US20100173965A1 - Antifungal composition - Google Patents

Antifungal composition Download PDF

Info

Publication number
US20100173965A1
US20100173965A1 US12/676,345 US67634508A US2010173965A1 US 20100173965 A1 US20100173965 A1 US 20100173965A1 US 67634508 A US67634508 A US 67634508A US 2010173965 A1 US2010173965 A1 US 2010173965A1
Authority
US
United States
Prior art keywords
pharmaceutical composition
group
compound selected
glycol
triacetin
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/676,345
Inventor
Takaaki Masuda
Naoto Nishida
Naoko Kobayashi
Hideaki Sasagawa
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pola Pharma Inc
Nihon Nohyaku Co Ltd
Original Assignee
Pola Pharma Inc
Nihon Nohyaku Co Ltd
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 Pola Pharma Inc, Nihon Nohyaku Co Ltd filed Critical Pola Pharma Inc
Assigned to NIHON NOHYAKU CO., LTD., POLA PHARMA INC. reassignment NIHON NOHYAKU CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KOBAYASHI, NAOKO, MASUDA, TAKAAKI, NISHIDA, NAOTO, SASAGAWA, HIDEAKI
Publication of US20100173965A1 publication Critical patent/US20100173965A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic 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
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/08Antiseborrheics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics

