase inhibitor
WO2016209061A1 (en) Composite preparation of mosapride and rabeprazole
WO2012077968A2 (en) Complex formulation comprising lercanidipine hydrochloride and valsartan and method for the preparation thereof
WO2010021473A2 (en) Pharmaceutical formulation
WO2021040257A1 (en) Pharmaceutical formulations comprising sodium palmitoyl-l-prolyl-l-prolyl-glycyl-l-tyrosinate and methods for preparing the same
WO2020130385A1 (en) Pharmaceutical composition containing tamsulosin hydrochloride with excellent acid resistance and preparation method therefor
WO2013169082A1 (en) Bosentan controlled release oral preparation
WO2016052866A1 (en) Solid pharmaceutical composition comprising amlodipine and losartan

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: 09798147

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09798147

Country of ref document: EP

Kind code of ref document: A2