Definitions

  • the present invention relates to a pharmaceutical composition, and more specifically, to a pharmaceutical composition for antifungal use.
  • mycoses In Japan having a hot and humid climatic property, measures against mycoses caused by pathogenic fungi are a particularly important pharmaceutical theme. Reoccurrence of such a disease is repeatedly caused by remaining living microbes even if the disease appears to have been perfectly cured. For example, summer is a season in which the disease easily reoccurs, and thus, repetitive treatments are required. Therefore, an antifungal agent having an excellent medicinal effect has been demanded. Further, simultaneously, mycoses include those which develop on hand and foot portions such as tinea pedis and tinea unguium, those which develop on the body skin such as tinea, and those which develop on the scalp such as seborrheic dermatitis, and there is also a demand for development of a versatile dosage form which can be applied to the respective portions.
  • an antifungal agent having an excellent antifungal effect for example, luliconazole, one of compounds represented by the general formula (1), has attracted attention as a medicament that allows a treatment period to be shortened (for example, see JP 09-100279 A and JP 02-275877 A).
  • Such compounds are also useful for onychomycosis, and a formulation thereof for onychomycosis, which is a hydrogel-like formulation, is also already known (for example, see WO 03/105841).
  • the compounds represented by the general formula (1) can be said to be excellent in antifungal property as well.
  • the compounds represented by the general formula (I) are poorly-soluble in an aqueous medium in terms of solubility, and must be formulated by using a solubilizing agent.
  • a solubilizing agent As a result, the control of a variation in the bioavailability of medicaments to be caused by such a solubilizing agent has been the single and greatest problem in a formulation study.
  • the addition of such a cellulose-based thickener also has an action of enhancing a horny cell layer storage (hereinafter, also referred to as a storage or a retention of a medicament in the horny cell layer).
  • a variation in the compounding amount of such a thickener for viscosity adjustment has also caused a change in the pharmacodynamic property and a change in the bioavailability of a formulation.
  • a medicine is always required to retain bioequivalence, and it can be said that the development of a system capable of compensating such a change in the pharmacodynamic property and of maintaining bioequivalence has been demanded for the formulation design of the compounds represented by the general formula (1).
  • any pharmaceutical composition for antifungal use containing: 1) one or more compounds selected from compounds represented by the general formula (1) below and physiologically acceptable salts thereof; 2) one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol; and 3) one or more compounds selected from glucono- ⁇ -lactone, propylene glycol, glycerin, and lactic acid. Further, there has been no finding that a change in the bioavailability of the compounds represented by the general formula (1) due to a variation in the formulation components can be compensated by adopting such a composition.
  • ingredients such as polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, polyoxyethylene-polyoxypropylene glycol, glucono- ⁇ -lactone, propylene glycol, glycerin, or lactic acid into a pharmaceutical composition for antifungal use for the purpose of serving as an absorption accelerant, a pH stabilizer, or the like (for example, see JP 2004-529935 A, JP 2003-252798 A, JP 2004-529923 A, WO 96/011710, and WO 01/003742).
  • the present invention has been made under such circumstances, and is characterized by providing a system for compensating a change in the bioavailability of compounds represented by the general formula (1) due to a variation in the formulation components.
  • the inventors of the present invention have intensively studied to search a system for compensating a change in the bioavailability of the compounds represented by the general formula (1) due to a variation in the formulation components.
  • the inventors have found that the horny cell layer storability of the compounds represented by the general formula (1) can be controlled by formulating a combination of a ingredient for inhibiting the absorption of the compounds of the general formula (1) to the horny cell layer, i.e., one or more compounds selected from polypropylene glycol, diesters of dibasic acid, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol, and a ingredient for promoting the absorption of the compounds of the general formula (I) to the horny cell layer, i.e., one or more compounds selected from glucono- ⁇ -lactone, propylene glycol, glycerin, and lactic acid
  • a pharmaceutical composition for antifungal use comprising: 1) one or more compounds selected from compounds represented by the general formula (1) and physiologically acceptable salts thereof; 2) one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol; and 3) one or more compounds selected from glucono- ⁇ -lactone, propylene glycol, glycerin, and lactic acid.
  • composition according to the item ⁇ 1> or ⁇ 2> comprising triacetin as one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol.
  • glucono- ⁇ -lactone as one or more compounds selected from glucono- ⁇ -lactone, propylene glycol, glycerin, and lactic acid.
  • composition according to any one of the item ⁇ 1> to ⁇ 5> further comprising ethanol and/or water.
  • ⁇ 7> The pharmaceutical composition according to any one of the item ⁇ 1> to ⁇ 6>, wherein the target disease is seborrheic dermatitis.
  • a method for adjusting the storability in the horny cell layer of compounds represented by the general formula (1) and/or salts thereof in the pharmaceutical composition according to any one of the item ⁇ 1> to ⁇ 7> which comprises decreasing the total content of one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol with respect to the total amount of the pharmaceutical composition, and/or increasing the total content of one or more compounds selected from glucono- ⁇ -lactone, propylene glycol, glycerin, and lactic acid with respect to the total amount of the pharmaceutical composition, in the case where the storability in the horny cell layer should be increased; or increasing the total content of one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexaned
  • a system for compensating a change in the bioavailability of compounds represented by the general formula (1) due to a variation in the formulation components may be provided.
  • the pharmaceutical composition of the present invention is one for antifungal use, and characterized by containing a compound represented by the general formula (1) and/or a salt thereof.
  • halogen when X in the compounds represented by the general formula (I) represents a halogen, preferred examples of the halogen include chlorine, bromine, iodine, and fluorine. Of those, chlorine is particularly preferred.
  • preferred specific examples of the compounds represented by the general formula (1) include (R)-( ⁇ )-(E)-[4-(2,4-dichlorophenyl)-1,3-dithiolane-2-ylidene]-1-imidazolylacetonitrile (luliconazole), (R)-(+)-(E)-[4-(2-chlorophenyl)-1,3-dithiolane-2-ylidene]-1-imidazolylacetonitrile, and (E)-[4-(2-chlorophenyl)-1,3-dithiolane-2-ylidene]-1-imidazolyl acetonitrile (lanoconazole).
  • luliconazole is preferred because it ensures the effect of the present invention to be particularly large.
  • physiologically acceptable salts thereof as described above are not particularly limited as long as the salts are physiologically acceptable, and favorable examples of the salts include: mineral acid salts such as a hydrochloride, a nitrate, a sulfate, and a phosphate; organic acid salts such as a citrate, an oxalate, a lactate, and an acetate; and sulfate-containing salts such as a mesylate and a tosylate.
  • a hydrochloride is more preferred in terms of safety and solubility.
  • One compound of compounds represented by the general formula (1) and/or physiologically acceptable salts thereof may be used alone, or two or more compounds thereof may also be used in combination.
  • the preferred total content of the compounds represented by the general formula (1) and/or physiologically acceptable salts thereof in the pharmaceutical composition for antifungal use of the present invention is preferably 0.01 to 20 w/v % and more preferably 0.1 to 10 w/v % with respect to the total amount of the pharmaceutical composition.
  • the amount of such compounds may be determined on the basis of solubility and formulation property thereof.
  • the pharmaceutical composition of the present invention contains, as an essential ingredient, one or more compounds selected from triacetin, polypropylene glycol, diesters of dibasic acids, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol, in addition to the compound represented by the general formula (1) and/or the salt thereof.
  • the polypropylene glycol the polymerization degree thereof is preferably 1000 or more, and more preferably 1500 or more, and the upper limit value of the polymerization degree is preferably 10,000, and more preferably 5000.
  • dibasic acids constituting the diesters of dibasic acids include tartaric acid, adipic acid, sebacic acid and succinic acid, and adipic acid or sebacic acid is particularly preferred.
  • ester moieties thereof preferred is a hydrocarbon group having 1 to 4 carbon atoms, and specifically, favorable examples of the ester moieties include a methyl ester, an ethyl ester, a propyl ester, an isopropyl ester, a butyl ester, a sec-butyl ester and a tert-butyl ester, and preferred examples thereof include an ethyl ester and an isopropyl ester.
  • diisopropyl adipate and diethyl sebacate are preferred.
  • the polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol have actions as solvents that stably dissolve the general formula (1), and as inhibitors of the horny cell layer storability of the compounds represented by the general formula (1).
  • each of such ingredients is incorporated at preferably 0.01 to 60 w/v % and more preferably 0.1 to 50 w/v % with respect to the total amount of the pharmaceutical composition.
  • the content is preferably set up to be a content that can be either decreased or increased.
  • Any of such ingredients also has a subsidiary effect of improving the preservation stability of the compounds represented by the general formula (1) even under a harsh condition of preservation at 60° C. for 1 week.
  • the pharmaceutical composition of the present invention is characterized by containing one or more compounds selected from glucono- ⁇ -lactone, propylene glycol, glycerin, and lactic acid, in addition to the compound represented by the general formula (1) and/or the salt thereof.
  • Such ingredients are solvents that stably dissolve the general formula (1) in many cases, and each have an action as an accelerator of the transdermal absorption of the compounds represented by the general formula (1).
  • each of such ingredients is incorporated at preferably 0.01 to 60 w/v % and more preferably 0.1 to 50 w/v % with respect to the total amount of the pharmaceutical composition.
  • the content is preferably set up to be a content that can be either decreased or increased. Any of such ingredients also has a subsidiary effect for improving the preservation stability of the compounds represented by the general formula (1) even under a harsh condition of preservation at 60° C. for 1 week.
  • the pharmaceutical composition of the present invention is one for antifungal use, and contains the above-mentioned essential ingredients.
  • the pharmaceutical composition of the present invention can be applied for any formulation with an dosage form known in an antifungal formulation, which formulation can contain the above-mentioned essential ingredients, however, is preferably applied for a skin agent for external use in view of an effect of “capable of controlling the absorbability to the horny cell layer”.
  • the pharmaceutical composition of the present invention is preferably formulated into the form of a single-phase solution without containing any flammable solvent such as methyl ethyl ketone, acetone, and ethyl acetate other than ethanol because many of the essential ingredients to be incorporated in the pharmaceutical composition of the present invention have a solvent property or an action of promoting dissolution, superior in an action of inhibiting the absorption and an action of promoting the absorption to the horny cell layer.
  • a cellulose-based thickener such as hydroxypropylcellulose, carboxymethylcellulose, hydroxypropylmethylcellulose, and hydroxyethylcellulose has effects of increasing a viscosity, inhibiting dripping, and enhancing usability, and also has an effect of enhancing the absorbability of a medicament to the horny cell layer. Therefore, in the viscosity adjustment of a system, in some cases, the fine adjustment of the content of such an ingredient causes a variation in horny cell layer storability, so that the maintenance of the bioequivalence becomes difficult.
  • the bioequivalence can be maintained by applying the pharmaceutical composition of the present invention to a system containing such an ingredient, and adjusting the amount ratio of the above-mentioned two essential ingredients involved in the absorbability to the horny cell layer. Accordingly, it is particularly preferred that the pharmaceutical composition of the present invention be applied for such a dosage form.
  • the term “single-phase solution” as used herein refers to a dissolved liquid substance in which no white turbidity is observed and neither liquid crystal nor microcrystal is observed under a polarized light.
  • such a viscous solution form can be administered without being spread from a site to be administered even if, for example, being administered to the hair line and the like, and hence is preferably applied as a formulation to be administered to such a site, i.e., in a medicine for antifungal use, a treatment drug for seborrheic dermatitis such as a therapeutic drug for seborrheic dermatitis and a preventive drug for seborrheic dermatitis.
  • the formulation can be performed without incorporating a solvent such as acetone and methyl ethyl ketone other than ethanol because many of the ingredients for promoting the absorption to the horny cell layer and the ingredients for inhibiting the absorption to the horny cell layer as essential ingredients have an excellent solvent effect or an action for promoting dissolution. Therefore, for a treatment drug for seborrheic dermatitis such as a therapeutic drug for seborrheic dermatitis and a preventive drug for seborrheic dermatitis to be administered to a site close to eyes, the probability that a volatile ingredient derived from a solvent irritates eyes is reduced, resulting in an advantageous property.
  • a solvent such as acetone and methyl ethyl ketone other than ethanol
  • the pharmaceutical composition of the present invention may contain, apart from the above ingredients, an arbitrary ingredient generally used for formulation.
  • an arbitrary ingredient include: oils and waxes such as macadamia nut oil, avocado oil, corn oil, olive oil, rapeseed oil, sesame oil, castor oil, safflower oil, cottonseed oil, jojoba oil, coconut oil, palm oil, liquid lanolin, hydrogenated coconut oil, hydrogenated oil, Japan wax, hydrogenated castor oil, beeswax, candelilla wax, carnauba wax, ibota wax, lanolin, reduced lanolin, hard lanolin, and jojoba wax; hydrocarbons such as liquid paraffin, squalane, pristane, ozokerite, paraffin, ceresin, vaseline, and microcrystalline wax; higher fatty acids such as oleic acid, isostearic acid, lauric acid, myristic acid, palmitic acid, stearic acid, behenic acid, and
  • ingredients which are preferably contained in the pharmaceutical composition of the present invention include thickeners such as hydroxypropylcellulose, hydroxypropylmethylcellulose, carboxymethylcellulose, ethylcellulose, and xanthane gum. This is because such ingredients act as variation factors of the bioavailability as well as variation factors of physical properties of the formulation, and as a result, the effect of the pharmaceutical composition of the present invention may be thoroughly exhibited.
  • the pharmaceutical composition of the present invention can be produced by treating those ingredients in accordance with a conventional method.
  • a method for adjusting the bioequivalence of the pharmaceutical composition of the present invention is characterized in that, in terms of the absorbability to the horny cell layer, the pharmaceutical composition is allowed to retain a constant absorption property to the horny cell layer at all times,
  • the phrase “maintaining bioequivalence” as used herein refers to keeping the absorption property of the compounds represented by the general formula (1) to the horny cell layer constant.
  • the content of this ingredient is adjusted in order to adjust the irregularity from lot to lot of the thickener ingredient, andmodulate a viscosity.
  • an operation for increasing the total content of the ingredient for promoting the absorption to the horny cell layer with respect to the total amount of the pharmaceutical composition there is performed an operation for decreasing the total content of the ingredient for inhibiting the absorption to the horny cell layer with respect to the total amount of the pharmaceutical composition.
  • the degree of this decrease or increase is preferably calculated and determined from a calibration curve which has been preliminarily drawn on a two-dimensional coordinate by using the mass ratio of the ingredient for promoting the absorption to the horny cell layer and the ingredient for inhibiting transdermal absorption as one axis, and the degree of the absorption to the horny cell layer as the other axis, for example.
  • a corrected value may be converted to a mathematical formula by, for example, a multiple regression analysis, and the mathematical formula may also be utilized.
  • the bioequivalence can be maintained without being affected by the irregularity from lot to lot of a formulation ingredient by adjusting the mixing ratio of the ingredient for promoting the absorption to the horny cell layer and the ingredient for inhibiting the absorption to the horny cell layer.
  • Skin agents 1 to 5 for external use each of which is the pharmaceutical composition of the present invention, were prepared in accordance with the following prescription. That is, luliconazole was dissolved by adding an appropriate amount of ethanol, and to the resulting solution, triacetin and glucono- ⁇ -lactone were added and uniformly dispersed. After that, hydroxypropylcellulose was gradually added, and water was further added to obtain homogenous skin agents for external use.
  • any of those agents had a viscosity of 100 to 800 mPa ⁇ s (viscometer: RE80R manufactured by Toki Sangyo Co., Ltd., and conditions for viscosity measurement: cone angle; 0.8°, cone radius; 2.4 cm, rotation frequency; 10 rpm, and temperature; 25° C.), was not so dripping, and thus was suited as a formulation for seborrheic dermatitis to be administered to the hair line and the like. Further, those agents were free of ingredients strongly exhibiting solvent characteristics likemethyl ethyl ketone (MEK) and acetone, did not provide any strong temporary irritation feeling, and could be suitably used for tinea pedis and the like as well.
  • MEK methyl ethyl ketone
  • the skin agent for external use was applied at room temperature, and the whole was left out at room temperature for 2 hours. After that, the skin agent for external use was wiped out from the applied site, and further, the applied site was subjected to 5 times of tape stripping with a cellophane tape. Thereafter, the plantar part was excised, and the horny cell layer was peeled off, formed into a piece having a certain size, and evaporated to dryness under reduced pressure for 24 hours. To the obtained dried product, methanol was added, luliconazole as a measurement target was extracted in an ultrasonicator, and filtered through a filter.
  • Skin agent 6 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription.
  • the concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 4.532 ⁇ g/cm 2
  • the concentration in Comparative Example 1 in which propylene glycol in the preparation was replaced by ethanol was 0.221 ⁇ g/cm 2 .
  • the S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 1.65% in Skin agent 6 for external use, which was higher compared with 1.45% in Comparative Example 1.
  • Propylene glycol is found to be usable as an ingredient for promoting the absorption to the horny cell layer.
  • Skin agent 7 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription.
  • the concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.584 ⁇ g/cm 2
  • the concentration in Comparative Example 2 in which 2-ethyl-1,3-hexanediol in the preparation was replaced by ethanol was 0.950 ⁇ g/cm 2 .
  • the S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 1.06% in Skin agent 7 for external use, which was higher compared with 0.38% in Comparative Example 2.
  • 2-ethyl-1,3-hexanediol is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • Skin agent 8 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription.
  • the concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.736 ⁇ g/cm 2
  • the concentration in Comparative Example 3 in which concentrated glycerin (100% glycerin) in the preparation was replaced by ethanol was 0.345 ⁇ g/cm 2 .
  • the S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.79% in Skin agent 8 for external use, which was higher compared with 0.47% in Comparative Example 3. Concentrated glycerin is found to be usable as an ingredient for promoting the absorption to the horny cell layer.
  • Skin agent 9 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription.
  • the concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 2.018 ⁇ g/cm 2
  • the concentration in Comparative Example 1 in which lactic acid in the preparation was replaced by ethanol was 0.221 ⁇ g/cm 2 .
  • the S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.14% in Skin agent 9 for external use, which was lower compared with 1.45% in Comparative Example 1. Lactic acid is found to be usable as an ingredient for promoting the absorption to the horny cell layer.
  • Skin agent 10 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription.
  • the concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.358 ⁇ g/cm 2
  • the concentration in Comparative Example 2 in which polyoxyethylene (20)-polyoxypropylene (20) glycol in the preparation was replaced by ethanol was 0.950 ⁇ g/cm 2 .
  • the S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 1.26% in Skin agent 10 for external use, which was higher compared with 0.38% in Comparative Example 2.
  • Polyoxyethylene (20)-polyoxypropylene (20) glycol is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • Skin agent 11 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription.
  • the concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.148 ⁇ g/cm 2
  • the concentration in Comparative Example 1 in which lauromacrogol in the preparation was replaced by ethanol was 0.221 ⁇ g/cm 2 .
  • the S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.19% in Skin agent 11 for external use, which was lower compared with 1.45% in Comparative Example 1. Lauromacrogol is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • Skin agent 12 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription.
  • the concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.199 ⁇ g/cm 2
  • the concentration in Comparative Example 1 in which polypropylene glycol 2000 in the preparation was replaced by ethanol was 0.221 ⁇ g/cm 2 .
  • the S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.78% in Skin agent 12 for external use, which was lower compared with 1.45% in Comparative Example 1.
  • Polypropylene glycol 2000 is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • Skin agent 13 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription.
  • the concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.306 ⁇ g/cm 2
  • the concentration in Comparative Example 2 in which diethyl sebacate in the preparation was replaced by ethanol was 0.950 ⁇ g/cm 2 .
  • the S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.14% in Skin agent 13 for external use, which was lower compared with 0.38% in Comparative Example 2. Diethyl sebacate is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • the present invention is applicable to a pharmaceutical composition such as a skin agent for external use for seborrheic dermatitis.

Abstract

Disclosed is a pharmaceutical composition for antifungal use, comprising: 1) one or more compounds selected from compounds represented by the general formula (1) below and physiologically acceptable salts thereof; 2) one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol; and 3) one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid. General formula (1) (In the formula, X represents a halogen or hydrogen).
Figure US20100173965A1-20100708-C00001
wherein X represents a halogen or hydrogen.

Description

    TECHNICAL FIELD
  • The present invention relates to a pharmaceutical composition, and more specifically, to a pharmaceutical composition for antifungal use.
  • BACKGROUND ART
  • In Japan having a hot and humid climatic property, measures against mycoses caused by pathogenic fungi are a particularly important pharmaceutical theme. Reoccurrence of such a disease is repeatedly caused by remaining living microbes even if the disease appears to have been perfectly cured. For example, summer is a season in which the disease easily reoccurs, and thus, repetitive treatments are required. Therefore, an antifungal agent having an excellent medicinal effect has been demanded. Further, simultaneously, mycoses include those which develop on hand and foot portions such as tinea pedis and tinea unguium, those which develop on the body skin such as tinea, and those which develop on the scalp such as seborrheic dermatitis, and there is also a demand for development of a versatile dosage form which can be applied to the respective portions.
  • As an antifungal agent having an excellent antifungal effect, for example, luliconazole, one of compounds represented by the general formula (1), has attracted attention as a medicament that allows a treatment period to be shortened (for example, see JP 09-100279 A and JP 02-275877 A). Such compounds are also useful for onychomycosis, and a formulation thereof for onychomycosis, which is a hydrogel-like formulation, is also already known (for example, see WO 03/105841). Thus, the compounds represented by the general formula (1) can be said to be excellent in antifungal property as well. However, the compounds represented by the general formula (I) are poorly-soluble in an aqueous medium in terms of solubility, and must be formulated by using a solubilizing agent. As a result, the control of a variation in the bioavailability of medicaments to be caused by such a solubilizing agent has been the single and greatest problem in a formulation study. In particular, in the case of allowing a cellulose-based thickener or the like to coexist for viscosity adjustment, the addition of such a cellulose-based thickener also has an action of enhancing a horny cell layer storage (hereinafter, also referred to as a storage or a retention of a medicament in the horny cell layer). Thus, in some cases, a variation in the compounding amount of such a thickener for viscosity adjustment has also caused a change in the pharmacodynamic property and a change in the bioavailability of a formulation. A medicine is always required to retain bioequivalence, and it can be said that the development of a system capable of compensating such a change in the pharmacodynamic property and of maintaining bioequivalence has been demanded for the formulation design of the compounds represented by the general formula (1).
  • Meanwhile, there has not been known any pharmaceutical composition for antifungal use containing: 1) one or more compounds selected from compounds represented by the general formula (1) below and physiologically acceptable salts thereof; 2) one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol; and 3) one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid. Further, there has been no finding that a change in the bioavailability of the compounds represented by the general formula (1) due to a variation in the formulation components can be compensated by adopting such a composition.
  • It should be noted that, there are already known techniques of incorporating ingredients such as polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, polyoxyethylene-polyoxypropylene glycol, glucono-δ-lactone, propylene glycol, glycerin, or lactic acid into a pharmaceutical composition for antifungal use for the purpose of serving as an absorption accelerant, a pH stabilizer, or the like (for example, see JP 2004-529935 A, JP 2003-252798 A, JP 2004-529923 A, WO 96/011710, and WO 01/003742).
  • Figure US20100173965A1-20100708-C00002
  • wherein X represents a halogen or hydrogen.
  • Figure US20100173965A1-20100708-C00003
  • DISCLOSURE OF THE INVENTION Technical Problem
  • The present invention has been made under such circumstances, and is characterized by providing a system for compensating a change in the bioavailability of compounds represented by the general formula (1) due to a variation in the formulation components.
  • In view of such circumstances, the inventors of the present invention have intensively studied to search a system for compensating a change in the bioavailability of the compounds represented by the general formula (1) due to a variation in the formulation components. As a result, the inventors have found that the horny cell layer storability of the compounds represented by the general formula (1) can be controlled by formulating a combination of a ingredient for inhibiting the absorption of the compounds of the general formula (1) to the horny cell layer, i.e., one or more compounds selected from polypropylene glycol, diesters of dibasic acid, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol, and a ingredient for promoting the absorption of the compounds of the general formula (I) to the horny cell layer, i.e., one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid, and changing the combination of those two ingredients. Thus, the present invention has been completed. That is, the present invention is as follows.
  • <1> A pharmaceutical composition for antifungal use, comprising: 1) one or more compounds selected from compounds represented by the general formula (1) and physiologically acceptable salts thereof; 2) one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol; and 3) one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid.
  • <2> The pharmaceutical composition according to the item <1>, wherein the compound represented by the general formula (1) is luliconazole.
  • <3> The pharmaceutical composition according to the item <1> or <2>, comprising triacetin as one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol.
  • <4> The pharmaceutical composition according to anyone of the item <1> to <3>, comprising glucono-δ-lactone as one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid.
  • <5> The pharmaceutical composition according to anyone of the item <1> to <4>, wherein the pharmaceutical composition for antifungal use is in the form of a single-phase solution.
  • <6> The pharmaceutical composition according to any one of the item <1> to <5>, further comprising ethanol and/or water.
  • <7> The pharmaceutical composition according to any one of the item <1> to <6>, wherein the target disease is seborrheic dermatitis.
  • <8> A method for adjusting the storability in the horny cell layer of compounds represented by the general formula (1) and/or salts thereof in the pharmaceutical composition according to any one of the item <1> to <7>, which comprises decreasing the total content of one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol with respect to the total amount of the pharmaceutical composition, and/or increasing the total content of one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid with respect to the total amount of the pharmaceutical composition, in the case where the storability in the horny cell layer should be increased; or increasing the total content of one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol with respect to the total amount of the pharmaceutical composition, and/or decreasing the total content of one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid with respect to the total amount of the pharmaceutical composition, in the case where the storability in the horny cell layer should be decreased.
  • EFFECT OF THE INVENTION
  • According to the present invention, there may be provided a system for compensating a change in the bioavailability of compounds represented by the general formula (1) due to a variation in the formulation components.
  • BEST MODE FOR CARRYING OUT THE INVENTION
  • <1> Compounds Represented by General Formula (1) as Essential Ingredients in Pharmaceutical Composition of Present Invention
  • The pharmaceutical composition of the present invention is one for antifungal use, and characterized by containing a compound represented by the general formula (1) and/or a salt thereof.
  • When X in the compounds represented by the general formula (I) represents a halogen, preferred examples of the halogen include chlorine, bromine, iodine, and fluorine. Of those, chlorine is particularly preferred.
  • In addition, preferred specific examples of the compounds represented by the general formula (1) include (R)-(−)-(E)-[4-(2,4-dichlorophenyl)-1,3-dithiolane-2-ylidene]-1-imidazolylacetonitrile (luliconazole), (R)-(+)-(E)-[4-(2-chlorophenyl)-1,3-dithiolane-2-ylidene]-1-imidazolylacetonitrile, and (E)-[4-(2-chlorophenyl)-1,3-dithiolane-2-ylidene]-1-imidazolyl acetonitrile (lanoconazole). Those compounds are known compounds and their production methods and antifungal properties have already been known (for example, see JP 62-93227 A). Of those, luliconazole is preferred because it ensures the effect of the present invention to be particularly large.
  • Further, the “physiologically acceptable salts thereof” as described above are not particularly limited as long as the salts are physiologically acceptable, and favorable examples of the salts include: mineral acid salts such as a hydrochloride, a nitrate, a sulfate, and a phosphate; organic acid salts such as a citrate, an oxalate, a lactate, and an acetate; and sulfate-containing salts such as a mesylate and a tosylate. A hydrochloride is more preferred in terms of safety and solubility. One compound of compounds represented by the general formula (1) and/or physiologically acceptable salts thereof may be used alone, or two or more compounds thereof may also be used in combination. The preferred total content of the compounds represented by the general formula (1) and/or physiologically acceptable salts thereof in the pharmaceutical composition for antifungal use of the present invention is preferably 0.01 to 20 w/v % and more preferably 0.1 to 10 w/v % with respect to the total amount of the pharmaceutical composition. The amount of such compounds may be determined on the basis of solubility and formulation property thereof.
  • <2> Polypropylene Glycol, Diesters of Dibasic Acids, Triacetin, 2-ethyl-1,3-hexanediol, Lauromacrogol, and Polyoxyethylene-Polyoxypropylene Glycol to be Incorporated in Pharmaceutical Composition of Present Invention
  • The pharmaceutical composition of the present invention contains, as an essential ingredient, one or more compounds selected from triacetin, polypropylene glycol, diesters of dibasic acids, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol, in addition to the compound represented by the general formula (1) and/or the salt thereof. As for the polypropylene glycol, the polymerization degree thereof is preferably 1000 or more, and more preferably 1500 or more, and the upper limit value of the polymerization degree is preferably 10,000, and more preferably 5000. Preferred examples of dibasic acids constituting the diesters of dibasic acids include tartaric acid, adipic acid, sebacic acid and succinic acid, and adipic acid or sebacic acid is particularly preferred. As the ester moieties thereof, preferred is a hydrocarbon group having 1 to 4 carbon atoms, and specifically, favorable examples of the ester moieties include a methyl ester, an ethyl ester, a propyl ester, an isopropyl ester, a butyl ester, a sec-butyl ester and a tert-butyl ester, and preferred examples thereof include an ethyl ester and an isopropyl ester. Specifically, preferred are diisopropyl adipate and diethyl sebacate. In the pharmaceutical composition of the present invention, the polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol have actions as solvents that stably dissolve the general formula (1), and as inhibitors of the horny cell layer storability of the compounds represented by the general formula (1). In order to exert such actions, each of such ingredients is incorporated at preferably 0.01 to 60 w/v % and more preferably 0.1 to 50 w/v % with respect to the total amount of the pharmaceutical composition. In particular, the content is preferably set up to be a content that can be either decreased or increased. Any of such ingredients also has a subsidiary effect of improving the preservation stability of the compounds represented by the general formula (1) even under a harsh condition of preservation at 60° C. for 1 week.
  • <3> Glucono-δ-Lactone, Propylene Glycol, Glycerin, and Lactic Acid to be Incorporated in Pharmaceutical Composition of Present Invention
  • The pharmaceutical composition of the present invention is characterized by containing one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid, in addition to the compound represented by the general formula (1) and/or the salt thereof. Such ingredients are solvents that stably dissolve the general formula (1) in many cases, and each have an action as an accelerator of the transdermal absorption of the compounds represented by the general formula (1). In order to exert such actions, each of such ingredients is incorporated at preferably 0.01 to 60 w/v % and more preferably 0.1 to 50 w/v % with respect to the total amount of the pharmaceutical composition. In particular, the content is preferably set up to be a content that can be either decreased or increased. Any of such ingredients also has a subsidiary effect for improving the preservation stability of the compounds represented by the general formula (1) even under a harsh condition of preservation at 60° C. for 1 week.
  • <4> Pharmaceutical Composition of Present Invention
  • The pharmaceutical composition of the present invention is one for antifungal use, and contains the above-mentioned essential ingredients. Without any particular limitation, the pharmaceutical composition of the present invention can be applied for any formulation with an dosage form known in an antifungal formulation, which formulation can contain the above-mentioned essential ingredients, however, is preferably applied for a skin agent for external use in view of an effect of “capable of controlling the absorbability to the horny cell layer”. The pharmaceutical composition of the present invention is preferably formulated into the form of a single-phase solution without containing any flammable solvent such as methyl ethyl ketone, acetone, and ethyl acetate other than ethanol because many of the essential ingredients to be incorporated in the pharmaceutical composition of the present invention have a solvent property or an action of promoting dissolution, superior in an action of inhibiting the absorption and an action of promoting the absorption to the horny cell layer. A cellulose-based thickener such as hydroxypropylcellulose, carboxymethylcellulose, hydroxypropylmethylcellulose, and hydroxyethylcellulose has effects of increasing a viscosity, inhibiting dripping, and enhancing usability, and also has an effect of enhancing the absorbability of a medicament to the horny cell layer. Therefore, in the viscosity adjustment of a system, in some cases, the fine adjustment of the content of such an ingredient causes a variation in horny cell layer storability, so that the maintenance of the bioequivalence becomes difficult. Thus, the bioequivalence can be maintained by applying the pharmaceutical composition of the present invention to a system containing such an ingredient, and adjusting the amount ratio of the above-mentioned two essential ingredients involved in the absorbability to the horny cell layer. Accordingly, it is particularly preferred that the pharmaceutical composition of the present invention be applied for such a dosage form. It should be noted that the term “single-phase solution” as used herein refers to a dissolved liquid substance in which no white turbidity is observed and neither liquid crystal nor microcrystal is observed under a polarized light. In addition, such a viscous solution form can be administered without being spread from a site to be administered even if, for example, being administered to the hair line and the like, and hence is preferably applied as a formulation to be administered to such a site, i.e., in a medicine for antifungal use, a treatment drug for seborrheic dermatitis such as a therapeutic drug for seborrheic dermatitis and a preventive drug for seborrheic dermatitis.
  • Further, in the above-mentioned composition, the formulation can be performed without incorporating a solvent such as acetone and methyl ethyl ketone other than ethanol because many of the ingredients for promoting the absorption to the horny cell layer and the ingredients for inhibiting the absorption to the horny cell layer as essential ingredients have an excellent solvent effect or an action for promoting dissolution. Therefore, for a treatment drug for seborrheic dermatitis such as a therapeutic drug for seborrheic dermatitis and a preventive drug for seborrheic dermatitis to be administered to a site close to eyes, the probability that a volatile ingredient derived from a solvent irritates eyes is reduced, resulting in an advantageous property.
  • The pharmaceutical composition of the present invention may contain, apart from the above ingredients, an arbitrary ingredient generally used for formulation. Favorable examples of such an arbitrary ingredient include: oils and waxes such as macadamia nut oil, avocado oil, corn oil, olive oil, rapeseed oil, sesame oil, castor oil, safflower oil, cottonseed oil, jojoba oil, coconut oil, palm oil, liquid lanolin, hydrogenated coconut oil, hydrogenated oil, Japan wax, hydrogenated castor oil, beeswax, candelilla wax, carnauba wax, ibota wax, lanolin, reduced lanolin, hard lanolin, and jojoba wax; hydrocarbons such as liquid paraffin, squalane, pristane, ozokerite, paraffin, ceresin, vaseline, and microcrystalline wax; higher fatty acids such as oleic acid, isostearic acid, lauric acid, myristic acid, palmitic acid, stearic acid, behenic acid, and undecylenic acid; higher alcohols such as cetyl alcohol, stearyl alcohol, isostearylalcohol, behenyl alcohol, octyldodecanol, myristyl alcohol, and cetostearyl alcohol; synthetic ester oils such as cetyl isooctanoate, isopropyl myristate, hexyldecyl isostearate, cetyl lactate, diisostearyl malate, ethylene glycol di-2-ethyl hexanoate, neopentyl glycol dicaprate, glyceryl di-2-heptyl undecanoate, glyceryl tri-2-ethylhexanoate, trimethylolpropane tri-2-ethylhexanoate, trimethylolpropane triisostearate, and pentaerythrityl tetra-2-ethylhexanoate; chain polysiloxanes such as dimethylpolysiloxane, methylphenylpolysiloxane, and diphenylpolysiloxane; cyclic polysiloxanes such as octamethylcyclotetrasiloxane, decamethylcyclopentasiloxane, and dodecamethylcyclohexanesiloxane; oil solutions such as silicone oils including modified polysiloxanes such as amino-modified polysiloxane, polyether-modified polysiloxane, alkyl-modified polysiloxane, and fluorine-modified polysiloxane; anionic surfactants such as fatty acid soaps (such as sodium laurate and sodium palmitate), potassium lauryl sulfate, and triethanolamine alkylether sulfate; cationic surfactants such as stearyl trimethyl ammonium chloride, benzalkonium chloride, and laurylamine oxide; amphoteric surfactants such as imidazoline-based amphoteric surfactants (such as a 2-cocoyl-2-imidazoliniumhydroxide-1-carboxyethyloxy-2-sodium salt), betaine-based surfactants (such as alkyl betaine, amide betaine, and sulfo betaine), and acylmethyl taurine; nonionic surfactants such as sorbitan fatty acid esters (such as sorbitan monostearate and sorbitan sesquioleate), glycerin fatty acids (such as glyceryl monostearate), propyleneglycol fatty acid esters (such as propyleneglycol monostearate), hydrogenated castor oil derivatives, glycerol alkyl ether, POE sorbitan fatty acid esters (such as POE sorbitan monooleate and polyoxyethylene sorbitan monostearate), POE sorbit fatty acid esters (such as POE-sorbit monolaurate), POE glycerol fatty acid esters (such as POE-glyceryl monoisostearate), POE fatty acid esters (such as polyethyleneglycol monooleate and POE distearate), POE alkyl ethers (such as POE2-octyldodecyl ether), POE alkyl phenyl ethers (such as POE nonylphenyl ether), POE/POP alkyl ethers (such as POE/POP2-decyltetradecyl ether), types of tetronic (registered trademark), POE castor oil/hydrogenated castor oil derivatives (such as POE castor oil and POE hydrogenated castor oil), sucrose fatty acid ester, and alkyl glycoside; polyhydric alcohols such as polyethylene glycol, 1,3-butylene glycol, erythritol, sorbitol, xylitol, maltitol, dipropylene glycol, diglycerin, isopreneglycol, 1,2-pentanediol, 2,4-hexanediol, 1,2-hexanediol, and 1,2-octanediol; moisturizing ingredients such as sodium pyrrolidone carboxylate, and sodium lactate; thickeners such as hydroxypropylcellulose, hydroxypropylmethylcellulose, a carboxyvinyl polymer which may be alkyl-modified and/or a salt thereof, carboxymethylcellulose, ethylcellulose, and xanthane gum; pH adjusters such as a buffer salt; and solvents such as benzyl alcohol and ethylene glycol salicylate. Of those ingredients, preferred examples of the ingredients which are preferably contained in the pharmaceutical composition of the present invention include thickeners such as hydroxypropylcellulose, hydroxypropylmethylcellulose, carboxymethylcellulose, ethylcellulose, and xanthane gum. This is because such ingredients act as variation factors of the bioavailability as well as variation factors of physical properties of the formulation, and as a result, the effect of the pharmaceutical composition of the present invention may be thoroughly exhibited. The pharmaceutical composition of the present invention can be produced by treating those ingredients in accordance with a conventional method.
  • <5> Method for Adjusting Bioequivalence of Pharmaceutical Composition of Present Invention
  • A method for adjusting the bioequivalence of the pharmaceutical composition of the present invention is characterized in that, in terms of the absorbability to the horny cell layer, the pharmaceutical composition is allowed to retain a constant absorption property to the horny cell layer at all times,
  • by taking a balance of the amount of the ingredient for promoting the absorption selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid and the amount of the ingredient for inhibiting the absorption selected from polypropylene glycols, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol. It should be noted that the phrase “maintaining bioequivalence” as used herein refers to keeping the absorption property of the compounds represented by the general formula (1) to the horny cell layer constant. The content of this ingredient is adjusted in order to adjust the irregularity from lot to lot of the thickener ingredient, andmodulate a viscosity. As a result, when the absorbability of a medicament to the horny cell layer is reduced, and thus, the absorbability to the horny cell layer must be increased, there is performed an operation for increasing the total content of the ingredient for promoting the absorption to the horny cell layer with respect to the total amount of the pharmaceutical composition, and/or an operation for decreasing the total content of the ingredient for inhibiting the absorption to the horny cell layer with respect to the total amount of the pharmaceutical composition. The degree of this decrease or increase is preferably calculated and determined from a calibration curve which has been preliminarily drawn on a two-dimensional coordinate by using the mass ratio of the ingredient for promoting the absorption to the horny cell layer and the ingredient for inhibiting transdermal absorption as one axis, and the degree of the absorption to the horny cell layer as the other axis, for example. As a matter of course, such a corrected value may be converted to a mathematical formula by, for example, a multiple regression analysis, and the mathematical formula may also be utilized.
  • In contrast to the above, when there is a need for decreasing the absorbability to the horny cell layer, there is performed an operation for decreasing the total content of the ingredient for promoting the absorption to the horny cell layer with respect to the total amount of the pharmaceutical composition, and/or an operation for increasing the total content of the ingredient for inhibiting the absorption to the horny cell layer with respect to the total amount the pharmaceutical composition.
  • As described above, the bioequivalence can be maintained without being affected by the irregularity from lot to lot of a formulation ingredient by adjusting the mixing ratio of the ingredient for promoting the absorption to the horny cell layer and the ingredient for inhibiting the absorption to the horny cell layer.
  • Hereinafter, the present invention is described in more detail by way of examples. However, it goes without saying that the present invention is not limited to only such examples.
  • EXAMPLE 1
  • Skin agents 1 to 5 for external use, each of which is the pharmaceutical composition of the present invention, were prepared in accordance with the following prescription. That is, luliconazole was dissolved by adding an appropriate amount of ethanol, and to the resulting solution, triacetin and glucono-δ-lactone were added and uniformly dispersed. After that, hydroxypropylcellulose was gradually added, and water was further added to obtain homogenous skin agents for external use. Any of those agents had a viscosity of 100 to 800 mPa·s (viscometer: RE80R manufactured by Toki Sangyo Co., Ltd., and conditions for viscosity measurement: cone angle; 0.8°, cone radius; 2.4 cm, rotation frequency; 10 rpm, and temperature; 25° C.), was not so dripping, and thus was suited as a formulation for seborrheic dermatitis to be administered to the hair line and the like. Further, those agents were free of ingredients strongly exhibiting solvent characteristics likemethyl ethyl ketone (MEK) and acetone, did not provide any strong temporary irritation feeling, and could be suitably used for tinea pedis and the like as well.
  • TABLE 1
    Ingredient w/v %
    Luliconazole 1
    Hydroxypropylcellulose 1.5
    Water 20
    Triacetin (TA)*
    Glucono-δ-lactone (GL)*
    Ethanol Appropriate amount
    Total 100 mL
    *See Table 2
  • TABLE 2
    GL TA Viscosity
    Sample (w/v %) (w/v %) (mPa · s)
    Skin agent 1 for external use 3.2 0 639.6
    Skin agent 2 for external use 3.2 12 648.8
    Skin agent 3 for external use 1.6 12 603.0
    Skin agent 4 for external use 1.6 24 656.6
    Skin agent 5 for external use 0 24 652.0
  • <Horny Cell Layer Storability Test>
  • To a plantar part of an isolated guinea pig hind leg, the skin agent for external use was applied at room temperature, and the whole was left out at room temperature for 2 hours. After that, the skin agent for external use was wiped out from the applied site, and further, the applied site was subjected to 5 times of tape stripping with a cellophane tape. Thereafter, the plantar part was excised, and the horny cell layer was peeled off, formed into a piece having a certain size, and evaporated to dryness under reduced pressure for 24 hours. To the obtained dried product, methanol was added, luliconazole as a measurement target was extracted in an ultrasonicator, and filtered through a filter. After that, the concentration of luliconazole in the horny cell layer at two hours after administration was measured with LC-MS/MS (manufactured by Micromass) under the following analysis condition. Table 3 shows the results. The results reveal that any absorption property can be designed by incorporating a combination of the ingredient for inhibiting the absorption to the horny cell layer and the ingredient for promoting the absorption to the horny cell layer, and varying the amount ratio thereof.
  • <HPLC Condition>
  • Column: Wakosil II5C18HG, 4.6×250 mm, 5 μm (Wako Pure Chemical Industries, Ltd.)
  • Mobile phase: 5 mmol/L formic acid aqueous solution:methanol (20:80, v/v)
    Flow rate:1.0 mL/min
    Split ratio ca.: 2:8 (MS/MS:drain)
    Injection volume: 2 μL
    Column temperature: 45° C.
    Sample temperature: 5° C.
  • <MS/MS Condition>
  • Ionization method: ESI, positive ion mode
    Compound: MS1 (m/z), MS2 (m/z)
    Measurement ion: luliconazole 354 150
  • TABLE 3
    Concentration of luliconazole
    Sample in horny cell layer (μg/cm2)
    Skin agent 1 for external use 1.591
    Skin agent 2 for external use 0.910
    Skin agent 3 for external use 0.598
    Skin agent 4 for external use 0.295
    Skin agent 5 for external use 0.327
  • <Stability in Preservation Under Harsh Condition>
  • Each of Skin agents 1 to 5 for external use was evaluated for the time-dependent stability of luliconazole under a harsh preservation condition (60° C., 1 week). The quantitative determination of the S-E isomer which was converted from luliconazole by changing the steric structure thereof was performed by HPLC (LC-20AD manufactured by Shimadzu Corporation, HPLC condition: column; CHIRALCEL OD-RH 4. 6×150 mm, column temperature; 35° C., mobile phase; a mixed solution of methanol/1.8% potassium hexafluorophosphate aqueous solution (83:17, v/v), flow rate; 0.56 mL/min., and detection; 295 nm). Further, the quantitative determination of Z isomers and other analogous substances was performed by HPLC (Agilent 1100 manufactured by Agilent Technologies, HPLC condition: column; Inertsil ODS-2 4.6×150 mm, column temperature; 40° C., mobile phase; 0.13% sodium 1-undecane-sulfonate mixed solution (water/acetonitrile/acetic acid (100) (54:45:1, v/v/v)), flow rate; 1.0 mL/min., and detection; 295 nm). Tables 4 and 5 show the results. The tables reveal that any sample stably exists. That is, in a system of glucono-δ-lactone, triacetin, and hydroxypropylcellulose/ethanol, it can be said that luliconazole stably exists even in preservation under a harsh condition, irrespective of the amount ratio of glucono-δ-lactone and triacetin.
  • TABLE 4
    Immediately after production (weight %)
    Total content of
    S-E Z other analogous
    Sample isomer isomer substances
    Skin agent 1 for external use 0.12 0.02 0.00
    Skin agent 2 for external use 0.13 0.02 0.00
    Skin agent 3 for external use 0.13 0.02 0.00
    Skin agent 4 for external use 0.14 0.02 0.00
    Skin agent 5 for external use 0.13 0.02 0.00
  • TABLE 5
    After preservation for 1 week (weight %)
    Total content of
    S-E Z other analogous
    Sample isomer isomer substances
    Skin agent 1 for external use 0.14 0.03 0.02
    Skin agent 2 for external use 0.14 0.04 0.02
    Skin agent 3 for external use 0.13 0.04 0.02
    Skin agent 4 for external use 0.14 0.03 0.00
    Skin agent 5 for external use 0.37 0.05 0.00
  • EXAMPLE 2
  • In the same manner as in Example 1, Skin agent 6 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription. The concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 4.532 μg/cm2, and the concentration in Comparative Example 1 in which propylene glycol in the preparation was replaced by ethanol was 0.221 μg/cm2. The S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 1.65% in Skin agent 6 for external use, which was higher compared with 1.45% in Comparative Example 1. Propylene glycol is found to be usable as an ingredient for promoting the absorption to the horny cell layer.
  • TABLE 6
    Ingredient w/v %
    Luliconazole 1
    Propylene glycol 50
    Ethanol Appropriate amount
    Total 100 mL
  • EXAMPLE 3
  • In the same manner as in Example 1, Skin agent 7 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription. The concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.584 μg/cm2, and the concentration in Comparative Example 2 in which 2-ethyl-1,3-hexanediol in the preparation was replaced by ethanol was 0.950 μg/cm2. The S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 1.06% in Skin agent 7 for external use, which was higher compared with 0.38% in Comparative Example 2. 2-ethyl-1,3-hexanediol is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • TABLE 7
    Ingredient w/v %
    Luliconazole 1
    Hydroxypropylcellulose 1.5
    Water 20
    2-ethyl-1,3-hexanediol 5
    Ethanol Appropriate amount
    Total 100 mL
  • EXAMPLE 4
  • In the same manner as in Example 1, Skin agent 8 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription. The concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.736 μg/cm2, and the concentration in Comparative Example 3 in which concentrated glycerin (100% glycerin) in the preparation was replaced by ethanol was 0.345 μg/cm2. The S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.79% in Skin agent 8 for external use, which was higher compared with 0.47% in Comparative Example 3. Concentrated glycerin is found to be usable as an ingredient for promoting the absorption to the horny cell layer.
  • TABLE 8
    Ingredient w/v %
    Luliconazole 1
    Water 10
    Concentrated glycerin 32
    Ethanol Appropriate amount
    Total 100 mL
  • EXAMPLE 5
  • In the same manner as in Example 1, Skin agent 9 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription. The concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 2.018 μg/cm2, and the concentration in Comparative Example 1 in which lactic acid in the preparation was replaced by ethanol was 0.221 μg/cm2. The S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.14% in Skin agent 9 for external use, which was lower compared with 1.45% in Comparative Example 1. Lactic acid is found to be usable as an ingredient for promoting the absorption to the horny cell layer.
  • TABLE 9
    Ingredient w/v %
    Luliconazole 1
    Lactic acid 8
    Ethanol Appropriate amount
    Total 100 mL
  • EXAMPLE 6
  • In the same manner as in Example 1, Skin agent 10 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription. The concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.358 μg/cm2, and the concentration in Comparative Example 2 in which polyoxyethylene (20)-polyoxypropylene (20) glycol in the preparation was replaced by ethanol was 0.950 μg/cm2. The S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 1.26% in Skin agent 10 for external use, which was higher compared with 0.38% in Comparative Example 2.
  • Polyoxyethylene (20)-polyoxypropylene (20) glycol is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • TABLE 10
    Ingredient w/v %
    Luliconazole 1
    Hydroxypropylcellulose 1.5
    Water 20
    POE (20) POP (20) glycol 12
    Ethanol Appropriate amount
    Total 100 mL
  • EXAMPLE 7
  • In the same manner as in Example 1, Skin agent 11 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription. The concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.148 μg/cm2, and the concentration in Comparative Example 1 in which lauromacrogol in the preparation was replaced by ethanol was 0.221 μg/cm2. The S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.19% in Skin agent 11 for external use, which was lower compared with 1.45% in Comparative Example 1. Lauromacrogol is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • TABLE 11
    Ingredient w/v %
    Luliconazole 1
    Lauromacrogol 12
    Ethanol Appropriate amount
    Total 100 mL
  • EXAMPLE 8
  • In the same manner as in Example 1, Skin agent 12 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription. The concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.199 μg/cm2, and the concentration in Comparative Example 1 in which polypropylene glycol 2000 in the preparation was replaced by ethanol was 0.221 μg/cm2. The S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.78% in Skin agent 12 for external use, which was lower compared with 1.45% in Comparative Example 1. Polypropylene glycol 2000 is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • TABLE 12
    Ingredient w/v %
    Luliconazole 1
    Polypropylene glycol 2000 20
    Ethanol Appropriate amount
    Total 100 mL
  • EXAMPLE 9
  • In the same manner as in Example 1, Skin agent 13 for external use as the pharmaceutical composition of the present invention was prepared in accordance with the following prescription. The concentration of luliconazole in the horny cell layer at 2 hours after the application of the agent was 0.306 μg/cm2, and the concentration in Comparative Example 2 in which diethyl sebacate in the preparation was replaced by ethanol was 0.950 μg/cm2. The S-E isomer measured by HPLC after harsh preservation (60° C., 1 week) was 0.14% in Skin agent 13 for external use, which was lower compared with 0.38% in Comparative Example 2. Diethyl sebacate is found to be usable as an ingredient for inhibiting the absorption to the horny cell layer.
  • TABLE 13
    Ingredient w/v %
    Luliconazole 1
    Hydroxypropylcellulose 1.5
    Water 20
    Diethyl sebacate 20
    Ethanol Appropriate amount
    Total 100 mL
  • INDUSTRIAL APPLICABILITY
  • The present invention is applicable to a pharmaceutical composition such as a skin agent for external use for seborrheic dermatitis.

Claims (19)

1. A pharmaceutical composition for antifungal use, comprising: 1) a compound selected from the group consisting of compounds represented by the general formula (1) below and physiologically acceptable salts thereof; 2) a compound selected from the group consisting of polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol; and 3) a compound selected from the group consisting of glucono-δ-lactone, propylene glycol, glycerin, and lactic acid.
Figure US20100173965A1-20100708-C00004
wherein X represents a halogen or hydrogen.
2. The pharmaceutical composition according to claim 1, wherein the compound selected from the group consisting of compounds represented by the general formula (1) and physiologically acceptable salts thereof is luliconazole and/or a physiologically acceptable salt thereof.
Figure US20100173965A1-20100708-C00005
3. The pharmaceutical composition according to claim 1 or 2, comprising triacetin as the compound selected from the group consisting of polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol.
4. The pharmaceutical composition according to claim 1, comprising glucono-δ-lactone as the compound selected from the group consisting of glucono-δ-lactone, propylene glycol, glycerin, and lactic acid.
5. The pharmaceutical composition according to claim 1, wherein the pharmaceutical composition for antifungal use is in the form of a single-phase solution.
6. The pharmaceutical composition according to claim 1, further comprising ethanol and/or water.
7. (canceled)
8. A method for adjusting the storability in the horny cell layer of compounds represented by the general formula (1) and/or salts thereof in the pharmaceutical composition according to claim 1, which comprises increasing the storability in the horny cell layer by decreasing the total content of one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol with respect to the total amount of the pharmaceutical composition, and/or by increasing the total content of one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid with respect to the total amount of the pharmaceutical composition; or decreasing the storability in the horny cell layer by increasing the total content of one or more compounds selected from polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol with respect to the total amount of the pharmaceutical composition, and/or by decreasing the total content of one or more compounds selected from glucono-δ-lactone, propylene glycol, glycerin, and lactic acid with respect to the total amount of the pharmaceutical composition, in the case.
9. A method for treating mycosis comprising administering transdermally to a subject in need thereof a composition comprising: 1) a compound selected from the group consisting of compounds represented by the general formula (1) below and physiologically acceptable salts thereof; 2) a compound selected from the group consisting of polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol; and 3) a compound selected from the group consisting of glucono-δ-lactone, propylene glycol, glycerin, and lactic acid.
Figure US20100173965A1-20100708-C00006
wherein X represents a halogen or hydrogen.
10. The method according to claim 9, wherein the compound selected from the group consisting of compounds represented by the general formula (1) and physiologically acceptable salts thereof is luliconazole and/or a physiologically acceptable salt thereof.
Figure US20100173965A1-20100708-C00007
11. The method according to claim 9, wherein the composition comprises triacetin as the compound selected from the group consisting of polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol.
12. The method according to claim 9, wherein the composition comprises glucono-δ-lactone as the compound selected from the group consisting of glucono-δ-lactone, propylene glycol, glycerin, and lactic acid.
13. The method according to claim 9, wherein the composition is in the form of a single-phase solution.
14. The method according to claim 9, wherein the composition further comprises ethanol and/or water.
15. The method according to claim 9, wherein the mycosis is seborrheic dermatitis.
16. A method for stabilizing a compound represented by the general formula (1) in a medicine, the method comprising a step of adding, to the compound, 1) a compound selected from the group consisting of polypropylene glycol, diesters of dibasic acids, triacetin, 2-ethyl-1,3-hexanediol, lauromacrogol, and polyoxyethylene-polyoxypropylene glycol; and 2) a compound selected from the group consisting of glucono-δ-lactone, propylene glycol, glycerin, and lactic acid.
17. The method according to claim 16, wherein the compound represented by the general formula (1) is luliconazole.
18. The method according to claim 16, wherein the stabilizing effects are depletion of production of their isomers.
19. The method according to claim 18, wherein the isomer(s) are E form and/or Z form of luliconazole.
US12/676,345 2007-09-05 2008-09-05 Antifungal composition Abandoned US20100173965A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2007229618 2007-09-05
JP2007-229618 2007-09-05
PCT/JP2008/066057 WO2009031643A1 (en) 2007-09-05 2008-09-05 Antifungal composition

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2008/066057 A-371-Of-International WO2009031643A1 (en) 2007-09-05 2008-09-05 Antifungal composition

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/539,530 Division US9968591B2 (en) 2007-09-05 2014-11-12 Antifungal composition

Publications (1)

Publication Number Publication Date
US20100173965A1 true US20100173965A1 (en) 2010-07-08

Family

ID=40428955

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/676,345 Abandoned US20100173965A1 (en) 2007-09-05 2008-09-05 Antifungal composition
US14/539,530 Active 2029-01-25 US9968591B2 (en) 2007-09-05 2014-11-12 Antifungal composition

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/539,530 Active 2029-01-25 US9968591B2 (en) 2007-09-05 2014-11-12 Antifungal composition

Country Status (6)

Country Link
US (2) US20100173965A1 (en)
EP (1) EP2191827B1 (en)
JP (1) JP5345937B2 (en)
KR (1) KR20100075476A (en)
CN (1) CN101808638B (en)
WO (1) WO2009031643A1 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090030059A1 (en) * 2006-03-08 2009-01-29 Nihon Nohyaku Co., Ltd. Pharmaceutical composition for external use
US20090076109A1 (en) * 2006-03-08 2009-03-19 Nihon Nohyaku Co., Ltd. Pharmaceutical Composition for External Use
US20090137651A1 (en) * 2006-03-08 2009-05-28 Hirokazu Kobayashi Pharmaceutical composition for external use
US20100168200A1 (en) * 2007-09-05 2010-07-01 Pola Pharma Inc. Antifungal pharmaceutical composition
US20100204293A1 (en) * 2007-09-05 2010-08-12 Pola Pharma Inc. Pharmaceutical composition
US20100210703A1 (en) * 2009-02-13 2010-08-19 Vontz Charles G Anti-fungal formulation
US8952044B2 (en) 2009-08-25 2015-02-10 Pola Pharma Inc. Antimycotic pharmaceutical composition
US8962669B2 (en) 2010-06-11 2015-02-24 Pola Pharma Inc. Antimycotic pharmaceutical composition
US8980931B1 (en) 2013-12-12 2015-03-17 Pola Pharma Inc. Method of evaluating pharmaceutical preparation containing luliconazole and index substance
US9012484B2 (en) 2012-09-14 2015-04-21 Pola Pharma Inc. Crystal and pharmaceutical preparation containing the same crystal
US9050271B2 (en) 2009-04-09 2015-06-09 Pola Pharma Inc. Antimycotic pharmaceutical composition
US9068958B1 (en) 2012-09-14 2015-06-30 Pola Pharma Inc. Use of surface free energy for differential evaluation of crystal, crystal evaluated on basis of surface free energy as index, and pharmaceutical composition prepared by containing the crystal
US9145401B2 (en) 2012-09-14 2015-09-29 Pola Pharma Inc. Amide derivative and use of the same as stability index of a luliconazole pharmaceutical formulation
US9199977B2 (en) 2012-09-14 2015-12-01 Pola Pharma Inc. Crystal having crystal habits and pharmaceutical composition obtained by processing the crystal
US9453006B2 (en) 2013-03-08 2016-09-27 Pola Pharma Inc. Crystalline form having specific crystal habit and pharmaceutical composition containing this crystalline form as active ingredient
US9527836B2 (en) 2013-09-06 2016-12-27 Pola Pharma Inc. Crystal having specific crystal habit and pharmaceutical composition containing the crystal as active ingredient
US9968591B2 (en) 2007-09-05 2018-05-15 Pola Pharma Inc. Antifungal composition
US10130610B2 (en) 2009-04-09 2018-11-20 Pola Pharma Inc. Antimycotic pharmaceutical composition
US20190142848A1 (en) * 2015-02-20 2019-05-16 Pedicis Research Llc Compositions and methods for treatment of skin infections
US10898470B1 (en) 2019-08-13 2021-01-26 Sato Pharmaceutical Co., Ltd. Pharmaceutical composition containing antifungal agent as active ingredient

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104619320A (en) * 2012-09-14 2015-05-13 宝丽制药股份有限公司 Pharmaceutical composition containing luliconazole
JP6067399B2 (en) * 2013-02-08 2017-01-25 株式会社ポーラファルマ Pharmaceutical composition
JP5926756B2 (en) * 2014-03-26 2016-05-25 株式会社ポーラファルマ Evaluation method and index substance of preparation containing luliconazole
JP5623671B1 (en) * 2014-07-23 2014-11-12 株式会社ポーラファルマ Evaluation method and index substance of preparation containing luliconazole
JP2015113343A (en) * 2014-09-24 2015-06-22 株式会社ポーラファルマ Evaluation method and index material of preparation containing luliconazole
WO2017170509A1 (en) * 2016-03-31 2017-10-05 株式会社ポーラファルマ Amorphizing agent, amorphous composition comprising amorphizing agent and utilization thereof
JP2017181387A (en) * 2016-03-31 2017-10-05 株式会社ポーラファルマ Evaluation method of film formed by pharmaceutical composition
ES2896680T3 (en) 2016-04-06 2022-02-25 Gedea Biotech Ab Glucono delta lactone for the treatment of vaginal fungal infections
WO2018179170A1 (en) * 2017-03-29 2018-10-04 日本農薬株式会社 Pharmaceutical composition for treatment of infection

Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4267169A (en) * 1978-07-22 1981-05-12 Toko Yakuhin Kogyo Kabushiki Kaisha Novel preparation of clotrimazole
US4636520A (en) * 1984-07-16 1987-01-13 Fujisawa Pharmaceutical Co., Ltd. Antifungal composition employing pyrrolnitrin in combination with an imidazole compound
US4764381A (en) * 1985-12-06 1988-08-16 Key Pharmaceuticals, Inc. Percutaneous penetration enhancer of oleic acid and 2-ethyl-1, 3-hexanediol
US5340836A (en) * 1991-02-19 1994-08-23 Mark S. Reinhard Composition and method for treatment of vaginal yeast infections
US5690923A (en) * 1993-01-07 1997-11-25 Yamanouchi Europe B.V. Stable topical retinoid compositions
US5753256A (en) * 1993-11-06 1998-05-19 Labtec Gesellschaft Fur Biotechnologische Forschung Und Entwicklung Mbh Plaster for the treatment of nail mycoses
US5814305A (en) * 1991-03-08 1998-09-29 Novartis Ag Use of hydrophilic penetration agents in dermatological compositions for the treatment of onychomycoses, and corresponding compositions
US5962536A (en) * 1998-07-31 1999-10-05 Komer; Gene Injectable propofol formulations
US5993787A (en) * 1996-09-13 1999-11-30 Johnson & Johnson Consumer Products, Inc. Composition base for topical therapeutic and cosmetic preparations
US6008256A (en) * 1995-08-28 1999-12-28 Showa Yakuhin Kako Co., Ltd. Composition for local anesthesia
US6007791A (en) * 1994-10-10 1999-12-28 Chiron Corporation Preparation of protein microspheres, films and coatings
US6017920A (en) * 1994-05-06 2000-01-25 Toko Yakuhin Kogyo Kabushiki Kaisha Antifungal composition for external use being retentive in stratum corneum
US6083518A (en) * 1995-06-20 2000-07-04 Bioglan Ab Composition comprising an active agent dissolved in a glass-forming carrier and a process for the preparation thereof
US6428654B1 (en) * 2000-04-05 2002-08-06 Hercules Incorporated Fungicidal method
US20030017207A1 (en) * 2001-05-01 2003-01-23 Lin Shun Y. Compositions and methods for treating vulvovaginitis and vaginosis
US6585963B1 (en) * 2001-02-15 2003-07-01 Watson Pharmaceuticals, Inc. Nail compositions and methods of administering same
US20030235541A1 (en) * 2002-06-21 2003-12-25 Maibach Howard I. Topical administration of basic antifungal compositions to treat fungal infections of the nails
US6740326B1 (en) * 1998-09-10 2004-05-25 Bioequal Ag Topical nail care compositions
US20040208906A1 (en) * 1999-07-12 2004-10-21 Daiichi Suntory Pharma Co., Ltd. Pharmaceutical composition for topical administration
US20050232879A1 (en) * 2002-06-18 2005-10-20 Hideaki Sasagawa Antifungal medicinal compositions
US20060140984A1 (en) * 2002-10-25 2006-06-29 Foamix Ltd. Cosmetic and pharmaceutical foam
US20070099932A1 (en) * 2003-06-25 2007-05-03 Toshihiro Shirouzu External preparation for athlete's foot treatment
US20090030059A1 (en) * 2006-03-08 2009-01-29 Nihon Nohyaku Co., Ltd. Pharmaceutical composition for external use
US20090076109A1 (en) * 2006-03-08 2009-03-19 Nihon Nohyaku Co., Ltd. Pharmaceutical Composition for External Use
US20090137651A1 (en) * 2006-03-08 2009-05-28 Hirokazu Kobayashi Pharmaceutical composition for external use
US20090202602A1 (en) * 2005-12-28 2009-08-13 Teikoku Seiyaku Co., Ltd. Pharmaceutical composition for application to nail
US20100168200A1 (en) * 2007-09-05 2010-07-01 Pola Pharma Inc. Antifungal pharmaceutical composition
US20100204293A1 (en) * 2007-09-05 2010-08-12 Pola Pharma Inc. Pharmaceutical composition
US20100210702A1 (en) * 2009-02-13 2010-08-19 Topica Pharmaceuticals, Inc. Anti-fungal formulation
US20100249202A1 (en) * 2007-08-27 2010-09-30 Nihon Nohyaku Co., Ltd. Agent for fungal dermatitis

Family Cites Families (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5815909A (en) 1981-07-22 1983-01-29 Toko Yakuhin Kogyo Kk Antimycotic agent for external use
JPH0236572B2 (en) 1984-11-13 1990-08-17 Hokuriku Pharmaceutical SUTEROIDOO177MONOESUTERUGANJUKURIIMUZAI
JPS6293227A (en) 1985-10-19 1987-04-28 Nippon Nohyaku Co Ltd Antimycotic agent
JPH0774144B2 (en) 1986-03-12 1995-08-09 久光製薬株式会社 Skin composition containing urea
US5686489A (en) 1986-12-23 1997-11-11 Tristrata Technology, Inc. Alpha hydroxyacid esters for skin aging
JPH01246219A (en) 1988-03-25 1989-10-02 Nippon Nohyaku Co Ltd Antimycotic cream composition for external use
JPH01242525A (en) 1988-03-25 1989-09-27 Nippon Nohyaku Co Ltd Antifungal agent for external use
JP3030780B2 (en) 1988-12-29 2000-04-10 日本農薬株式会社 Optically active ketene dithioacetal derivative and method for producing the same
JPH02264723A (en) 1989-04-06 1990-10-29 Taisho Pharmaceut Co Ltd Antifungal agent
EP0473707A4 (en) 1989-05-26 1992-12-09 Abbott Laboratories Injectable clarithromycin composition
US5061700A (en) * 1989-11-16 1991-10-29 Gordon Jay Dow Glyceryl acetate ointment vehicles
IL97012A0 (en) 1990-01-30 1992-03-29 Gist Brocades Nv Topical preparations for treating human nails
JP2555555B2 (en) 1991-07-03 1996-11-20 武田薬品工業株式会社 Antifungal topical formulation
JPH06199701A (en) * 1992-12-29 1994-07-19 Lion Corp Anti-inflammatory analgesic agent for external use
JPH06211651A (en) 1993-01-12 1994-08-02 Hisamitsu Pharmaceut Co Inc Composition for treating nail trichophytosis
JPH0774144A (en) 1993-06-29 1995-03-17 Sony Corp Cleaning method for plasma apparatus
JP2860748B2 (en) 1993-12-24 1999-02-24 エスエス製薬株式会社 Stable hydrocortisone butyrate-containing cream
JP3803393B2 (en) 1994-01-12 2006-08-02 久光製薬株式会社 Nail ringworm treatment composition
JP3629283B2 (en) 1994-02-15 2005-03-16 ポーラ化成工業株式会社 Skin preparation
WO1996011710A1 (en) * 1994-10-13 1996-04-25 Hisamitsu Pharmaceutical Co., Inc. External preparation for nail ringworm
SE9403541L (en) * 1994-10-14 1996-04-15 Sven Moberg Antimicrobial composition
KR100386491B1 (en) 1995-07-08 2003-08-21 니혼노야쿠가부시키가이샤 Antifungal agents, compounds and methods for their preparation
JP4253047B2 (en) 1996-09-27 2009-04-08 杏林製薬株式会社 Film-forming antifungal composition
JP4227677B2 (en) 1996-12-10 2009-02-18 杏林製薬株式会社 Film-forming antifungal composition
JPH10226686A (en) 1996-12-10 1998-08-25 Nippon Nohyaku Co Ltd (r)-(e)-(4-substituted phenyl-1,3-dithiolan-2-iriden)-1-imidazole-acetonitrile derivative as optically active substance, antifungal agent and production of the same
AU7936198A (en) 1998-07-01 2000-01-24 Lead Chemical Co., Ltd. Ketotifen preparation for percutaneous absorption
JP2001064206A (en) 1999-08-30 2001-03-13 Isp Japan Kk Percutaneous absorption-promoting composition
US6541517B1 (en) * 1999-09-03 2003-04-01 Donald M. Murphy Treatment of skin disorders
US7074392B1 (en) 2000-03-27 2006-07-11 Taro Pharmaceutical Industries Limited Controllled delivery system of antifungal and keratolytic agents for local treatment of fungal infections
JP2002114680A (en) 2000-07-31 2002-04-16 Nippon Nohyaku Co Ltd Antifungal agent
JP2002193755A (en) * 2000-12-28 2002-07-10 Sansho Seiyaku Co Ltd Antidandruff and anti-itching hair cosmetic and hair- shampooing cosmetic
JP2002284702A (en) 2001-01-19 2002-10-03 Teika Seiyaku Kk Antifungal pharmaceutical preparation for external use
KR100423666B1 (en) 2001-02-07 2004-03-18 보령제약 주식회사 Composition of Antifungal Topical preparations
JP2002363070A (en) 2001-06-06 2002-12-18 Yuutoku Yakuhin Kogyo Kk Transdermal patch preparation
BR0212311A (en) * 2001-09-04 2004-10-13 Trommsdorff Arzneimittel Plaster for the treatment of nail disorders and disorders
JP2003252798A (en) * 2002-02-28 2003-09-10 Shiseido Co Ltd Antibacterial gel preparation
JP4387639B2 (en) * 2002-07-16 2009-12-16 久光製薬株式会社 Transdermal absorption preparation
CA2492976C (en) 2002-09-05 2011-11-01 Galderma Research & Development, S.N.C. Solution for ungual and peri-ungual application
EP2106805B1 (en) 2003-03-21 2011-06-15 Nexmed Holdings, Inc. Antifungal nail coat and method of use
ES2535045T3 (en) 2003-04-10 2015-05-04 Vanderbilt Royalty Sub L.P. Uses and compositions for capsaicin administration
JP2005104924A (en) 2003-09-30 2005-04-21 Kobayashi Pharmaceut Co Ltd External pharmaceutical composition
JP4431369B2 (en) 2003-11-21 2010-03-10 久光製薬株式会社 Antifungal aerosol topical preparation
JP4429035B2 (en) 2004-02-27 2010-03-10 久光製薬株式会社 Cream preparation for external use with enhanced keratin retention
JP3783104B2 (en) 2004-03-31 2006-06-07 小林製薬株式会社 Antifungal composition for external use
WO2005123136A1 (en) 2004-06-15 2005-12-29 Hisamitsu Pharmaceutical Co., Inc. Antiinflammatory and analgesic preparation for external use
JP4690672B2 (en) 2004-07-20 2011-06-01 岩城製薬株式会社 Emulsion skin external preparation
JP3800232B2 (en) 2004-09-30 2006-07-26 小林製薬株式会社 Antifungal composition for external use
JP2006306734A (en) 2005-04-26 2006-11-09 Tsumura & Co External preparation blended with urea
WO2008075207A2 (en) 2006-04-04 2008-06-26 Foamix Ltd. Anti-infection augmentation foamable compositions and kit and uses thereof
EP2191827B1 (en) 2007-09-05 2013-10-30 Pola Pharma Inc. Antifungal composition

Patent Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4267169A (en) * 1978-07-22 1981-05-12 Toko Yakuhin Kogyo Kabushiki Kaisha Novel preparation of clotrimazole
US4636520A (en) * 1984-07-16 1987-01-13 Fujisawa Pharmaceutical Co., Ltd. Antifungal composition employing pyrrolnitrin in combination with an imidazole compound
US4764381A (en) * 1985-12-06 1988-08-16 Key Pharmaceuticals, Inc. Percutaneous penetration enhancer of oleic acid and 2-ethyl-1, 3-hexanediol
US5340836A (en) * 1991-02-19 1994-08-23 Mark S. Reinhard Composition and method for treatment of vaginal yeast infections
US5814305A (en) * 1991-03-08 1998-09-29 Novartis Ag Use of hydrophilic penetration agents in dermatological compositions for the treatment of onychomycoses, and corresponding compositions
US5690923A (en) * 1993-01-07 1997-11-25 Yamanouchi Europe B.V. Stable topical retinoid compositions
US5753256A (en) * 1993-11-06 1998-05-19 Labtec Gesellschaft Fur Biotechnologische Forschung Und Entwicklung Mbh Plaster for the treatment of nail mycoses
US6017920A (en) * 1994-05-06 2000-01-25 Toko Yakuhin Kogyo Kabushiki Kaisha Antifungal composition for external use being retentive in stratum corneum
US6007791A (en) * 1994-10-10 1999-12-28 Chiron Corporation Preparation of protein microspheres, films and coatings
US6083518A (en) * 1995-06-20 2000-07-04 Bioglan Ab Composition comprising an active agent dissolved in a glass-forming carrier and a process for the preparation thereof
US6008256A (en) * 1995-08-28 1999-12-28 Showa Yakuhin Kako Co., Ltd. Composition for local anesthesia
US5993787A (en) * 1996-09-13 1999-11-30 Johnson & Johnson Consumer Products, Inc. Composition base for topical therapeutic and cosmetic preparations
US5962536A (en) * 1998-07-31 1999-10-05 Komer; Gene Injectable propofol formulations
US6740326B1 (en) * 1998-09-10 2004-05-25 Bioequal Ag Topical nail care compositions
US20040208906A1 (en) * 1999-07-12 2004-10-21 Daiichi Suntory Pharma Co., Ltd. Pharmaceutical composition for topical administration
US6428654B1 (en) * 2000-04-05 2002-08-06 Hercules Incorporated Fungicidal method
US6585963B1 (en) * 2001-02-15 2003-07-01 Watson Pharmaceuticals, Inc. Nail compositions and methods of administering same
US20030017207A1 (en) * 2001-05-01 2003-01-23 Lin Shun Y. Compositions and methods for treating vulvovaginitis and vaginosis
US20050232879A1 (en) * 2002-06-18 2005-10-20 Hideaki Sasagawa Antifungal medicinal compositions
US20030235541A1 (en) * 2002-06-21 2003-12-25 Maibach Howard I. Topical administration of basic antifungal compositions to treat fungal infections of the nails
US20060140984A1 (en) * 2002-10-25 2006-06-29 Foamix Ltd. Cosmetic and pharmaceutical foam
US20070099932A1 (en) * 2003-06-25 2007-05-03 Toshihiro Shirouzu External preparation for athlete's foot treatment
US20090099202A1 (en) * 2003-06-25 2009-04-16 Toshihiro Shirouzu External Preparation for Athlete's Foot Treatment
US20090202602A1 (en) * 2005-12-28 2009-08-13 Teikoku Seiyaku Co., Ltd. Pharmaceutical composition for application to nail
US20090030059A1 (en) * 2006-03-08 2009-01-29 Nihon Nohyaku Co., Ltd. Pharmaceutical composition for external use
US20090076109A1 (en) * 2006-03-08 2009-03-19 Nihon Nohyaku Co., Ltd. Pharmaceutical Composition for External Use
US20090137651A1 (en) * 2006-03-08 2009-05-28 Hirokazu Kobayashi Pharmaceutical composition for external use
US20100249202A1 (en) * 2007-08-27 2010-09-30 Nihon Nohyaku Co., Ltd. Agent for fungal dermatitis
US20100168200A1 (en) * 2007-09-05 2010-07-01 Pola Pharma Inc. Antifungal pharmaceutical composition
US20100204293A1 (en) * 2007-09-05 2010-08-12 Pola Pharma Inc. Pharmaceutical composition
US20100210702A1 (en) * 2009-02-13 2010-08-19 Topica Pharmaceuticals, Inc. Anti-fungal formulation
US20100210703A1 (en) * 2009-02-13 2010-08-19 Vontz Charles G Anti-fungal formulation

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8349882B2 (en) 2006-03-08 2013-01-08 Nihon Nohyaku Co., Ltd. Pharmaceutical composition for external use
US20090076109A1 (en) * 2006-03-08 2009-03-19 Nihon Nohyaku Co., Ltd. Pharmaceutical Composition for External Use
US20090137651A1 (en) * 2006-03-08 2009-05-28 Hirokazu Kobayashi Pharmaceutical composition for external use
US8058303B2 (en) 2006-03-08 2011-11-15 Nihon Nohyaku Co, Ltd Pharmaceutical composition for external use
US20090030059A1 (en) * 2006-03-08 2009-01-29 Nihon Nohyaku Co., Ltd. Pharmaceutical composition for external use
US8268876B2 (en) 2006-03-08 2012-09-18 Nihon Nohyaku Co., Ltd. Pharmaceutical composition for external use
US20100168200A1 (en) * 2007-09-05 2010-07-01 Pola Pharma Inc. Antifungal pharmaceutical composition
US20100204293A1 (en) * 2007-09-05 2010-08-12 Pola Pharma Inc. Pharmaceutical composition
US9480678B2 (en) 2007-09-05 2016-11-01 Pola Pharma Inc. Antifungal pharmaceutical composition
US9968591B2 (en) 2007-09-05 2018-05-15 Pola Pharma Inc. Antifungal composition
US8513296B2 (en) 2007-09-05 2013-08-20 Pola Pharma Inc. Pharmaceutical composition
US8362059B2 (en) 2009-02-13 2013-01-29 Topica Pharmaceuticals, Inc. Anti-fungal formulation
US8193233B2 (en) 2009-02-13 2012-06-05 Topica Pharmaceuticals, Inc. Anti-fungal formulation
US8193232B2 (en) 2009-02-13 2012-06-05 Topica Pharmaceuticals, Inc. Anti-fungal formulation
US20100210702A1 (en) * 2009-02-13 2010-08-19 Topica Pharmaceuticals, Inc. Anti-fungal formulation
US20100210703A1 (en) * 2009-02-13 2010-08-19 Vontz Charles G Anti-fungal formulation
US10130610B2 (en) 2009-04-09 2018-11-20 Pola Pharma Inc. Antimycotic pharmaceutical composition
US9050271B2 (en) 2009-04-09 2015-06-09 Pola Pharma Inc. Antimycotic pharmaceutical composition
US8952044B2 (en) 2009-08-25 2015-02-10 Pola Pharma Inc. Antimycotic pharmaceutical composition
US8962669B2 (en) 2010-06-11 2015-02-24 Pola Pharma Inc. Antimycotic pharmaceutical composition
US9012484B2 (en) 2012-09-14 2015-04-21 Pola Pharma Inc. Crystal and pharmaceutical preparation containing the same crystal
US9068958B1 (en) 2012-09-14 2015-06-30 Pola Pharma Inc. Use of surface free energy for differential evaluation of crystal, crystal evaluated on basis of surface free energy as index, and pharmaceutical composition prepared by containing the crystal
US9145401B2 (en) 2012-09-14 2015-09-29 Pola Pharma Inc. Amide derivative and use of the same as stability index of a luliconazole pharmaceutical formulation
US9199977B2 (en) 2012-09-14 2015-12-01 Pola Pharma Inc. Crystal having crystal habits and pharmaceutical composition obtained by processing the crystal
US9453006B2 (en) 2013-03-08 2016-09-27 Pola Pharma Inc. Crystalline form having specific crystal habit and pharmaceutical composition containing this crystalline form as active ingredient
US9527836B2 (en) 2013-09-06 2016-12-27 Pola Pharma Inc. Crystal having specific crystal habit and pharmaceutical composition containing the crystal as active ingredient
US9085554B2 (en) * 2013-12-12 2015-07-21 Pola Pharma Inc. Method of evaluating pharmaceutical preparation containing luliconazole and index substance
US20150168357A1 (en) * 2013-12-12 2015-06-18 Pola Pharma Inc. Method of evaluating pharmaceutical preparation containing luliconazole and index substance
US8980931B1 (en) 2013-12-12 2015-03-17 Pola Pharma Inc. Method of evaluating pharmaceutical preparation containing luliconazole and index substance
US20190142848A1 (en) * 2015-02-20 2019-05-16 Pedicis Research Llc Compositions and methods for treatment of skin infections
US11504384B1 (en) 2015-02-20 2022-11-22 Pedicis Research Llc Compositions and methods for treatment of skin infections
US11833161B2 (en) 2015-02-20 2023-12-05 Pedicis Research Llc Compositions and methods for treatment of skin infections
US10898470B1 (en) 2019-08-13 2021-01-26 Sato Pharmaceutical Co., Ltd. Pharmaceutical composition containing antifungal agent as active ingredient
US11738006B2 (en) 2019-08-13 2023-08-29 Sato Pharmaceutical Co., Ltd. Pharmaceutical composition containing antifungal agent as active ingredient

Also Published As

Publication number Publication date
WO2009031643A1 (en) 2009-03-12
JPWO2009031643A1 (en) 2010-12-16
US9968591B2 (en) 2018-05-15
EP2191827B1 (en) 2013-10-30
EP2191827A1 (en) 2010-06-02
EP2191827A4 (en) 2010-09-15
JP5345937B2 (en) 2013-11-20
CN101808638B (en) 2012-07-25
KR20100075476A (en) 2010-07-02
US20150073028A1 (en) 2015-03-12
CN101808638A (en) 2010-08-18

Similar Documents

Publication Publication Date Title
US9968591B2 (en) Antifungal composition
KR101409792B1 (en) Antimycotic pharmaceutical composition
US8513296B2 (en) Pharmaceutical composition
US9480678B2 (en) Antifungal pharmaceutical composition
KR101409789B1 (en) Antimycotic pharmaceutical composition
JP5635075B2 (en) Antifungal pharmaceutical composition
WO2012011566A1 (en) Tacrolimus-containing oil-in-water type creamy composition
US11607399B2 (en) Pharmaceutical formulation for histone deacetylase inhibitors
WO2021177256A1 (en) Drug containing sofpironium bromide
US20230172937A1 (en) Topical formulations of ruxolitinib with an organic amine ph adjusting agent for treatment of skin diseases
RU2699674C1 (en) Percutaneous absorption composition
JP6625208B2 (en) Transdermal formulation
WO2007086582A1 (en) OIL-IN-WATER TYPE EMULSION LOTION CONTAINING 22-OXA-1α,25-DIHYDROXYVITAMIN D3 AND METHOD OF TREATING SKIN DISEASE BY USING THE SAME
JP4060347B2 (en) Oil-in-water emulsion lotion containing 22-oxa-1α, 25-dihydroxyvitamin D3
JP2014208618A (en) Pharmaceutical liquid composition
WO2023102559A1 (en) Topical formulations of ruxolitinib with an organic amine ph adjusting agent for treatment of skin diseases
JP5674786B2 (en) Oil-in-water cream composition containing tacrolimus
JP2020158501A (en) Skin external preparation containing loxoprofen and solubilizer

Legal Events

Date Code Title Description
AS Assignment

Owner name: POLA PHARMA INC., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MASUDA, TAKAAKI;NISHIDA, NAOTO;KOBAYASHI, NAOKO;AND OTHERS;REEL/FRAME:024029/0967

Effective date: 20100208

Owner name: NIHON NOHYAKU CO., LTD., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MASUDA, TAKAAKI;NISHIDA, NAOTO;KOBAYASHI, NAOKO;AND OTHERS;REEL/FRAME:024029/0967

Effective date: 20100208

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION