US20150290178A1 - Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient - Google Patents

Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient Download PDF

Info

Publication number
US20150290178A1
US20150290178A1 US14/731,964 US201514731964A US2015290178A1 US 20150290178 A1 US20150290178 A1 US 20150290178A1 US 201514731964 A US201514731964 A US 201514731964A US 2015290178 A1 US2015290178 A1 US 2015290178A1
Authority
US
United States
Prior art keywords
pde
roflumilast
dosage form
inhibitor
treatment
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
US14/731,964
Inventor
Rango Dietrich
Klaus Eistetter
Hartmut Ney
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.)
AstraZeneca AB
Original Assignee
Takeda GmbH
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27758398&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20150290178(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from DE2002107160 external-priority patent/DE10207160A1/en
Application filed by Takeda GmbH filed Critical Takeda GmbH
Priority to US14/731,964 priority Critical patent/US20150290178A1/en
Publication of US20150290178A1 publication Critical patent/US20150290178A1/en
Assigned to ASTRAZENECA AB reassignment ASTRAZENECA AB ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TAKEDA GMBH
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0002Galenical forms characterised by the drug release technique; Application systems commanded by energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • 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
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to the field of pharmaceutical technology and describes a dosage form for oral administration of a PDE 4 inhibitor as active ingredient in tablet or pellet form for treating diseases such as asthma or airway obstructions.
  • the invention additionally relates to processes for producing the dosage form.
  • PDE Cyclic nucleotide phosphodiesterase
  • WO00/50011 and WO01/32165 which relate to dosage forms with controlled or sustained delivery of a PDE 4 inhibitor, it is pointed out that unwanted CNS side effects may become manifest on delivery of certain PDE 4 inhibitors such as Ariflo® (INN: cilomilast) in higher dosages.
  • PDE 4 inhibitors such as Ariflo® (INN: cilomilast) in higher dosages.
  • WO00/50011 and WO01/32165 see this as being a particular risk with immediate release dosage forms of the active ingredient and therefore propose administering the PDE 4 inhibitor Ariflo® (INN: cilomilast) in dosage forms with controlled or sustained release.
  • U.S. Pat. No. 5,286,494 proposes a dosage form with controlled or sustained release for the PDE 4 inhibitor Rolipram whose solubility is slight.
  • production of dosage forms with controlled or sustained release of slightly soluble active ingredients may be technically complicated, reference being made thereto for example in U.S. Pat. No. 5,286,494.
  • the solubility of active ingredients of the PDE 4 inhibitor class in water and aqueous systems may, depending on the chemical structure, be low.
  • the solubility in water found for the PDE 4 inhibitor N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (INN: roflumilast) which is described in WO95/01338, is only 0.53 mg/l at 21° C.
  • the bioavailability of a medicinal substance depends essentially on the release of the medicinal substance from the pharmaceutical form. Faster release and dissolution of the medicinal substance from the formulation means faster absorption thereof. With medicinal substances which are slightly soluble in water, therefore, the bioavailability is frequently limited by the solubility or rate of dissolution. This makes it very difficult to produce suitable dosage forms.
  • the oral dosage form of the invention is preferably a solid dosage form in tablet or pellet form. It is preferably a solid oral dosage form with immediate release of the active ingredient (immediate release solid oral dosage form).
  • the invention therefore relates to a dosage form in tablet or pellet form for oral administration of a PDE 4 inhibitor whose solubility is slight, comprising the PDE 4 inhibitor whose solubility is slight together with polvinylpyrrolidone as binder, and one or more other suitable pharmaceutical excipients.
  • the PDE 4 inhibitor whose solubility is slight is preferably according to the invention a compound from the group of compounds of the formula I
  • R1 is 3-7C cyotoalkoxy, 3-7C cycloalkylmethoxy or benzyloxy and
  • R2 is 1-4C alkoxy which is completely or partly substituted by fluorine, or
  • R1 is 1-4C alkoxy which is completely or partly substituted by fluorine and
  • R2 is 3-7C cycloalkylmethoxy or benzyloxy
  • R3 is phenyl, pyridyl, phenyl substituted by R31, R32 and R33, or pyridyl substituted by R34, R35, R36 and R37, where
  • Cycloalkoxy is, for example, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy and cycloheptyloxy, of which cyclopropyloxy, cyclobutyloxy and cyclopentyloxy are preferred.
  • Cycloalkylmethoxy is, for example, cyclopropylmethoxy, cyclobutylmethoxy, cyclopentylmethoxy, cyclohexylmethoxy and cycloheptylmethoxy, of which cyclopropylmethoxy, cyclobutylmethoxy and cyclopentylmethoxy are preferred.
  • Examples which may be mentioned of 1-4C alkoxy which is completely or partly substituted by fluorine are 1,2,2-trifluoroethoxy, 2,2,3,3,3-pentafluoropropoxy, perfluoroethoxy and, in particular, 1,1,2,2-tetra-fluoroethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy and difluoromethoxy radicals.
  • Halogen for the purposes of the present invention is bromine, chlorine and fluorine.
  • Alkyl represents straight-chain or branched alkyl radicals having 1 to 4 carbon atoms. Examples which may be mentioned are butyl, isobutyl, sec-butyl, tert-butyl, propyl, isopropyl, ethyl and methyl radicals.
  • Alkoxy is a radical which, besides the oxygen atom, contains one of the aforementioned
  • 1-4C Alkoxycarbonyl is a carbonyl group to which one of the aforementioned 1-4C alkoxy radicals is bonded. Examples which may be mentioned are the methoxycarbonyl (CH 3 O—CO—) and ethoxycarbonyl (CH 3 CH 2 O—CO—) radicals.
  • 1-4C Alkylcarbonyl is a carbonyl group to which one of the aforementioned 1-4C alkyl radicals is bonded.
  • An example which may be mentioned is the acetyl radical (CH 3 CO—).
  • 1-4C Alkylcarbonyloxy radicals comprise besides the oxygen atom one of the aforementioned 1-4C alkylcarbonyl radicals.
  • An example which may be mentioned is the acetoxy radical (CH 3 CO—O—).
  • Examples of mono- or di-1-4C alkylamino radicals which may be mentioned are the methylamino, dimethylamino and diethylamino radicals.
  • a 1-4C alkylcarbonylamino radical which may be mentioned is the acetylamino radical (—NH—CO—CH 3 ).
  • phenyl radicals substituted by R31, R32 and R33 which may be mentioned are the radicals 2-acetylphenyl, 2-aminophenyl, 2-bromophenyl, 2-chlorophenyl, 2,3-dichlorophenyl, 2,4-dichiorophenyl, 4-diethylamino-2-methylphenyl, 4-bromo-2-trifluoromethylphenyl, 2-carboxy-5-chlorophenyl, 3,5-dichloro-2-hydroxyphenyl, 2-bromo-4-carboxy-5-hydroxyphenyl, 2,6-dichiorophenyl, 2,5-dichiorophenyl, 2,4,6-trichlorophenyl, 2,4,6-trifluorophenyl, 2,6-dibromophenyl, 2-cyanophenyl, 4-cyano-2-fluorophenyl, 2-fluorophenyl, 2,4-difluorophenyl, 2,6
  • pyridyl radicals substituted by R34, R35, R36 and R37 which may be mentioned are the radicals 3,5-dichloropyrid-4-yl, 2,6-diaminopyrid-3-yl, 4-aminopyrid-3-yl, 3-methylpyrid-2-yl, 4-methylpyrid-2-yl, 5-hydroxypyrid-2-yl, 4-chloropyrid-3-yl, 3-chloropyrid-2-yl, 3-chloropyrid-4-yl, 2-chloropyrid-3-yl, 2,3,5,6-tetrafluoropyrid-4-yl, 3,5-dichloro-2,6-difluoropyrid-4-yl, 3,5-dibromopyrid-2-yl, 3,5-dibromopyrid-4-yl, 3,5-dichloropyrid-4-yl, 2,6-dichloropyrid-3-yl, 3,5-dii
  • water-soluble and water-insoluble acid addition salts with acids such as, for example, hydrochloric acid, hydrobromic acid, phosphoric acid, nitric acid, sulfuric acid, acetic acid, citric acid, D-gluconic acid, benzoic acid, 2-(4-hydroxybenzoyl)benzoic acid, butyric acid, sulfosalicylic acid, maleic acid, lauric acid, malic acid, fumaric acid, succinic acid, oxalic acid, tartaric acid, embonic acid, stearic acid, toluenesulfonic acid, methanesulfonic acid, or 3-hydroxy-2-naphthoic acid, the acids being employed to prepare the salts in the equimolar ratio of amounts, or one differing therefrom—depending on whether the acid is monobasic or polybasic and depending on which salt is desired.
  • acids such as, for example, hydrochloric acid, hydrobromic acid, phosphoric acid, ni
  • salts with bases are also particularly suitable.
  • basic salts which may be mentioned are lithium, sodium, potassium, calcium, aluminum, magnesium, titanium, ammonium, meglumine or guanidinium salts, once again the bases being employed to prepare the salts in the equimolar ratio of amounts or one differeing therefrom.
  • R1 is 3-5C cycloalkoxy, 3-5C cycloalkylmethoxy or benzyloxy and
  • R2 is 1-4C alkoxy which is completely or partly substituted by fluorine, or
  • R1 is 1-4C alkoxy which is completely or partly substituted by fluorine and
  • R2 is 3-5C cycloalkylmethoxy or benzyloxy
  • R3 is phenyl, pyridyl, phenyl substituted by R31, R32 and R33, or pyridyl substituted by R34, R35, R36 and R37, where
  • R1 is 3-5C cycloalkoxy, 3-5C cycloalkylmethoxy or benzyloxy and
  • R2 is 1-4C alkoxy which is completely or partly substituted by fluorine, or
  • R1 is 1-4C alkoxy which is completely or partly substituted by fluorine and
  • R2 is 3-5C cycloalkylmethoxy or benzyloxy
  • R3 is 2-bromophenyl, 2,6-dichloro-4-ethoxycarbonylphenyl, 2,6-dimethoxyphenyl, 4-cyano-2-fluoro-phenyl, 2,4,6-trifluorophenyl, 2-chloro-6-methylphenyl, 2,6-dimethylphenyl, 2,6-difluorophenyl, 2,6-dichlorophenyl, 3,5-dichloropyrid-4-yl, 3-methylpyrid-2-yl, 2-chloropyrid-3-yl, 3,5-dibromopyrid-2-yl, 2,3,5,6-tetrafluoropyrid-4-yl, 3-chloro-2,5,6-trifluoropyrid-4-yl, 3,5-dichloro-2,6-difluoropyrid-4-yl or 2,6-dichloropyrid-3-yl,
  • Preferred compounds of the formula I are those in which
  • R1 is difluoromethoxy
  • R2 is cyclopropylmethoxy
  • R3 is 2-bromophenyl, 2,6-dichloro-4-ethoxycarbonylphenyl, 2,6-dimethoxyphenyl, 4-cyano-2-fluorophenyl, 2,4,6-trifluorophenyl, 2-chloro-6-methylphenyl, 2,6-dimethylphenyl, 2,6-difluoro-phenyl, 2,6-dichlorophenyl, 3,5-dichloropyrid-4-yl, 3-methylpyrid-2-yl, 2-chloropyrid-3-yl, 3,5-dibromopyrid-2-yl, 2,3,5,6-tetrafluoropyrid-4-yl, 3-chloro-2,5,6-trifluoropyrid-4-yl, 3,5-dichloro-2,6-difluoropyrid-4-yl or 2,6-dichloropyrid-3-yl,
  • a particularly preferred compound of the formula I is the one in which
  • R1 is difluoromethoxy
  • R2 is cyclopropylmethoxy
  • R3 is 3,5-dichloropyrid-4-yl
  • This compound has the chemical name N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (INN: roflumilast).
  • the PDE 4 inhibitor whose solubility is slight is preferably a PDE 4 inhibitor with a solubility in water of less than or equal to 100 milligram/liter, particularly preferably with a solubility in water of less than or equal to 1 milligram/liter, at a temperature of 15 to 25° C., in particular at 21° C.
  • This compound is particularly preferably one of the formula I.
  • suitable pharmaceutical excipients which may be used in the dosage form of the invention are pharmaceutical excipients such as fillers, additional binders, tablet disintegrants or else lubricants and release agents.
  • suitable excipients which may be present in the dosage form of the invention are, for example, flavoring substances (such as flavors and sweeteners), buffer substances, preservatives, coloring substances (such as iron oxid yellow or red) or else emulsifiers.
  • Flavors are usually added in a proportion of from 0.05 to 1% by weight.
  • Other flavoring substances by way of example are acids such as citric acid, sweeteners such as saccharin, aspartame, cyclamate sodium or maltol, which are added according to the desired result.
  • the polyvinylpyrrolidone (PVP) employed according to the invention is, in particular, a water-soluble PVP with an average molecular weight above 2 000, preferably above 20 000.
  • PVP polyvinylpyrrolidone
  • Examples which may be mentioned are Kollidon 12 PF (molecular weight 2 000-3 000), Kollidon 17 PF (molecular weight 7 000-11 000), Kollidon 25 (molecular weight 28 000-34 000), Kollidon 30 (molecular weight 44 000-54 000), Kollidon 90 F (molecular weight 1 000 000-1 500 000).
  • PVP of higher molecular weight such as, for example, Kollidon 25, Kollidon 30 and Kollidon 90 F may be mentioned as preferred.
  • PVP polyvinyl acetate
  • gelatin e.g. Kollidon VA 64
  • corn starch mucilage e.g. corn starch mucilage
  • preswollen starches Starch 1500
  • HPMC hydroxypropyl-methylceflulose
  • L-HPC hydroxypropylcellulose
  • Fillers suitable according to the invention are fillers such as calcium carbonate (e.g. MagGran® CC or Destab® 95) and sodium carbonate, sugar alcohols such as mannitol (e.g. Perlitol® or Parteck0 M), sorbitol (e.g. Karion®), xylitol or maltitol, starches such as corn starch, potato starch and wheat starch, microcrystalline cellulose, saccharides such as glucose, lactose (e.g. lactose monohydrate), levulose, sucrose and dextrose. It is also possible if desired to use mixtures thereof. Corn starch, microcrystalline cellulose and lactose may be mentioned as preferred.
  • sugar alcohols such as mannitol (e.g. Perlitol® or Parteck0 M), sorbitol (e.g. Karion®), xylitol or maltitol, starches such as corn starch, potato starch and wheat starch, microcrystalline cellulose
  • Suitable lubricants and release agents which may be mentioned are sodium stearyl fumarate, magnesium stearate, calcium stearate, stearic acid, talc and colloidal anhydrous silica (Aerosil).
  • the proportion (in percent by weight based on the finished dosage form) of PDE 4 inhibitor in the dosage form of the invention is usually, depending on the nature of the PDE 4 inhibitor, from 0.01 to 50% by weight.
  • the proportion of PDE 4 inhibitor is preferably up to 20% by weight.
  • the proportion (in percent by weight based on the finished dosage form) of binder (PVP and, where appropriate, other binders) may preferably be according to the invention from 0.5 to 20% by weight.
  • the proportion of PVP is preferably from 1 to 5% by weight, particularly preferably 2 to 3% by weight.
  • the proportion (in percent by weight based on the finished dosage form) of filler in the tablet of the invention is advantageously from 40 to 99% by weight.
  • the proportion of filler is preferably from 60 to 97% by weight.
  • the proportion (in percent by weight based on the finished dosage form) of disintegrant in the rapidly disintegrating tablet can usually be up to 35% by weight.
  • the proportion of disintegrant is preferably from 2 to 20% by weight.
  • the proportion of disintegrant is particularly preferably from 5 to 10% by weight.
  • the proportion (in percent by weight based on the finished dosage form) of lubricant or release agent in the rapidly disintegrating tablet is usually from 0.1 to 5% by weight.
  • the proportion of lubricant or release agent is preferably from 0.3 to 3% by weight.
  • the proportion of lubricant or release agent is particularly preferably from 0.5 to 2% by weight.
  • the dosage form is a tablet. It is preferred for the tablet, besides the PDE 4 inhibitor whose solubility is slight and PVP, to comprise as further pharmaceutical excipients at least one filler and at least one lubricant or release agent.
  • the pharmaceutical preparation of the invention can be produced by processes known to the skilled worker for producing tablets and pellets.
  • the pharmaceutical form of the invention is produced by producing a solid solution of the PDE 4 inhibitor whose solubility is slight in the binder PVP as carrier.
  • This can take place for example by the solvent method in which PVP, the PDE 4 inhibitor and, where appropriate, other pharmaceutical excipients are dissolved in a suitable solvent, and the solvent is subsequently removed again by spray drying, normal drying, vacuum drying or freeze-drying. It has been found, surprisingly, that production of the solid solution is also possible by the mixing method in which a PDE 4 inhibitor whose solubility is slight and, where appropriate, other pharmaceutical excipients are vigorously mixed together with PVP.
  • the invention also relates further to a solid solution of a PDE 4 inhibitor whose solubility is slight in the binder PVP as carrier.
  • a solid solution of the PDE 4 inhibitor in the binder PVP as carrier means according to the invention a solid solution with amorphous structure in which the PDE 4 inhibitor is in the form of a molecular dispersion in the carrier material.
  • the solid solution may be processed as active ingredient component together with the filler, binder, disintegrant and lubricant components by production processes familiar to the skilled worker to give the oral dosage forms of the invention.
  • the invention therefore also relates to a process for producing a dosage form in tablet or pellet form for oral administration of a PDE 4 inhibitor, comprising the steps: (a) production of an active ingredient preparation in the form of a solid solution in PVP of the PDE 4 inhibitor whose solubility is slight, (b) production of a mixture of an active ingredient preparation and pharmaceutical excipients and (c) granulation of the mixture obtained in (b) with an aqueous solution of PVP.
  • the granules obtained in (c) can, after drying and mixing with lubricants or release agents, be compressed in a tablet press.
  • the wet granules obtained in (c) can be processed by the extruder/spheroidizer process to suitable pellets.
  • dispersions/suspensions of an active ingredient preparation can be applied in the form of a solid solution In PVP of the PDE 4 inhibitor whose solubility is slight in a suitable solvent to pellet-like carriers (e.g. nonpareils or HPMC-containing pellets).
  • the dosage form of the invention is produced by granulating a mixture of active ingredient and pharmaceutical excipients with an aqueous PVP solution, drying the granules and, if desired, admixing other pharmaceutical excipients. Wet preparations obtained after granulation can then be further processed to pellets and can subsequently be packed into capsules. Dried granules can—if desired after admixture of other pharmaceutical excipients—after mixing with a release agent be compressed in a tablet press. The granulation preferably takes place in a fluidized bed granulator under suitable conditions.
  • the active ingredient is admixed to the other pharmaceutical excipients in the form of a trituration with a pharmaceutical excipient (especially a filler).
  • a pharmaceutical excipient especially a filler
  • Such a trituration can normally be obtained by grinding the active ingredient with a pharmaceutical excipient (especially a filler).
  • the invention therefore also relates to a process for producing a dosage form in tablet or pellet form for oral administration of a PDE 4 inhibitor comprising the steps:
  • the dosage form of the invention is particularly preferably produced by granulation of a mixture of
  • the PDE 4 inhibitor whose solubility is slight is in this case particularly preferably roflumilast, the salts thereof, the N-oxide of the pyridine and salts thereof.
  • the dosage form of the invention is particularly preferably produced by granulation of a mixture of
  • the PDE 4 inhibitor whose solubility is slight is in this case particularly preferably roflumilast, the salts thereof, the N-oxide of the pyridine and salts thereof.
  • the dosage form of the invention is produced by granulation of a mixture of pharmaceutical excipients with a suspension of the active ingredient in an aqueous PVP solution, drying of the granules and, if desired, admixture of further pharmaceutical excipients.
  • the preparations obtained in this way can then, after mixing with a release agent, be compressed in a tablet press.
  • the granulation preferably takes place in a fluidized bed granulator under suitable conditions.
  • the invention therefore also relates to a process for producing a dosage form in tablet or pellet form for oral administration of a PDE 4 inhibitor comprising the steps:
  • the dosage form of the invention is particularly preferably produced by granulation of a mixture of corn starch and lactose monohydrate with a suspension of the PDE 4 inhibitor whose solubility is slight in an aqueous solution of PVP, drying of the granules, mixing of the granules with a release agent and compression in a tablet press.
  • dosage forms of the invention produced employing physical mixtures or triturations of the PDE 4 inhibitor whose solubility is slight with a filler (e.g. by grinding, vigorous mixing or extrusion) and subsequent granulation with aqueous PVP solutions, or produced employing granulation suspensions of PDE 4 inhibitors in aqueous PVP solutions, have similar advantageous properties in relation to the bioavailability of the PDE 4 inhibitor whose solubility is slight as do dosage forms produced by first producing solid solutions of PVP and PDE 4 inhibitor.
  • a filler e.g. by grinding, vigorous mixing or extrusion
  • FIG. 1 shows the time course of the average serum concentration of roflumilast after oral administration of 0.5 mg (2 tablets each containing 0.25 mg) of roflumilast from dosage forms of the invention compared with a dosage form containing no PVP.
  • (1) is mixed with part of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions.
  • (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.1 mg.
  • (1) is mixed with part of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions.
  • (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.125 mg.
  • (1) is mixed with part of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2), (5) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions.
  • (6) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 59.5 mg.
  • (1) is mixed with part of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions.
  • (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.25 mg.
  • (1) is mixed with part of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions.
  • (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.500 mg.
  • (1) is mixed with part of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions.
  • (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 130.5 mg.
  • (1) is mixed with part of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2), (5) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions.
  • (6) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 61.75 mg.
  • (1) is mixed with 70 g of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on.
  • Spray pressure 3 bar; product temperature: 28-33° C.; air flow rate in the first third of the spraying process: 100 m 3 /h; air flow rate subsequently during the spraying process: 150 m 3 /h; inlet air temperature: 40-70° C.; spraying rate: 30-40 g/min).
  • spraying is complete, drying is carried out until the product temperature reaches 34° C.
  • the granules are passed through a stainless steel sieve with a mesh width of 0.8 mm, and the relative surface moisture is measured and adjusted to a value in the range 20-50%. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.1 mg.
  • the granules are passed through a stainless steel sieve with a mesh width of 0.8 mm, and the relative surface moisture is measured and adjusted to a value in the range 20-50%. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.5 mg.
  • (1) is homogeneously suspended in a granulation solution of (4) in purified water.
  • (2) and (3) are put into the product container of a suitable fluidized bed granulation system and granulated with the granulation suspension described above, and then dried.
  • (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.1 mg.
  • (1) is homogeneously suspended in a granulation solution of (4) in purified water.
  • (2) and (3) are put into the product container of a suitable fluidized bed granulation system and granulated with the granulation suspension described above, and then dried.
  • (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.25 mg.
  • a dispersion is produced from (4) and water, and (1) is homogeneously suspended therein. (5) is dissolved in water and added to the dispersion. (2) and (3) are granulated in a suitable fluidized bed granulation system with the dispersion under suitable conditions. (6) is added to this mixture, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.650 mg.
  • (1) is mixed with part of (3), and a trituration is produced in a planetary mill.
  • the trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) and (5) in purified water is sprayed on and dried under suitable conditions.
  • (6) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 66.55 mg.
  • the formulation is produced according to a process disclosed above.
  • Disintegration time the disintegration time was determined using a disintegration tester by the method described in the European Pharmacopoeia.
  • a dispersion is produced from (4) and water, and (1) is homogeneously suspended therein. (2) and (3) are granulated in a suitable fluidized bed granulation system with the dispersion under suitable conditions. (5) is added to the dry granules, and a homogeneous mixture is produced. (5) is added to this mixture, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 96.438 mg.
  • treatment A Tablet corresponding to example D, referred to as “treatment A” hereinafter.
  • treatment B Tablet corresponding to example K, referred to as “treatment B” hereinafter.
  • treatment C Tablet corresponding to example 0, referred to as “treatment C” hereinafter.
  • the dosage forms of the invention can be employed for the treatment and prevention of all diseases regarded as treatable or preventable through the use of PDE 4 inhibitors.
  • Selective cyclic nucleotide phosphodiesterase (PDE) inhibitors are suitable on the one hand as bronchial therapeutic agents (for the treatment of airway obstructions owing to their dilating effect but also owing to their effect increasing the respiratory rate and respiratory drive) and for eliminating erectile dysfunction owing to the vasodilating effect, but on the other hand especially for the treatment of disorders, especially of an inflammatory nature, e.g.
  • mediators such as histamine, PAF (platelet-activating factor), arachidonic acid derivatives such as leukotrienes and prostaglandins, cytokines, interleukins, chemokines, alpha-, beta- and gamma-interferon, tumor necrosis factor (TNF) or oxygen free radicals and proteases.
  • mediators such as histamine, PAF (platelet-activating factor), arachidonic acid derivatives such as leukotrienes and prostaglandins, cytokines, interleukins, chemokines, alpha-, beta- and gamma-interferon, tumor necrosis factor (TNF) or oxygen free radicals and proteases.
  • mediators such as histamine, PAF (platelet-activating factor), arachidonic acid derivatives such as leukotrienes and prostaglandins, cytokines, interleukins, chemokines, alpha-, beta- and gamma-interferon
  • the pharmaceutical preparations of the invention can therefore be used in human and veterinary medicine for example for the treatment and prophylaxis of the following diseases: acute and chronic (especially inflammatory and allergen-induced) airway disorders of various etiologies (bronchitis, allergic bronchitis, bronchial asthma, COPD); dermatoses (especially of a proliferative, inflammatory and allergic nature) such as, for example, psoriasis (vulgaris), toxic and allergic contact eczema, atopic eczema, seborrhoic eczema, lichen simplex, sunburn, pruritus in the genitoanal region, alopecia areata, hypertrophic scars, discoid lupus erythematosus, follicular and extensive pyodermas, endogenous and exogenous acne, acne rosacea and other proliferative, inflammatory and allergic skin disorders; disorders based on excessive release of TNF an le
  • disorders of the arthritic type rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis and other arthritic states
  • disorders of the immune system AIDS, multiple sclerosis
  • types of shock septic shock, endotoxin shock, gram-negative sepsis, toxic shock syndrome and ARDS (adult respiratory distress syndrome)] and generalized inflammations in the gastrointestinal region (Crohn's disease and ulcerative colitis
  • cardiac disorders which can be treated by PDE inhibitors such as, for example, heart failure, or disorders which can be treated owing to the tissue-relaxant effect of PDE inhibitors
  • the invention further relates to a method for the treatment of mammals, including humans, suffering from one of the abovementioned diseases.
  • the method is characterized by administration of a therapeutically effective and pharmacologically suitable amount of a PDE 4 inhibitor to the mammalian patient, the PDE 4 inhibitor being present in a dosage form of the invention.
  • the dosage forms of the invention comprise the PDE 4 inhibitor in the dose customary for the treatment of the particular disease.
  • the dosage of the active ingredient is of the order of magnitude customary for PDE inhibitors, it being possible to administer the daily dose in one or more dosage units.
  • the normal dose on systemic therapy is between 0.001 mg and 3 mg per kilogram and day.
  • Dosage forms preferred according to the invention contain from 0.01 mg to 5 mg of roflumilast, preferably from 0.05 mg to 2.5 mg, particularly preferably 0.1 mg to 0.5 mg of roflumilast per dosage unit.
  • Examples of pharmaceutical preparations of the invention contain 0.1 mg, 0.125 mg, 0.25 mg and 0.5 mg of roflumilast per dosage unit. Normally, one or more than one dosage unit of the invention is administered once a day. If desired, it is also possible for one or more dosage units of the invention to be administered more than once a day.

Abstract

Dosage forms for oral administration of a PDE 4 inhibitor whose solubility is slight are described. They contain PVP as binder.

Description

    TECHNICAL FIELD
  • The present invention relates to the field of pharmaceutical technology and describes a dosage form for oral administration of a PDE 4 inhibitor as active ingredient in tablet or pellet form for treating diseases such as asthma or airway obstructions. The invention additionally relates to processes for producing the dosage form.
  • PRIOR ART
  • Cyclic nucleotide phosphodiesterase (PDE) inhibitors (specifically of type 4) are currently of special interest as a new generation of active ingredients for treating inflammatory disorders, especially inflammations of the airways such as asthma or airway obstructions (such as, for example, COPD=chronic obstructive pulmonary disease). A number of PDE 4 inhibitors is currently undergoing advanced clinical testing.
  • In WO00/50011 and WO01/32165, which relate to dosage forms with controlled or sustained delivery of a PDE 4 inhibitor, it is pointed out that unwanted CNS side effects may become manifest on delivery of certain PDE 4 inhibitors such as Ariflo® (INN: cilomilast) in higher dosages. WO00/50011 and WO01/32165 see this as being a particular risk with immediate release dosage forms of the active ingredient and therefore propose administering the PDE 4 inhibitor Ariflo® (INN: cilomilast) in dosage forms with controlled or sustained release.
  • U.S. Pat. No. 5,286,494 proposes a dosage form with controlled or sustained release for the PDE 4 inhibitor Rolipram whose solubility is slight. However, production of dosage forms with controlled or sustained release of slightly soluble active ingredients may be technically complicated, reference being made thereto for example in U.S. Pat. No. 5,286,494.
  • The solubility of active ingredients of the PDE 4 inhibitor class in water and aqueous systems may, depending on the chemical structure, be low. Thus, the solubility in water found for the PDE 4 inhibitor N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (INN: roflumilast), which is described in WO95/01338, is only 0.53 mg/l at 21° C. The bioavailability of a medicinal substance depends essentially on the release of the medicinal substance from the pharmaceutical form. Faster release and dissolution of the medicinal substance from the formulation means faster absorption thereof. With medicinal substances which are slightly soluble in water, therefore, the bioavailability is frequently limited by the solubility or rate of dissolution. This makes it very difficult to produce suitable dosage forms.
  • DESCRIPTION OF THE INVENTION
  • It is an object of the present invention to provide a dosage form for oral administration of PDE 4 inhibitors whose solubility is slight, which form can be produced without great technical complexity, which takes account of the low solubility of the PDE 4 inhibitor whose solubility is slight, and which results in rapid, acceptable bioavailability of the PDE 4 inhibitor whose solubility is slight, so as to attain serum levels which are required in order to obtain the desired pharmacological effect quickly without side effects becoming manifest.
  • It has now been found, surprisingy, that this object can be achieved by a dosage form for oral administration of a PDE 4 inhibitor whose solubility is slight, employing polyvinylpyrrolidone (PVP) as binder for the dosage form. Compared with dosage forms in which no PVP is employed as binder, the dosage form of the invention shows distinctly improved pharmacokinetic properties. Thus, in particular in relation to the bioavailability of the PDE 4 inhibitor whose solubility is slight, a faster absorption and thus faster onset of the pharmacological effect is observed with the dosage forms of the invention compared with dosage forms without PVP. The oral dosage form of the invention is preferably a solid dosage form in tablet or pellet form. It is preferably a solid oral dosage form with immediate release of the active ingredient (immediate release solid oral dosage form).
  • The invention therefore relates to a dosage form in tablet or pellet form for oral administration of a PDE 4 inhibitor whose solubility is slight, comprising the PDE 4 inhibitor whose solubility is slight together with polvinylpyrrolidone as binder, and one or more other suitable pharmaceutical excipients.
  • The PDE 4 inhibitor whose solubility is slight is preferably according to the invention a compound from the group of compounds of the formula I
  • Figure US20150290178A1-20151015-C00001
  • in which either
  • R1 is 3-7C cyotoalkoxy, 3-7C cycloalkylmethoxy or benzyloxy and
  • R2 is 1-4C alkoxy which is completely or partly substituted by fluorine, or
  • R1 is 1-4C alkoxy which is completely or partly substituted by fluorine and
  • R2 is 3-7C cycloalkylmethoxy or benzyloxy, and
  • R3 is phenyl, pyridyl, phenyl substituted by R31, R32 and R33, or pyridyl substituted by R34, R35, R36 and R37, where
      • R31 is hydroxyl, halogen, cyano, carboxyl, trifluoromethyl, 1-4C alkyl, 1-4C alkoxy, 1-4C alkoxy-carbonyl, 1-4C alkylcarbonyl, 1-4C alkylcarbonyloxy, amino, mono- or di-1-4C alkylamino or 1-4C alkylcarbonylamino,
      • R32 is hydrogen, hydroxyl, halogen, amino, trifluoromethyl, 1-4C alkyl or 1-4C alkoxy,
      • R33 is hydrogen, halogen, 1-4C alkyl or 1-4C alkoxy,
      • R34 is hydroxyl, halogen, cyano, carboxyl, 1-4C alkyl, 1-4C alkoxy, 1-4C alkoxycarbonyl or amino,
      • R35 is hydrogen, halogen, amino or 1-4C alkyl,
      • R36 is hydrogen or halogen and
      • R37 is hydrogen or halogen,
  • the salts of these compounds and the N-oxides of the pyridines and the salts thereof.
  • 3-7C Cycloalkoxy is, for example, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy and cycloheptyloxy, of which cyclopropyloxy, cyclobutyloxy and cyclopentyloxy are preferred.
  • 3-7C Cycloalkylmethoxy is, for example, cyclopropylmethoxy, cyclobutylmethoxy, cyclopentylmethoxy, cyclohexylmethoxy and cycloheptylmethoxy, of which cyclopropylmethoxy, cyclobutylmethoxy and cyclopentylmethoxy are preferred.
  • Examples which may be mentioned of 1-4C alkoxy which is completely or partly substituted by fluorine are 1,2,2-trifluoroethoxy, 2,2,3,3,3-pentafluoropropoxy, perfluoroethoxy and, in particular, 1,1,2,2-tetra-fluoroethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy and difluoromethoxy radicals.
  • Halogen for the purposes of the present invention is bromine, chlorine and fluorine.
  • 1-4C Alkyl represents straight-chain or branched alkyl radicals having 1 to 4 carbon atoms. Examples which may be mentioned are butyl, isobutyl, sec-butyl, tert-butyl, propyl, isopropyl, ethyl and methyl radicals.
  • 1-4C Alkoxy is a radical which, besides the oxygen atom, contains one of the aforementioned
  • 1-4C alkyl radicals. Examples which may be mentioned are the methoxy and ethoxy radicals.
  • 1-4C Alkoxycarbonyl is a carbonyl group to which one of the aforementioned 1-4C alkoxy radicals is bonded. Examples which may be mentioned are the methoxycarbonyl (CH3O—CO—) and ethoxycarbonyl (CH3CH2O—CO—) radicals.
  • 1-4C Alkylcarbonyl is a carbonyl group to which one of the aforementioned 1-4C alkyl radicals is bonded. An example which may be mentioned is the acetyl radical (CH3CO—).
  • 1-4C Alkylcarbonyloxy radicals comprise besides the oxygen atom one of the aforementioned 1-4C alkylcarbonyl radicals. An example which may be mentioned is the acetoxy radical (CH3CO—O—).
  • Examples of mono- or di-1-4C alkylamino radicals which may be mentioned are the methylamino, dimethylamino and diethylamino radicals.
  • An example of a 1-4C alkylcarbonylamino radical which may be mentioned is the acetylamino radical (—NH—CO—CH3).
  • Examples of phenyl radicals substituted by R31, R32 and R33 which may be mentioned are the radicals 2-acetylphenyl, 2-aminophenyl, 2-bromophenyl, 2-chlorophenyl, 2,3-dichlorophenyl, 2,4-dichiorophenyl, 4-diethylamino-2-methylphenyl, 4-bromo-2-trifluoromethylphenyl, 2-carboxy-5-chlorophenyl, 3,5-dichloro-2-hydroxyphenyl, 2-bromo-4-carboxy-5-hydroxyphenyl, 2,6-dichiorophenyl, 2,5-dichiorophenyl, 2,4,6-trichlorophenyl, 2,4,6-trifluorophenyl, 2,6-dibromophenyl, 2-cyanophenyl, 4-cyano-2-fluorophenyl, 2-fluorophenyl, 2,4-difluorophenyl, 2,6-difluorophenyl, 2-chloro-6-fluorophenyl, 2-hydroxyphenyl, 2-hydroxy-4-methoxyphenyl, 2,4-dihydroxyphenyl, 2-methoxyphenyl, 2,3-dimethoxy-phenyl, 2,4-dimethoxyphenyl, 2,6-dimethoxyphenyl, 2-dimethylaminophenyl, 2-methylphenyl, 2-chloro-6-methylphenyl, 2,4-dimethylphenyl, 2,6-dimethylphenyl, 2,3-dimethylphenyl, 2-methoxycarbonylphenyl, 2-trifluoromethylphenyl, 2,6-dichloro-4-methoxyphenyl, 2,6-dichloro-4-cyanophenyl, 2,6-dichloro-4-aminophenyl, 2,6-dichloro-4-methoxycarbonylphenyl, 4-acetylamino-2,6-dichlorophenyl and 2,6-dichloro-4-ethoxycarbonylphenyl.
  • Examples of pyridyl radicals substituted by R34, R35, R36 and R37 which may be mentioned are the radicals 3,5-dichloropyrid-4-yl, 2,6-diaminopyrid-3-yl, 4-aminopyrid-3-yl, 3-methylpyrid-2-yl, 4-methylpyrid-2-yl, 5-hydroxypyrid-2-yl, 4-chloropyrid-3-yl, 3-chloropyrid-2-yl, 3-chloropyrid-4-yl, 2-chloropyrid-3-yl, 2,3,5,6-tetrafluoropyrid-4-yl, 3,5-dichloro-2,6-difluoropyrid-4-yl, 3,5-dibromopyrid-2-yl, 3,5-dibromopyrid-4-yl, 3,5-dichloropyrid-4-yl, 2,6-dichloropyrid-3-yl, 3,5-dimethylpyrid-4-yl, 3-chloro-2,5,6-trifluoropyrid-4-yl and 2,3,5-trifluoropyrid-4-yl.
  • Salts suitable for compounds of the formula I—depending on the substitution—are all acid addition salts but, in particular, all salts with bases. Particular mention may be made of the pharmacologically acceptable salts of the inorganic and organic acids and bases normally used in pharmaceutical technology. Pharmacologically unacceptable salts which, for example, may be the initial products of the process for preparing the compounds of the invention on the industrial scale are converted into pharmacologically acceptable salts by processes known to the skilled worker. Those suitable on the one hand are water-soluble and water-insoluble acid addition salts with acids such as, for example, hydrochloric acid, hydrobromic acid, phosphoric acid, nitric acid, sulfuric acid, acetic acid, citric acid, D-gluconic acid, benzoic acid, 2-(4-hydroxybenzoyl)benzoic acid, butyric acid, sulfosalicylic acid, maleic acid, lauric acid, malic acid, fumaric acid, succinic acid, oxalic acid, tartaric acid, embonic acid, stearic acid, toluenesulfonic acid, methanesulfonic acid, or 3-hydroxy-2-naphthoic acid, the acids being employed to prepare the salts in the equimolar ratio of amounts, or one differing therefrom—depending on whether the acid is monobasic or polybasic and depending on which salt is desired.
  • On the other hand, salts with bases are also particularly suitable. Examples of basic salts which may be mentioned are lithium, sodium, potassium, calcium, aluminum, magnesium, titanium, ammonium, meglumine or guanidinium salts, once again the bases being employed to prepare the salts in the equimolar ratio of amounts or one differeing therefrom.
  • Compounds of the formula Ito be emphasized are those in which either
  • R1 is 3-5C cycloalkoxy, 3-5C cycloalkylmethoxy or benzyloxy and
  • R2 is 1-4C alkoxy which is completely or partly substituted by fluorine, or
  • R1 is 1-4C alkoxy which is completely or partly substituted by fluorine and
  • R2 is 3-5C cycloalkylmethoxy or benzyloxy, and
  • R3 is phenyl, pyridyl, phenyl substituted by R31, R32 and R33, or pyridyl substituted by R34, R35, R36 and R37, where
      • R31 is halogen, cyano, carboxyl, 1-4C alkyl, 1-4C alkoxy or 1-4C alkoxycarbonyl,
      • R32 is hydrogen, halogen, 1-4C alkyl or 1-4C alkoxy,
      • R33 is hydrogen, halogen, 1-4C alkyl or 1-4C alkoxy,
      • R34 is halogen or 1-4C alkyl,
      • R35 is hydrogen or halogen,
      • R36 is hydrogen or halogen and
      • R37 is hydrogen or halogen,
  • the salts of these compounds, and the N-oxides of the pyridines and salts thereof.
  • Compounds of the formula Ito be particularly emphasized are those in which either
  • R1 is 3-5C cycloalkoxy, 3-5C cycloalkylmethoxy or benzyloxy and
  • R2 is 1-4C alkoxy which is completely or partly substituted by fluorine, or
  • R1 is 1-4C alkoxy which is completely or partly substituted by fluorine and
  • R2 is 3-5C cycloalkylmethoxy or benzyloxy and
  • R3 is 2-bromophenyl, 2,6-dichloro-4-ethoxycarbonylphenyl, 2,6-dimethoxyphenyl, 4-cyano-2-fluoro-phenyl, 2,4,6-trifluorophenyl, 2-chloro-6-methylphenyl, 2,6-dimethylphenyl, 2,6-difluorophenyl, 2,6-dichlorophenyl, 3,5-dichloropyrid-4-yl, 3-methylpyrid-2-yl, 2-chloropyrid-3-yl, 3,5-dibromopyrid-2-yl, 2,3,5,6-tetrafluoropyrid-4-yl, 3-chloro-2,5,6-trifluoropyrid-4-yl, 3,5-dichloro-2,6-difluoropyrid-4-yl or 2,6-dichloropyrid-3-yl,
  • the salts of these compounds, and the N-oxides of the pyridines and salts thereof.
  • Preferred compounds of the formula I are those in which
  • R1 is difluoromethoxy,
  • R2 is cyclopropylmethoxy and
  • R3 is 2-bromophenyl, 2,6-dichloro-4-ethoxycarbonylphenyl, 2,6-dimethoxyphenyl, 4-cyano-2-fluorophenyl, 2,4,6-trifluorophenyl, 2-chloro-6-methylphenyl, 2,6-dimethylphenyl, 2,6-difluoro-phenyl, 2,6-dichlorophenyl, 3,5-dichloropyrid-4-yl, 3-methylpyrid-2-yl, 2-chloropyrid-3-yl, 3,5-dibromopyrid-2-yl, 2,3,5,6-tetrafluoropyrid-4-yl, 3-chloro-2,5,6-trifluoropyrid-4-yl, 3,5-dichloro-2,6-difluoropyrid-4-yl or 2,6-dichloropyrid-3-yl,
  • the salts of these compounds, and the N-oxides of the pyridines and salts thereof.
  • A particularly preferred compound of the formula I is the one in which
  • R1 is difluoromethoxy,
  • R2 is cyclopropylmethoxy and
  • R3 is 3,5-dichloropyrid-4-yl,
  • the salts of these compounds, and the N-oxide of the pyridine and salts thereof.
  • This compound has the chemical name N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (INN: roflumilast).
  • The PDE 4 inhibitor whose solubility is slight is preferably a PDE 4 inhibitor with a solubility in water of less than or equal to 100 milligram/liter, particularly preferably with a solubility in water of less than or equal to 1 milligram/liter, at a temperature of 15 to 25° C., in particular at 21° C. This compound is particularly preferably one of the formula I.
  • The abovementioned compounds of the formula I and the use of these compounds as phosphodiesterase (PDE) 4 inhibitors are described in the international patent application WO95/01338.
  • Further suitable pharmaceutical excipients which may be used in the dosage form of the invention are pharmaceutical excipients such as fillers, additional binders, tablet disintegrants or else lubricants and release agents. Other suitable excipients which may be present in the dosage form of the invention are, for example, flavoring substances (such as flavors and sweeteners), buffer substances, preservatives, coloring substances (such as iron oxid yellow or red) or else emulsifiers. Flavors are usually added in a proportion of from 0.05 to 1% by weight. Other flavoring substances by way of example are acids such as citric acid, sweeteners such as saccharin, aspartame, cyclamate sodium or maltol, which are added according to the desired result.
  • The polyvinylpyrrolidone (PVP) employed according to the invention is, in particular, a water-soluble PVP with an average molecular weight above 2 000, preferably above 20 000. Examples which may be mentioned are Kollidon 12 PF (molecular weight 2 000-3 000), Kollidon 17 PF (molecular weight 7 000-11 000), Kollidon 25 (molecular weight 28 000-34 000), Kollidon 30 (molecular weight 44 000-54 000), Kollidon 90 F (molecular weight 1 000 000-1 500 000). PVP of higher molecular weight such as, for example, Kollidon 25, Kollidon 30 and Kollidon 90 F may be mentioned as preferred.
  • It is possible if desired to employ in addition to PVP other binders such as polyvinyl acetate (e.g. Kollidon VA 64), gelatin, corn starch mucilage, preswollen starches (Starch 1500), hydroxypropyl-methylceflulose (HPMC) or hydroxypropylcellulose (L-HPC).
  • Fillers suitable according to the invention are fillers such as calcium carbonate (e.g. MagGran® CC or Destab® 95) and sodium carbonate, sugar alcohols such as mannitol (e.g. Perlitol® or Parteck0 M), sorbitol (e.g. Karion®), xylitol or maltitol, starches such as corn starch, potato starch and wheat starch, microcrystalline cellulose, saccharides such as glucose, lactose (e.g. lactose monohydrate), levulose, sucrose and dextrose. It is also possible if desired to use mixtures thereof. Corn starch, microcrystalline cellulose and lactose may be mentioned as preferred.
  • Examples of suitable lubricants and release agents which may be mentioned are sodium stearyl fumarate, magnesium stearate, calcium stearate, stearic acid, talc and colloidal anhydrous silica (Aerosil).
  • Disintegrants suitable according to the invention are, in particular, insoluble polyvinylpyrrolidone (insoluble PVP, crospovidone), carboxymethylstarch sodium [=sodium starch glycolate], sodium carboxymethylcellulose, alginic acid, and starches able to carry out the function of a disintegrant (e.g. Starch 1500).
  • The proportion (in percent by weight based on the finished dosage form) of PDE 4 inhibitor in the dosage form of the invention is usually, depending on the nature of the PDE 4 inhibitor, from 0.01 to 50% by weight. The proportion of PDE 4 inhibitor is preferably up to 20% by weight.
  • The proportion (in percent by weight based on the finished dosage form) of binder (PVP and, where appropriate, other binders) may preferably be according to the invention from 0.5 to 20% by weight. The proportion of PVP is preferably from 1 to 5% by weight, particularly preferably 2 to 3% by weight.
  • The proportion (in percent by weight based on the finished dosage form) of filler in the tablet of the invention is advantageously from 40 to 99% by weight. The proportion of filler is preferably from 60 to 97% by weight.
  • The proportion (in percent by weight based on the finished dosage form) of disintegrant in the rapidly disintegrating tablet can usually be up to 35% by weight. The proportion of disintegrant is preferably from 2 to 20% by weight. The proportion of disintegrant is particularly preferably from 5 to 10% by weight.
  • The proportion (in percent by weight based on the finished dosage form) of lubricant or release agent in the rapidly disintegrating tablet is usually from 0.1 to 5% by weight. The proportion of lubricant or release agent is preferably from 0.3 to 3% by weight. The proportion of lubricant or release agent is particularly preferably from 0.5 to 2% by weight.
  • In a preferred embodiment of the invention, the dosage form is a tablet. It is preferred for the tablet, besides the PDE 4 inhibitor whose solubility is slight and PVP, to comprise as further pharmaceutical excipients at least one filler and at least one lubricant or release agent.
  • The pharmaceutical preparation of the invention can be produced by processes known to the skilled worker for producing tablets and pellets.
  • In one embodiment of the invention, the pharmaceutical form of the invention is produced by producing a solid solution of the PDE 4 inhibitor whose solubility is slight in the binder PVP as carrier. This can take place for example by the solvent method in which PVP, the PDE 4 inhibitor and, where appropriate, other pharmaceutical excipients are dissolved in a suitable solvent, and the solvent is subsequently removed again by spray drying, normal drying, vacuum drying or freeze-drying. It has been found, surprisingly, that production of the solid solution is also possible by the mixing method in which a PDE 4 inhibitor whose solubility is slight and, where appropriate, other pharmaceutical excipients are vigorously mixed together with PVP.
  • The invention also relates further to a solid solution of a PDE 4 inhibitor whose solubility is slight in the binder PVP as carrier. A solid solution of the PDE 4 inhibitor in the binder PVP as carrier means according to the invention a solid solution with amorphous structure in which the PDE 4 inhibitor is in the form of a molecular dispersion in the carrier material.
  • In the event of further processing of a solid solution to tablets or pellets, the solid solution may be processed as active ingredient component together with the filler, binder, disintegrant and lubricant components by production processes familiar to the skilled worker to give the oral dosage forms of the invention.
  • The invention therefore also relates to a process for producing a dosage form in tablet or pellet form for oral administration of a PDE 4 inhibitor, comprising the steps: (a) production of an active ingredient preparation in the form of a solid solution in PVP of the PDE 4 inhibitor whose solubility is slight, (b) production of a mixture of an active ingredient preparation and pharmaceutical excipients and (c) granulation of the mixture obtained in (b) with an aqueous solution of PVP.
  • In the case of dosage forms of the invention in the form of tablets, the granules obtained in (c) can, after drying and mixing with lubricants or release agents, be compressed in a tablet press. In the case of dosage forms of the invention in the form of pellets, the wet granules obtained in (c) can be processed by the extruder/spheroidizer process to suitable pellets. Alternatively, dispersions/suspensions of an active ingredient preparation can be applied in the form of a solid solution In PVP of the PDE 4 inhibitor whose solubility is slight in a suitable solvent to pellet-like carriers (e.g. nonpareils or HPMC-containing pellets).
  • In another preferred embodiment of the invention, the dosage form of the invention is produced by granulating a mixture of active ingredient and pharmaceutical excipients with an aqueous PVP solution, drying the granules and, if desired, admixing other pharmaceutical excipients. Wet preparations obtained after granulation can then be further processed to pellets and can subsequently be packed into capsules. Dried granules can—if desired after admixture of other pharmaceutical excipients—after mixing with a release agent be compressed in a tablet press. The granulation preferably takes place in a fluidized bed granulator under suitable conditions. It is moreover possible if desired for the active ingredient to be admixed to the other pharmaceutical excipients in the form of a trituration with a pharmaceutical excipient (especially a filler). This is particularly preferred when the active ingredient content in the dosage form is less than 5% by weight. Such a trituration can normally be obtained by grinding the active ingredient with a pharmaceutical excipient (especially a filler).
  • The invention therefore also relates to a process for producing a dosage form in tablet or pellet form for oral administration of a PDE 4 inhibitor comprising the steps:
  • (a) production of a mixture of active ingredient and pharmaceutical excipients and
  • (b) granulation of the mixture obtained in (a) with an aqueous solution of PVP.
  • The dosage form of the invention is particularly preferably produced by granulation of a mixture of
  • (a) PDE 4 inhibitor whose solubility is slight, or a trituration of the PDE 4 inhibitor whose solubility is slight with corn starch,
  • (b) corn starch and
  • (c) lactose monohydrate
  • with an aqueous PVP solution, drying of the granules, mixing of the granules with a release agent and compression in a tablet press. The PDE 4 inhibitor whose solubility is slight is in this case particularly preferably roflumilast, the salts thereof, the N-oxide of the pyridine and salts thereof.
  • Alternatively, the dosage form of the invention is particularly preferably produced by granulation of a mixture of
  • (a) PDE 4 inhibitor whose solubility is slight, or a trituration of the PDE 4 inhibitor whose solubility is slight with corn starch,
  • (b) corn starch,
  • (c) microcrystalline cellulose and
  • (d) sodium carboxymethylstarch
  • with an aqueous PVP solution, drying of the granules, mixing of the granules with a release agent and compression in a tablet press. The PDE 4 inhibitor whose solubility is slight is in this case particularly preferably roflumilast, the salts thereof, the N-oxide of the pyridine and salts thereof.
  • In a further preferred embodiment of the invention, the dosage form of the invention is produced by granulation of a mixture of pharmaceutical excipients with a suspension of the active ingredient in an aqueous PVP solution, drying of the granules and, if desired, admixture of further pharmaceutical excipients. The preparations obtained in this way can then, after mixing with a release agent, be compressed in a tablet press. The granulation preferably takes place in a fluidized bed granulator under suitable conditions.
  • The invention therefore also relates to a process for producing a dosage form in tablet or pellet form for oral administration of a PDE 4 inhibitor comprising the steps:
  • (a) production of a mixture of pharmaceutical excipients and
  • (b) granulation of the mixture obtained in (a) with a suspension of the active ingredient in an aqueous solution of PVP.
  • The dosage form of the invention is particularly preferably produced by granulation of a mixture of corn starch and lactose monohydrate with a suspension of the PDE 4 inhibitor whose solubility is slight in an aqueous solution of PVP, drying of the granules, mixing of the granules with a release agent and compression in a tablet press.
  • It has surprisingly been found that dosage forms of the invention produced employing physical mixtures or triturations of the PDE 4 inhibitor whose solubility is slight with a filler (e.g. by grinding, vigorous mixing or extrusion) and subsequent granulation with aqueous PVP solutions, or produced employing granulation suspensions of PDE 4 inhibitors in aqueous PVP solutions, have similar advantageous properties in relation to the bioavailability of the PDE 4 inhibitor whose solubility is slight as do dosage forms produced by first producing solid solutions of PVP and PDE 4 inhibitor. This suggests that in the production of the dosage forms of the invention based on physical mixtures or triturations of the PDE 4 inhibitor whose solubility is slight with a filler, which are subsequently granulated with aqueous PVP solutions, or in whose preparation granulation suspensions of PDE 4 inhibitors in aqueous PVP solutions are employed, there are, surprisingly, interactions between PVP and PDE 4 inhibitor whose solubility is slight, like those occurring in the solid solution of PVP and PDE 4 inhibitor. In the production of the dosage forms of the invention it is therefore also possible to dispense with the more technically elaborate variant of production of a solid solution by the solvent method.
  • DESCRIPTION OF THE FIGURE
  • FIG. 1 shows the time course of the average serum concentration of roflumilast after oral administration of 0.5 mg (2 tablets each containing 0.25 mg) of roflumilast from dosage forms of the invention compared with a dosage form containing no PVP.
  • The production of tablets and preparations of the invention is described by way of example below. The following examples explain the invention in more detail without restricting it.
  • EXAMPLES Production of Tablets of the Invention Example A Weight Based on a Tablet Containing 0.1 mg of Roflumilast
  • 1. Roflumilast (micronized) 0.100 mg
    2. Lactose monohydrate 49.660 mg
    3. Corn starch 13.390 mg
    4. Polyvidone K90 1.300 mg
    5. Magnesium stearate (vegetable) 0.650 mg
    Total 65.100 mg
  • Production: (1) is mixed with part of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.1 mg.
  • Example B Weight Based on a Tablet Containing 0.125 mg of Roflumilast
  • 1. Roflumilast 0.125 mg
    2. Lactose monohydrate 49.660 mg
    3. Corn starch 13.390 mg
    4. Polyvidone K90 1.300 mg
    5. Magnesium stearate (vegetable) 0.650 mg
    Total 65.125 mg
  • Production: (1) is mixed with part of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.125 mg.
  • Example C
  • Weight Based on a Tablet Containing 0.25 mg of Roflumilast
  • 1. Roflumilast 0.250 mg
    2. Microcrystalline cellulose 33.900 mg
    3. Corn starch 2.500 mg
    4. Polyvidone K90 2.250 mg
    5. Sodium carboxymethylstarch (type A) 20.000 mg
    6. Magnesium stearate (vegetable) 0.600 mg
    Total 59.500 mg
  • Production: (1) is mixed with part of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2), (5) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions. (6) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 59.5 mg.
  • Example D Weight Based on a Tablet Containing 0.25 mg of Roflumilast
  • 1. Roflumilast 0.250 mg
    2. Lactose monohydrate 49.660 mg
    3. Corn starch 13.390 mg
    4. Polyvidone K90 1.300 mg
    5. Magnesium stearate (vegetable) 0.650 mg
    Total 65.250 mg
  • Production: (1) is mixed with part of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.25 mg.
  • Example E Weight Based on a Tablet Containing 0.5 mg of Roflumilast
  • 1. Roflumilast 0.500 mg
    2. Lactose monohydrate 49.660 mg
    3. Corn starch 13.390 mg
    4. Polyvidone K90 1.300 mg
    5. Magnesium stearate (vegetable) 0.650 mg
    Total 65.500 mg
  • Production: (1) is mixed with part of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.500 mg.
  • Example F Weight Based on a Tablet Containing 0.5 mg of Roflumilast
  • 1. Roflumilast 0.500 mg
    2. Lactose monohydrate 99.320 mg
    3. Corn starch 26.780 mg
    4. Polyvidone K90 2.600 mg
    5. Magnesium stearate (vegetable) 1.300 mg
    Total 130.500 mg
  • Production: (1) is mixed with part of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 130.5 mg.
  • Example G Weight Based on a Tablet Containing 2.5 mg of Roflumilast
  • 1. Roflumilast 2.500 mg
    2. Microcrystalline cellulose 33.900 mg
    3. Corn starch 2.500 mg
    4. Polyvidone K90 2.250 mg
    5. Sodium carboxymethylstarch (type A) 20.000 mg
    6. Magnesium stearate (vegetable) 0.600 mg
    Total 61.750 mg
  • Production: (1) is mixed with part of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2), (5) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on and dried under suitable conditions. (6) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 61.75 mg.
  • Example H Production of Tablets Containing 0.1 mg of Roflumilast as Active Ingredient (Weight for a Batch of 70 000 Tablets)
  • 1. Roflumilast (micronized) 7.000 g
    2. Lactose monohydrate 3476.200 g
    3. Corn starch 937.300 g
    4. Polyvidone K90 91.000 g
    5. Magnesium stearate (vegetable) 45.500 g
    Total 4557.000 g
  • Production: (1) is mixed with 70 g of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on. (Spraying pressure: 3 bar; product temperature: 28-33° C.; air flow rate in the first third of the spraying process: 100 m3/h; air flow rate subsequently during the spraying process: 150 m3/h; inlet air temperature: 40-70° C.; spraying rate: 30-40 g/min). After spraying is complete, drying is carried out until the product temperature reaches 34° C. The granules are passed through a stainless steel sieve with a mesh width of 0.8 mm, and the relative surface moisture is measured and adjusted to a value in the range 20-50%. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.1 mg.
  • Example I Production of Tablets Containing 0.25 mg of Roflumilast as Active Ingredient (Weight for a Batch of 70 000 Tablets)
  • 1. Roflumilast (micronized) 35.000 g
    2. Lactose monohydrate 3476.200 g
    3. Corn starch 937.300 g
    4. Polyvidone K90 91.000 g
    5. Magnesium stearate (vegetable) 45.500 g
    Total 4585.000 g
  • Production: 19.25 g of (1) are mixed with 192.5 g of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) in purified water is sprayed on. (Spraying pressure: 3 bar; product temperature: 28-33° C.; air flow rate in the first third of the spraying process: 100 m3/h; air flow rate subsequently during the spraying process: 150 m3/h; inlet air temperature: 40-70° C.; spraying rate: 30-40 g/min). After spraying is complete, drying is carried out until the product temperature reaches 34° C. The granules are passed through a stainless steel sieve with a mesh width of 0.8 mm, and the relative surface moisture is measured and adjusted to a value in the range 20-50%. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.5 mg.
  • Example J Production of Tablets Containing 0.1 mg of Roflumilast as Active Ingredient (Weight for a Batch of 70 000 Tablets)
  • 1. Roflumilast (micronized) 7.000 g
    2. Lactose monohydrate 3476.200 g
    3. Corn starch 937.300 g
    4. Polyvidone K90 91.000 g
    5. Magnesium stearate (vegetable) 45.500 g
    Total 4557.000 g
  • Production: (1) is homogeneously suspended in a granulation solution of (4) in purified water. (2) and (3) are put into the product container of a suitable fluidized bed granulation system and granulated with the granulation suspension described above, and then dried. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.1 mg.
  • Example K Production of Tablets Containing 0.25 mg of Roflumilast as Active Ingredient (Weight for a Batch of 70 000 Tablets)
  • 1. Roflumilast (micronized) 35.000 g
    2. Lactose monohydrate 3476.200 g
    3. Corn starch 937.300 g
    4. Polyvidone K90 91.000 g
    5. Magnesium stearate (vegetable) 45.500 g
    Total 4585.000 g
  • Production: (1) is homogeneously suspended in a granulation solution of (4) in purified water. (2) and (3) are put into the product container of a suitable fluidized bed granulation system and granulated with the granulation suspension described above, and then dried. (5) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.25 mg.
  • Example L Weight Based on a Tablet Containing 0.25 mg of Roflumilast
  • 1. Roflumilast 0.250 mg
    2. Lactose monohydrate 49.660 mg
    3. Potato starch 10.000 mg
    4. Corn starch 3.590 mg
    5. PVP 25 1.500 mg
    6. Magnesium stearate (vegetable) 0.650 mg
    Total 65.650 mg
  • Production: A dispersion is produced from (4) and water, and (1) is homogeneously suspended therein. (5) is dissolved in water and added to the dispersion. (2) and (3) are granulated in a suitable fluidized bed granulation system with the dispersion under suitable conditions. (6) is added to this mixture, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 65.650 mg.
  • Example M Weight Based on a Tablet Containing 0.25 mg of Roflumilast
  • 1. Roflumilast 0.250 mg
    2. Lactose monohydrate 49.660 mg
    3. Corn starch 13.390 mg
    4. Polyvidone K90 1.300 mg
    5. Gelatin 1.300 mg
    6. Magnesium stearate (vegetable) 0.650 mg
    Total 66.550 mg
  • Production: (1) is mixed with part of (3), and a trituration is produced in a planetary mill. The trituration is put together with (2) and the remaining amount of (3) in the product container of a fluidized bed granulation system, and a 5% granulation solution of (4) and (5) in purified water is sprayed on and dried under suitable conditions. (6) is added to the granules, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 66.55 mg.
  • Example M1 Formulation for Pediatric Use Weight Based on a Tablet Containing 0.125 mg of Roflumilast
  • 1. Roflumilast 0.125 mg
    2. Lactose monohydrate 49.660 mg
    3. Corn starch 13.390 mg
    4. Polyvidone K90 1.300 mg
    5. Mannit 32.238 mg
    6. Flavor (Tutti Frutti) 0.329 mg
    7. PVP (insoluble) 12.895 mg
    5. Magnesium stearate (vegetable) 1.649 mg
    Total 111.586 mg
  • The formulation is produced according to a process disclosed above.
  • Physical Investigations and Comparative Tests with Dosage Forms in Which no PVP was Used as Binder
  • Example N
  • The disintegration time and the release of active ingredient were determined for a dosage form corresponding to example D.
  • Disintegration time: the disintegration time was determined using a disintegration tester by the method described in the European Pharmacopoeia.
  • Result: 7.08 minutes.
  • Release of active ingredient: the release of active ingredient was determined as described in the US Pharmacopeia (USP XXV; apparatus 2).
  • Result: 78% of the active ingredient are released after 15 minutes, and quantitative release is observed after 60 minutes.
  • Example O
  • Production of a dosage form containing roflumilast in which no PVP is used:
  • Weight Based on a Tablet Containing 0.25 mg of Roflumilast
  • 1. Roflumilast 0.250 mg
    2. Lactose monohydrate 70.300 mg
    3. Potato starch 19.475 mg
    4. Corn starch 3.563 mg
    5. Sodium carboxymethylstarch (Type A) 1.900 mg
    6. Magnesium stearate (vegetable) 0.950 mg
    Total 96.438 mg
  • Production: A dispersion is produced from (4) and water, and (1) is homogeneously suspended therein. (2) and (3) are granulated in a suitable fluidized bed granulation system with the dispersion under suitable conditions. (5) is added to the dry granules, and a homogeneous mixture is produced. (5) is added to this mixture, and the mixture obtained after mixing is compressed in a tablet press to tablets having an average weight of 96.438 mg.
  • Comparative Study
  • Design: 24 subjects, 3-period changeover, randomized; dose in each case 0.5 mg (2 tablets each containing 0.25 mg of roflumilast). The serum concentration of roflumilast after oral administration of 0.5 mg (2 tablets each containing 0.25 mg) of roflumilast was investigated for the following dosage forms:
  • With PVP as Binder:
  • Tablet corresponding to example D, referred to as “treatment A” hereinafter.
  • Tablet corresponding to example K, referred to as “treatment B” hereinafter.
  • Without PVP as Binder:
  • Tablet corresponding to example 0, referred to as “treatment C” hereinafter.
  • The results are depicted in FIG. 1. Higher serum levels were observed considerably more quickly after oral administration for dosage forms with PVP as binder compared with dosage forms without PVP. The rate of absorption is thus distinctly increased for the dosage forms of the invention.
  • INDUSTRIAL APPLICABILITY
  • The dosage forms of the invention can be employed for the treatment and prevention of all diseases regarded as treatable or preventable through the use of PDE 4 inhibitors. Selective cyclic nucleotide phosphodiesterase (PDE) inhibitors (specifically of type 4) are suitable on the one hand as bronchial therapeutic agents (for the treatment of airway obstructions owing to their dilating effect but also owing to their effect increasing the respiratory rate and respiratory drive) and for eliminating erectile dysfunction owing to the vasodilating effect, but on the other hand especially for the treatment of disorders, especially of an inflammatory nature, e.g. of the airways (asthma prophylaxis), of the skin, of the central nervous system, of the intestine, of the eyes and of the joints, which are promoted by mediators such as histamine, PAF (platelet-activating factor), arachidonic acid derivatives such as leukotrienes and prostaglandins, cytokines, interleukins, chemokines, alpha-, beta- and gamma-interferon, tumor necrosis factor (TNF) or oxygen free radicals and proteases. The pharmaceutical preparations of the invention can therefore be used in human and veterinary medicine for example for the treatment and prophylaxis of the following diseases: acute and chronic (especially inflammatory and allergen-induced) airway disorders of various etiologies (bronchitis, allergic bronchitis, bronchial asthma, COPD); dermatoses (especially of a proliferative, inflammatory and allergic nature) such as, for example, psoriasis (vulgaris), toxic and allergic contact eczema, atopic eczema, seborrhoic eczema, lichen simplex, sunburn, pruritus in the genitoanal region, alopecia areata, hypertrophic scars, discoid lupus erythematosus, follicular and extensive pyodermas, endogenous and exogenous acne, acne rosacea and other proliferative, inflammatory and allergic skin disorders; disorders based on excessive release of TNF an leukotrienes, e.g. disorders of the arthritic type (rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis and other arthritic states), disorders of the immune system (AIDS, multiple sclerosis), types of shock [septic shock, endotoxin shock, gram-negative sepsis, toxic shock syndrome and ARDS (adult respiratory distress syndrome)] and generalized inflammations in the gastrointestinal region (Crohn's disease and ulcerative colitis); disorders based on allergic and/or chronic abnormal immunological reactions in the region of the upper airways (pharyngeal space, nose) and adjacent regions (paranasal sinuses, eyes), such as, for example, allergic rhinitis/sinusitis, chronic rhinitis/sinusitis, allergic conjunctivitis and nasal polyps; but also cardiac disorders which can be treated by PDE inhibitors, such as, for example, heart failure, or disorders which can be treated owing to the tissue-relaxant effect of PDE inhibitors, such as, for example, erectile dysfunction or colic of the kidneys and ureters connected with kidney stones; or else disorders of the CNS such as, for example, depressions or arteriosclerotic dementia.
  • The invention further relates to a method for the treatment of mammals, including humans, suffering from one of the abovementioned diseases. The method is characterized by administration of a therapeutically effective and pharmacologically suitable amount of a PDE 4 inhibitor to the mammalian patient, the PDE 4 inhibitor being present in a dosage form of the invention. The disease is preferably asthma or airway obstructions, especially COPD (=chronic obstructive pulmonary disease).
  • The dosage forms of the invention comprise the PDE 4 inhibitor in the dose customary for the treatment of the particular disease. The dosage of the active ingredient is of the order of magnitude customary for PDE inhibitors, it being possible to administer the daily dose in one or more dosage units. The normal dose on systemic therapy (oral) is between 0.001 mg and 3 mg per kilogram and day. Dosage forms preferred according to the invention contain from 0.01 mg to 5 mg of roflumilast, preferably from 0.05 mg to 2.5 mg, particularly preferably 0.1 mg to 0.5 mg of roflumilast per dosage unit. Examples of pharmaceutical preparations of the invention contain 0.1 mg, 0.125 mg, 0.25 mg and 0.5 mg of roflumilast per dosage unit. Normally, one or more than one dosage unit of the invention is administered once a day. If desired, it is also possible for one or more dosage units of the invention to be administered more than once a day.

Claims (12)

1.-11. (canceled)
12. A method for the treatment of chronic obstructive pulmonary disease (COPD), comprising:
orally administering to a patient suffering from chronic obstructive pulmonary disease an immediate release solid dosage form including between 250 and 500 μg of a PDE 4 inhibitor that is N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (roflumilast) or a salt thereof, and one or more suitable pharmaceutical excipients.
13. The method of treatment according to claim 12, wherein the PDE 4 inhibitor is roflumilast.
14. The method of treatment according to claim 12, wherein the immediate release solid dosage form is a tablet.
15. The method of treatment of claim 14, wherein the PDE4 inhibitor is roflumilast.
16. A method for the treatment of chronic obstructive pulmonary disease (COPD), comprising:
orally administering to a patient suffering from chronic obstructive pulmonary disease an immediate release oral solid dosage form in tablet or pellet form including between 250 and 500 μg of N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (roflumilast) or a salt thereof, and one or more suitable pharmaceutical excipients.
17. The method of treatment of claim 16 wherein said immediate release oral dosage form is administered once per day.
18. The method of treatment of claim 16, wherein said immediate release oral solid dosage form in tablet or pellet form contains 500 μg of N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (roflumilast).
19. The method of treatment of claim 17, wherein said immediate release oral solid dosage form in tablet or pellet form contains 500 μg of N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (roflumilast).
20. The method of treatment of claim 12, wherein said immediate release oral solid dosage form contains 500 μg of N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (roflumilast).
21. The method of treatment of claim 12 wherein said immediate release oral dosage form is administered once per day.
22. The method of treatment of claim 21, wherein said immediate release oral solid dosage form contains 500 μg of N-(3,5-dichloropyrid-4-yl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide (roflumilast).
US14/731,964 2002-02-20 2015-06-05 Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient Abandoned US20150290178A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/731,964 US20150290178A1 (en) 2002-02-20 2015-06-05 Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
EP02003811 2002-02-20
EP02003811.3 2002-02-20
DE2002107160 DE10207160A1 (en) 2002-02-20 2002-02-20 Dosage form useful for treating diseases e.g. psoriasis, allergic contact eczema, atopic eczema, sunburn and pruritis comprises phosphodiesterase inhibitor and polyvinylpyrrolidone
DE10207160.8 2002-02-20
PCT/EP2003/001650 WO2003070279A1 (en) 2002-02-20 2003-02-19 Oral dosage form containing a pde 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US50513804A 2004-08-19 2004-08-19
US13/008,842 US8431154B2 (en) 2002-02-20 2011-01-18 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidone as excipient
US13/874,065 US9468598B2 (en) 2002-02-20 2013-04-30 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US14/038,678 US20140031397A1 (en) 2002-02-20 2013-09-26 Oral Dosage Form Containing A Pde 4 Inhibitor As An Active Ingredient And Polyvinylpyrrolidon As Excipient
US14/731,964 US20150290178A1 (en) 2002-02-20 2015-06-05 Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/038,678 Continuation US20140031397A1 (en) 2002-02-20 2013-09-26 Oral Dosage Form Containing A Pde 4 Inhibitor As An Active Ingredient And Polyvinylpyrrolidon As Excipient

Publications (1)

Publication Number Publication Date
US20150290178A1 true US20150290178A1 (en) 2015-10-15

Family

ID=27758398

Family Applications (11)

Application Number Title Priority Date Filing Date
US10/505,138 Expired - Lifetime US7951397B2 (en) 2002-02-20 2003-02-19 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US11/501,836 Abandoned US20060269600A1 (en) 2002-02-20 2006-08-10 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US12/876,996 Abandoned US20110060016A1 (en) 2002-02-20 2010-09-07 Oral dosage form containing a pde 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US13/008,842 Expired - Lifetime US8431154B2 (en) 2002-02-20 2011-01-18 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidone as excipient
US13/739,457 Abandoned US20130131123A1 (en) 2002-02-20 2013-01-11 Oral Dosage Form Containing A Pde 4 Inhibitor As An Active Ingredient And Polyvinylpyrrolidon As Excipient
US13/874,065 Expired - Lifetime US9468598B2 (en) 2002-02-20 2013-04-30 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US14/038,678 Abandoned US20140031397A1 (en) 2002-02-20 2013-09-26 Oral Dosage Form Containing A Pde 4 Inhibitor As An Active Ingredient And Polyvinylpyrrolidon As Excipient
US14/038,666 Abandoned US20140031396A1 (en) 2002-02-20 2013-09-26 Oral Dosage Form Containing A Pde 4 Inhibitor As An Active Ingredient And Polyvinylpyrrolidon As Excipient
US14/731,964 Abandoned US20150290178A1 (en) 2002-02-20 2015-06-05 Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient
US14/735,015 Abandoned US20150290180A1 (en) 2002-02-20 2015-06-09 Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient
US14/735,004 Abandoned US20150290179A1 (en) 2002-02-20 2015-06-09 Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient

Family Applications Before (8)

Application Number Title Priority Date Filing Date
US10/505,138 Expired - Lifetime US7951397B2 (en) 2002-02-20 2003-02-19 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US11/501,836 Abandoned US20060269600A1 (en) 2002-02-20 2006-08-10 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US12/876,996 Abandoned US20110060016A1 (en) 2002-02-20 2010-09-07 Oral dosage form containing a pde 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US13/008,842 Expired - Lifetime US8431154B2 (en) 2002-02-20 2011-01-18 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidone as excipient
US13/739,457 Abandoned US20130131123A1 (en) 2002-02-20 2013-01-11 Oral Dosage Form Containing A Pde 4 Inhibitor As An Active Ingredient And Polyvinylpyrrolidon As Excipient
US13/874,065 Expired - Lifetime US9468598B2 (en) 2002-02-20 2013-04-30 Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US14/038,678 Abandoned US20140031397A1 (en) 2002-02-20 2013-09-26 Oral Dosage Form Containing A Pde 4 Inhibitor As An Active Ingredient And Polyvinylpyrrolidon As Excipient
US14/038,666 Abandoned US20140031396A1 (en) 2002-02-20 2013-09-26 Oral Dosage Form Containing A Pde 4 Inhibitor As An Active Ingredient And Polyvinylpyrrolidon As Excipient

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/735,015 Abandoned US20150290180A1 (en) 2002-02-20 2015-06-09 Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient
US14/735,004 Abandoned US20150290179A1 (en) 2002-02-20 2015-06-09 Oral Dosage Form Containing A PDE 4 Inhibitor as an Active Ingredient and Polyvinylpyrrolidon as Excipient

Country Status (34)

Country Link
US (11) US7951397B2 (en)
EP (2) EP2258394B1 (en)
JP (1) JP4163120B2 (en)
KR (2) KR101386843B1 (en)
CN (2) CN1635909B (en)
AR (1) AR038527A1 (en)
AT (1) ATE550038T1 (en)
AU (1) AU2003206924B2 (en)
BR (1) BR0307739A (en)
CA (1) CA2475923C (en)
CY (2) CY1112811T1 (en)
DK (2) DK2258394T3 (en)
EA (1) EA008219B1 (en)
EC (1) ECSP045238A (en)
ES (2) ES2424634T3 (en)
GE (1) GEP20074079B (en)
HK (1) HK1077752A1 (en)
IL (3) IL162843A0 (en)
IS (1) IS7410A (en)
MA (1) MA27100A1 (en)
ME (1) ME00566A (en)
MX (1) MXPA04005759A (en)
MY (1) MY140561A (en)
NO (1) NO332844B1 (en)
NZ (1) NZ535197A (en)
PE (1) PE20030823A1 (en)
PL (1) PL226401B1 (en)
PT (2) PT1478399E (en)
RS (1) RS52548B (en)
SI (2) SI2258394T1 (en)
TN (1) TNSN04124A1 (en)
TW (1) TWI363636B (en)
UA (1) UA84266C2 (en)
WO (1) WO2003070279A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9468598B2 (en) 2002-02-20 2016-10-18 Astrazeneca Ab Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient

Families Citing this family (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1606261B1 (en) 2003-03-10 2009-11-04 Nycomed GmbH Novel process for the preparation of roflumilast
CL2004000983A1 (en) * 2003-05-08 2005-03-04 Altana Pharma Ag ORAL PHARMACEUTICAL COMPOSITION IN THE FORM OF A TABLET THAT INCLUDES DIHYDRATED MAGNETIC PANTOPRAZOL, WHERE THE TABLET FORM IS COMPOSED BY A NUCLEUS, A MIDDLE COAT AND AN OUTER LAYER; AND USE OF PHARMACEUTICAL COMPOSITION IN ULCERAS AND
EP1628682B1 (en) 2003-05-22 2013-09-25 Takeda GmbH Composition comprising a pde4 inhibitor and a pde5 inhibitor for the treatment of copd
DE10350528A1 (en) * 2003-10-29 2005-06-09 Boehringer Ingelheim Pharma Gmbh & Co. Kg Drug formulation containing an LTB4 antagonist, as well as processes for their preparation and their use
ES2245891B1 (en) * 2004-07-09 2006-11-16 Clinmet S.L. "METHOD FOR OBTAINING A UNIVERSAL EXCIPIENT FOR ORAL ADMINISTRATION OF PHARMACEUTICAL ACTIVE PRINCIPLES AND COMPOSITIONS OF EXCIPIENTS RESULTING FROM THE METHOD".
DE102004046235A1 (en) * 2004-09-22 2006-03-30 Altana Pharma Ag drug preparation
JP5383183B2 (en) * 2005-03-16 2014-01-08 タケダ ゲゼルシャフト ミット ベシュレンクテル ハフツング A tasted dosage form containing roflumilast
SI2366393T1 (en) * 2005-04-19 2013-12-31 Takeda Gmbh Roflumilast for the treatment of pulmonary hypertension
MX2008016123A (en) 2006-07-05 2009-01-15 Nycomed Gmbh Combination of hmg-coa reductase inhibitors with phosphodiesterase 4 inhibitors for the treatment of inflammatory pulmonary diseases.
US20110160213A1 (en) * 2007-02-01 2011-06-30 Glenmark Pharmaceuticals, S.A. Pharmaceutical compositions for the treatment of inflammatory and allergic disorders
BRPI0809205B8 (en) * 2007-03-29 2021-05-25 Daiichi Sankyo Co Ltd pharmaceutical composition
EP2140861A1 (en) * 2008-06-30 2010-01-06 Abbott GmbH & Co. KG Pharmaceutical dosage form comprising polymeric carrier composition
EP2186521A1 (en) * 2008-11-14 2010-05-19 Mergemeier Steffen Compositons for the treatment and prevention of diseases involving bacterial, viral and fungal pathogens and fragments thereof with polyvinylpyrrolidone and/or polyvinylpolypyrrolidone as therapeutically active compound
KR101792299B1 (en) 2010-03-19 2017-10-31 다이이찌 산쿄 가부시키가이샤 Method for improving dissolution of anticoagulant agent
US20130299501A1 (en) 2010-10-22 2013-11-14 Daewoo Shipbuilding & Marine Engineering Co., Ltd. Storage container for liquefied natural gas
HUE031652T2 (en) 2011-02-07 2017-07-28 Scipharm Sarl Novel composition for the treatment of cystic fibrosis
KR101940840B1 (en) 2011-08-10 2019-01-21 다이이찌 산쿄 가부시키가이샤 Pharmaceutical composition containing diamine derivative
CN102274222B (en) * 2011-08-18 2013-04-10 天津市汉康医药生物技术有限公司 High-bioavailability roflumilast medicinal composition and preparation method thereof
WO2013030789A1 (en) 2011-08-30 2013-03-07 Ranbaxy Laboratories Limited Pharmaceutical oral solid dosage form containing a poorly water soluble pde - iv inhibitor
SI2797581T1 (en) * 2011-12-27 2020-08-31 Amgen (Europe) GmbH Formulations of (+)-2-(1-(3-ethoxy-4-methoxy-phenyl)-2-methanesulfonyl-ethyl)-4-acetyl aminoisoindoline-1,3-dione
CN103446070B (en) * 2012-05-31 2016-06-22 天津康鸿医药科技发展有限公司 A kind of roflumilast solid immediate release preparation and preparation method thereof
WO2014012954A1 (en) 2012-07-18 2014-01-23 Takeda Gmbh Treatment of partly controlled or uncontrolled severe asthma
CN103565806A (en) * 2012-07-31 2014-02-12 江苏柯菲平医药有限公司 Roflumilast oral preparation and preparation method thereof
DK2892540T3 (en) * 2012-09-06 2018-11-19 Mcpharma Biotech Inc TREATMENT OF DIARRES AND DRAWN DIARRES WITH RESISTANT POTATO STARCH
CN102871976A (en) * 2012-09-29 2013-01-16 华润赛科药业有限责任公司 Tablet containing roflumilast as active ingredients and preparation method of tablet
CN102949370B (en) * 2012-11-27 2018-05-04 贵州信邦制药股份有限公司 A kind of Roflumilast tablet and preparation method thereof and detection method
CN103127011B (en) * 2012-12-27 2014-11-26 深圳万乐药业有限公司 Roflumilast tablet and preparation method thereof
CN103330689A (en) * 2013-03-31 2013-10-02 北京万全德众医药生物技术有限公司 Orally disintegrating tablet comprising roflumilast
ES2888074T3 (en) 2013-08-16 2021-12-30 Univ Maastricht Treatment of cognitive impairment with PDE4 inhibitor
WO2015037017A2 (en) * 2013-09-13 2015-03-19 Hetero Research Foundation Pharmaceutical compositions of roflumilast and process for preparation thereof
WO2015132708A1 (en) * 2014-03-07 2015-09-11 Torrent Pharmaceuticals Limited Pharmaceutical composition of roflumilast
EP3174999A1 (en) 2014-07-29 2017-06-07 Fundació Hospital Universitari Vall d' Hebron - Institut de Recerca Differential diagnosis and therapy selection for rheumatoid arthritis and psoriatic arthritis
KR101878621B1 (en) 2014-10-24 2018-07-13 히사미쓰 세이야꾸 가부시키가이샤 Prodrug
WO2016120746A1 (en) * 2015-01-29 2016-08-04 Micro Labs Limited A pharmaceutical composition comprising roflumilast
CN106176639A (en) * 2015-04-30 2016-12-07 四川科伦药物研究院有限公司 A kind of method preparing Roflumilast tablet
RU2659206C2 (en) * 2016-04-14 2018-06-28 Общество с ограниченной ответственностью "Технофарм" Composition and method for industrial production of digoxin trituration (options)
CN106139161A (en) * 2016-08-12 2016-11-23 合肥久诺医药科技有限公司 A kind of roflumilast clathrate and solid preparation thereof
JP7019978B2 (en) * 2017-06-30 2022-02-16 富士電機株式会社 Equipment management system, equipment management method and program
US11534493B2 (en) 2017-09-22 2022-12-27 Arcutis Biotherapeutics, Inc. Pharmaceutical compositions of roflumilast in aqueous blends of water-miscible, pharmaceutically acceptable solvents
WO2019147824A1 (en) 2018-01-26 2019-08-01 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a pde4 inhibitor
WO2020106750A1 (en) 2018-11-19 2020-05-28 Progenity, Inc. Methods and devices for treating a disease with biotherapeutics
CN115666704A (en) 2019-12-13 2023-01-31 比奥拉治疗股份有限公司 Ingestible device for delivery of therapeutic agents to the gastrointestinal tract
CN111643470A (en) * 2020-04-30 2020-09-11 山东希尔康泰药业有限公司 Preparation process of roflumilast film-coated tablets
US11672761B2 (en) 2020-11-16 2023-06-13 Orcosa Inc. Rapidly infusing platform and compositions for therapeutic treatment in humans
US11504332B2 (en) * 2021-03-23 2022-11-22 Vk Research Associates Inc. Phosphodiesterase-4 inhibitor combinations, methods of making, and methods of use thereof
US20240108609A1 (en) 2022-09-15 2024-04-04 Arcutis Biotherapeutics, Inc. Pharmaceutical compositions of roflumilast and solvents capable of dissolving high amounts of the drug

Family Cites Families (218)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3065142A (en) * 1958-07-30 1962-11-20 Armour Pharma Gastric resistant medicinal preparation
US4006227A (en) * 1973-11-15 1977-02-01 Gallegos Alfred J Compositions and methods for fertility control
US4024240A (en) * 1975-07-18 1977-05-17 Eli Lilly And Company Antibiotic A-32390 compositions
US4343804A (en) * 1979-03-26 1982-08-10 A. H. Robins Company, Inc. 4-Amino-3-quinolinecarboxylic acids and esters-antisecretory anti-ulcer compounds
ZA81219B (en) 1980-01-23 1982-01-27 Schering Corp Imidazo (1,2-a) pyridines ,process for their preparation and pharmaceutical compositions containing them
NZ196700A (en) * 1980-04-18 1983-04-12 Smith & Nephew Ass Anti-inflammatory compositions containing 5-benzoyl-1-methylpyrrole-2-acetic acid derivatives
EP0068378B1 (en) * 1981-06-26 1986-03-05 Schering Corporation Novel imidazo(1,2-a)pyridines and pyrazines, processes for their preparation and pharmaceutical compositions containing them
US4464372A (en) * 1982-08-16 1984-08-07 Schering Corporation Imidazo[1,2-b]pyridazines
GB8305245D0 (en) * 1983-02-25 1983-03-30 Fujisawa Pharmaceutical Co Imidazo-heterocyclic compounds
DE3308239A1 (en) 1983-03-09 1984-09-13 Basf Ag, 6700 Ludwigshafen N-ACYL-ANTHRANILE ACID DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE FOR CONTROLLING UNWANTED PLANT GROWTH
GB8307865D0 (en) * 1983-03-22 1983-04-27 Fujisawa Pharmaceutical Co Benzimidazole derivatives
DK159431C (en) 1984-05-10 1991-03-18 Byk Gulden Lomberg Chem Fab 6-PHENYL-3 (2H) -PYRIDAZINONES, METHOD OF PREPARING THEREOF, PHARMACEUTICALS CONTAINING THESE AND USING THE COMPOUNDS FOR THE PREPARATION OF MEDICINAL PRODUCTS
GB8415540D0 (en) 1984-06-18 1984-07-25 Fujisawa Pharmaceutical Co Imidazoisoquinoline compounds
JPS61205208A (en) 1985-03-08 1986-09-11 Yamanouchi Pharmaceut Co Ltd Rapidly solubilizing preparation for hard-soluble medicines
US4725601A (en) 1985-06-04 1988-02-16 Fujisawa Pharmaceutical Co., Ltd. Certain imidazo[1,2-a]pyridines useful in the treatment of ulcers
JPS61205208U (en) 1985-06-12 1986-12-24
JPS625966A (en) * 1985-07-03 1987-01-12 Nippon Shinyaku Co Ltd Benzimidazole derivative
EP0228006A1 (en) * 1985-12-16 1987-07-08 Fujisawa Pharmaceutical Co., Ltd. Imidazopyridine compounds and processes for preparation thereof
US4753945A (en) * 1986-02-19 1988-06-28 Eye Research Institute Of Retina Foundation Stimulation of tear secretion with phosphodiesterase inhibitors
JPH0249720Y2 (en) 1986-05-08 1990-12-27
US5286494A (en) * 1986-07-02 1994-02-15 Schering Aktiengesellschaft Medicinal agents with sustained action
GB8621425D0 (en) 1986-09-05 1986-10-15 Smith Kline French Lab Compounds
US4791117A (en) 1986-09-22 1988-12-13 Ortho Pharmaceutical Corporation 2- or 3-aryl substituted imidazo[1,2-a]pyridines and their use as calcium channel blockers
US4833149A (en) * 1986-09-22 1989-05-23 Ortho Pharmaceutical Corporation 2- or 3-aryl substituted imidazo[1,2-a]pyridines
NZ221996A (en) 1986-10-07 1989-08-29 Yamanouchi Pharma Co Ltd Imidazo-pyridine derivatives and pharmaceutical compositions
EP0264883A3 (en) 1986-10-21 1990-04-04 Banyu Pharmaceutical Co., Ltd. Substituted pyridine derivatives
US4831041A (en) * 1986-11-26 1989-05-16 Fujisawa Pharmaceutical Co., Ltd. Imidazopyridine compounds and processes for preparation thereof
WO1989000570A1 (en) 1987-07-16 1989-01-26 Byk Gulden Lomberg Chemische Fabrik Gmbh New diazols
GB8717644D0 (en) 1987-07-24 1987-09-03 Smithkline Beckman Intercredit Compounds
GB8722488D0 (en) 1987-09-24 1987-10-28 Fujisawa Pharmaceutical Co Imidazopyridine compound
GB8804444D0 (en) * 1988-02-25 1988-03-23 Smithkline Beckman Intercredit Compounds
US4900775A (en) * 1988-02-29 1990-02-13 Gaf Chemicals Corporation Solubilization of complexes of water-insoluble organic compounds by aqueous solutions of polyvinylpyrrolidone
US5011843A (en) * 1988-05-31 1991-04-30 Iolab Corporation Treatment of glaucoma using phosphodiesterase inhibitors
EP0368158A1 (en) 1988-11-07 1990-05-16 Byk Gulden Lomberg Chemische Fabrik GmbH Imidazo pyridines
JPH02270873A (en) 1989-03-13 1990-11-05 Fujisawa Pharmaceut Co Ltd Imidazopyridine compound and production thereof
CA2011086A1 (en) 1989-03-17 1990-09-17 Karl-Heinz Geiss 2-alkyl-4-arylmethylaminoquinolines, the use thereof and drugs prepared therefrom
GB8908229D0 (en) 1989-04-12 1989-05-24 Smithkline Beckman Intercredit Compounds
ATE124401T1 (en) 1989-04-17 1995-07-15 Byk Gulden Lomberg Chem Fab NEW ARYLPYRIDAZINE, THEIR PREPARATION, USE AND MEDICINAL PRODUCTS CONTAINING THEM.
DE3917232A1 (en) 1989-05-26 1990-11-29 Basf Ag New N-aryl:methyl 4-amino-quinoline derivs - useful as gastrointestinal medicaments
DE3917233A1 (en) 1989-05-26 1990-11-29 Basf Ag 8-SUBSTITUTED 4- (HETEROCYCLYLMETHYLAMINO) -INCHINOLINES, THEIR USE AND DRUGS DERIVED THEREFROM
DE3943385A1 (en) 1989-12-27 1991-07-04 Schering Ag AGENT FOR TRANSDERMAL APPLICATION CONTAINING ROLIPRAM
FR2657257B1 (en) * 1990-01-19 1994-09-02 Rhone Poulenc Sante PROCESS FOR THE PREPARATION OF DRUGS IN THE FORM OF PEARLS.
ES2112272T3 (en) 1990-03-28 1998-04-01 Otsuka Pharma Co Ltd DERIVATIVE OF QUINOLINE, ANTI-ULCEROUS MEDICINE CONTAINING THIS DERIVATIVE AND PRODUCTION OF THIS DERIVATIVE.
JP2851117B2 (en) 1990-03-30 1999-01-27 日清製粉株式会社 Indole derivatives and anti-ulcer drugs containing them as active ingredients
JP2816227B2 (en) 1990-03-30 1998-10-27 日清製粉株式会社 Anti-ulcer drug
AU7754691A (en) 1990-04-27 1991-11-27 Byk Gulden Lomberg Chemische Fabrik Gmbh Novel pyridazines
GB9012592D0 (en) 1990-06-06 1990-07-25 Smithkline Beecham Intercredit Compounds
US5041442A (en) * 1990-07-31 1991-08-20 Syntex (U.S.A.) Inc. Pyrrolo(1,2-a)pyrazines as inhibitors of gastric acid secretion
WO1992006979A1 (en) 1990-10-15 1992-04-30 Byk Gulden Lomberg Chemische Fabrik Gmbh New diazines
ATE168101T1 (en) 1990-10-16 1998-07-15 Byk Gulden Lomberg Chem Fab ARYLPYRIDAZINONES
IE71647B1 (en) 1991-01-28 1997-02-26 Rhone Poulenc Rorer Ltd Benzamide derivatives
US5698711A (en) * 1991-01-28 1997-12-16 Rhone-Poulenc Rorer Limited Compounds containing phenyl linked to aryl or heteroaryl by an aliphatic- or heteroatom-containing linking group
US5935978A (en) 1991-01-28 1999-08-10 Rhone-Poulenc Rorer Limited Compounds containing phenyl linked to aryl or heteroaryl by an aliphatic- or heteroatom-containing linking group
CZ153293A3 (en) 1991-01-29 1993-12-15 Smithkline Beecham Intercredit Salts of 4-amino-acylquinoline derivative
SE9100920D0 (en) 1991-03-27 1991-03-27 Astra Ab NEW ACTIVE COMPOUNDS
US5449676A (en) 1991-04-26 1995-09-12 Byk Gulden Lomberg Chemische Fabrik Gmbh Pyridazines
US5320848A (en) * 1991-05-28 1994-06-14 Affinity Biotech, Inc. Chewable drug-delivery composition
JP3108483B2 (en) * 1991-09-30 2000-11-13 日清製粉株式会社 Indole derivatives and anti-ulcer drugs containing the same as active ingredients
JP3038064B2 (en) * 1991-10-07 2000-05-08 日清製粉株式会社 Indole derivatives and anti-ulcer drugs containing the same as active ingredients
US5534515A (en) * 1991-10-25 1996-07-09 Byk Gulden Lomberg Chemische Fabrik Gmbh Pyrrolopyridazines having gastrointestinal protective effects
US5262171A (en) * 1991-11-25 1993-11-16 Isp Investments Inc. Pharmaceutical tablet with PVP having enhanced drug dissolution rate
GB9126438D0 (en) 1991-12-12 1992-02-12 Smithkline Beecham Intercredit New quinoline derivatives
ATE157864T1 (en) 1991-12-18 1997-09-15 Warner Lambert Co METHOD FOR PRODUCING A SOLID DISPERSION
GB9200607D0 (en) 1992-01-13 1992-03-11 Ethical Pharma Ltd Pharmaceutical compositions containing nifedipine and process for the preparation thereof
GB9201694D0 (en) 1992-01-27 1992-03-11 Smithkline Beecham Intercredit Compounds
GB9201692D0 (en) 1992-01-27 1992-03-11 Smithkline Beecham Intercredit Compounds
GB9201693D0 (en) * 1992-01-27 1992-03-11 Smithkline Beecham Intercredit Compounds
WO1993015045A1 (en) 1992-01-29 1993-08-05 Smithkline Beecham Corporation N-(3-phenylpropyl)oxamic acid, oxamate, and oxamide derivatives
AU3588693A (en) 1992-01-29 1993-09-01 Smithkline Beecham Corporation N-benzyloxamic acid, oxamate, and oxamide derivatives and their use as TNF and PDE IV inhibitors
JP3523275B2 (en) 1992-03-26 2004-04-26 東光薬品工業株式会社 Patch
DE69309030T2 (en) 1992-06-15 1997-09-25 Celltech Therapeutics Ltd TRISUBSTITUTED PHENYL DERIVATIVES AS SELECTIVE PHOSPHODIESTERASE IV INHIBITORS
GB9212693D0 (en) * 1992-06-15 1992-07-29 Celltech Ltd Chemical compounds
HUT72656A (en) 1992-07-28 1996-05-28 Rhone Poulenc Rorer Ltd Inhibitors of c-amp phosphodiesterase and tnf, their preparation pharmaceuticals contg. them and their use
US5891904A (en) * 1992-09-14 1999-04-06 Wolf-Georg Forssmann Use of inhibitors of phosphodiesterase IV
JP3284622B2 (en) 1992-10-23 2002-05-20 ソニー株式会社 Disk unit
US5429824A (en) * 1992-12-15 1995-07-04 Eastman Kodak Company Use of tyloxapole as a nanoparticle stabilizer and dispersant
KR0144833B1 (en) 1992-12-28 1998-07-15 김태훈 Novel quinazoline derivatives and their preparation
IL108520A (en) * 1993-02-15 1997-09-30 Byk Gulden Lomberg Chem Fab 2, 3, 8-TRISUBSTITUTED IMIDAZO £1, 2-a| PYRIDINE DERIVATIVES, PROCESSES FOR THE PREPARATION THEREOF AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
WO1994024130A1 (en) 1993-04-22 1994-10-27 Byk Gulden Lomberg Chemische Fabrik Gmbh New pyridinium salts and their use for controlling helicobacter bacteria
DE4314963A1 (en) 1993-05-06 1994-11-10 Bayer Ag Substituted pyridines and 2-oxo-1,2-dihydropyridines
US5556863A (en) 1993-06-11 1996-09-17 Astra Aktiebolag Compound for gastric acid secretion inhibition
CA2165192C (en) 1993-07-02 2001-04-24 Hermann Amschler Fluoroalkoxy-substituted benzamides and their use as cyclic nucleotide phosphodiesterase inhibitors
CN1128955A (en) 1993-08-10 1996-08-14 藤泽药品工业株式会社 Percutaneously absorbable prepn.
JP3284686B2 (en) 1993-08-30 2002-05-20 株式会社明電舎 Brake torque control method of brake dynamometer system
UA48122C2 (en) * 1993-10-11 2002-08-15 Бік Гульден Ломберг Хеміше Фабрік Гмбх IMIDAZO[1,2-a]PYRIDINE ALKOXYALKYL CARBAMATES, A PROCESS OF THEIR PREPARING AND A DRUG ON THEIR BASIS
US5559110A (en) * 1994-03-09 1996-09-24 The Dupont Merck Pharmaceutical Company Pharmaceutical formulations of cyclic urea type compounds
SE9401197D0 (en) 1994-04-11 1994-04-11 Astra Ab Active compounds
ZA954599B (en) 1994-06-07 1996-01-26 Allergan Inc Stable gel formulation for topical treatment of skin conditions
HU220874B1 (en) * 1994-07-28 2002-06-29 Byk Gulden Lomberg Chem Fab Imidazopyridine-azolidinones, process for producing them, the use thereof and pharmaceutical compositions containing them
JP3031280B2 (en) 1994-08-19 2000-04-10 東陶機器株式会社 Toilet bowl cleaning equipment
GB9514473D0 (en) * 1995-07-14 1995-09-13 Glaxo Lab Sa Chemical compounds
JPH0959152A (en) 1995-08-18 1997-03-04 Teisan Seiyaku Kk Isosorbide dinitrate-containing plaster
SE512835C2 (en) 1996-01-08 2000-05-22 Astrazeneca Ab Dosage form containing a plurality of units all containing acid labile H + K + ATPase inhibitors
US5677302A (en) * 1996-02-26 1997-10-14 Apotex Inc. Thiadiazole compounds useful as proton pump inhibitors
US6114537A (en) * 1996-02-26 2000-09-05 Apotex Inc. Process for scavenging thiols
FR2746800B1 (en) * 1996-03-29 1998-06-05 Jouveinal Inst Rech DIAZEPINO-INDOLES PHOSPHODIESTERASE INHIBITORS 4
JP3031280U (en) 1996-05-15 1996-11-22 サン トップ コンピューター システムズ コーポレーション Linear CCD bar code reader
NZ332341A (en) 1996-05-20 2000-05-26 Darwin Discovery Ltd Quinoline carboxamides as TNF inhibitors and as PDE-IV inhibitors
US5972381A (en) * 1996-06-28 1999-10-26 Schering Corporation Solid solution of an antifungal agent with enhanced bioavailability
US5762953A (en) * 1996-08-22 1998-06-09 Theratech, Inc. Transdermal propentofylline compositions for the treatment of Alzheimers disease
KR100338610B1 (en) 1996-09-04 2002-05-27 디. 제이. 우드, 스피겔 알렌 제이 Indazole Derivatives and Their Use as Inhibitors of Phosphodiesterase (PDE) type IV and the Production of Tumor Necrosis Factor (TNF)
JP2001503766A (en) 1996-11-12 2001-03-21 フアーマシア・アンド・アツプジヨン・アー・ベー Compact member comprising a plurality of porous cellulose matrices (PCM), methods of making and using the same
KR100640025B1 (en) * 1997-02-17 2006-10-31 알타나 파마 아게 Compositions for the treatment of ards or irds containing 3-cyclopropylmethoxy-n-3,5-dichloro-4-pyridinyl-4-difluoromethoxybenzamide and lung surfactant
SE9700661D0 (en) * 1997-02-25 1997-02-25 Astra Ab New compounds
US6624181B1 (en) * 1997-02-28 2003-09-23 Altana Pharma Ag Synergistic combination
EP0971922B1 (en) 1997-03-24 2004-04-28 ALTANA Pharma AG Tetrahydropyrido compounds
FR2762841B1 (en) 1997-04-30 1999-07-02 Jouveinal Inst Rech DIAZEPINO-INDOLONES INHIBITING PHOSPHODIESTERASES IV
JP2001526703A (en) * 1997-05-28 2001-12-18 ビイク グルデン ロンベルク ヒエーミツシエ フアブリーク ゲゼルシヤフト ミツト ベシユレンクテル ハフツング Condensed dihydropyran
IT1294748B1 (en) * 1997-09-17 1999-04-12 Permatec Tech Ag FORMULATION FOR A TRANSDERMAL DEVICE
HUP0100043A2 (en) * 1997-12-08 2001-08-28 Byk Gulden Lomberg Chemische Fabrik Gmbh. Novel administration form comprising an acid-sensitive active compound
SE9801526D0 (en) 1998-04-29 1998-04-29 Astra Ab New compounds
DE69937343T2 (en) * 1998-05-07 2008-07-24 Corixa Corp., Seattle ADJUVANEAN COMPOSITION AND METHODS OF USE THEREOF
CA2319495A1 (en) 1998-06-08 1999-12-16 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
CA2338753C (en) 1998-07-24 2006-11-21 Jago Research Ag Medicinal aerosol formulations
SE9802793D0 (en) * 1998-08-21 1998-08-21 Astra Ab New compounds
SE9802794D0 (en) 1998-08-21 1998-08-21 Astra Ab New compounds
NZ527232A (en) * 1998-08-26 2005-03-24 Smithkline Beecham Corp A composition contaning a PDE 4 inhibitor and a beta adrenergic bronchodilator useful for treating pulmonary diseases
DK1109810T3 (en) 1998-08-31 2004-10-18 Altana Pharma Ag Benzonaphthydine N-oxides with PDE3 and PDE4 inhibitory activity
IL142063A0 (en) * 1998-09-23 2002-03-10 Byk Gulden Lomberg Chem Fab Tetrahydropyridoether derivatives and pharmaceutical compositions containing the same
EP1118615A4 (en) 1998-09-29 2002-02-27 Fujisawa Pharmaceutical Co Novel salts of pyridopyrazine compound and crystals thereof
US6395746B1 (en) 1998-09-30 2002-05-28 Alcon Manufacturing, Ltd. Methods of treating ophthalmic, otic and nasal infections and attendant inflammation
US6743443B1 (en) * 1998-10-05 2004-06-01 Eisai Co., Ltd. Tablets immediately disintegrating in the oral cavity
US20020006418A1 (en) * 1998-10-13 2002-01-17 John Kung Composition to enhance permeation of topical skin agents
ATE386742T1 (en) 1998-11-03 2008-03-15 Nycomed Gmbh IMIDAZONAPHTHYRIDINES
DE19858331A1 (en) 1998-12-17 2000-06-21 Boehringer Ingelheim Pharma Tricyclic nitrogen heterocycles as PDE IV inhibitors
US6417190B1 (en) * 1998-12-17 2002-07-09 Boehringer Ingelheim Pharma Kg Tricyclic nitrogen heterocycles as PDE IV inhibitors
MY121142A (en) 1999-02-23 2005-12-30 Smithkline Beecham Corp Controlled release formulation for treating copd
DZ3019A1 (en) * 1999-03-01 2005-05-20 Smithkline Beecham Corp Use of a pde4 inhibitor in the preparation of a drug against copd.
US6270807B1 (en) * 1999-03-02 2001-08-07 L. Perrigo Company Taste-masked pharmaceutical composition
AU3284000A (en) 1999-03-10 2000-09-28 Byk Gulden Lomberg Chemische Fabrik Gmbh 3-cyclopropylmethoxy-4-difluoromethoxy-n-(3,5-dichloropyrid- 4-yl)benzamide in the treatment of multiple sclerosis
CN1126468C (en) 1999-04-08 2003-11-05 王力田 Black corn stalk beverage and preparing process thereof
US6503923B1 (en) * 1999-04-17 2003-01-07 Altana Pharma Ag Haloalkoxy imidazonaphthyridines
KR100328972B1 (en) * 1999-04-23 2002-03-20 서경배 Method for Producing Cosmetic or Skincare-pharmaceutical Composition Containing Nutritional Supplement and the Cosmetic or Skincare-pharmaceutical Composition Produced by the Above-method
PT1176960E (en) * 1999-05-04 2005-02-28 Altana Pharma Ag COMBINATION SINERGICA UNDERSTANDING ROFLUMILAST AND A PDE-3 INHIBITOR
WO2000074654A1 (en) * 1999-06-07 2000-12-14 Byk Gulden Lomberg Chemische Fabrik Gmbh Novel preparation and administration form comprising an acid-labile active compound
DE19925710C2 (en) 1999-06-07 2002-10-10 Byk Gulden Lomberg Chem Fab New preparation and dosage form containing an acid labile proton pump inhibitor
EP1200091B1 (en) 1999-08-03 2008-07-02 Lilly Icos LLC Pharmaceutical composition comprising tadalafil
DK1212089T3 (en) * 1999-08-21 2006-07-24 Altana Pharma Ag Synergistic combination of Roflumilast and Salmeterol
US6174878B1 (en) 1999-08-31 2001-01-16 Alcon Laboratories, Inc. Topical use of kappa opioid agonists to treat otic pain
US6375968B1 (en) * 1999-10-22 2002-04-23 3M Innovative Properties Company Encapsulated active material immobilized in hydrogel microbeads
EP1158416A4 (en) 1999-10-25 2007-12-05 Sony Corp Method for managing content data
US7076437B1 (en) 1999-10-29 2006-07-11 Victor Levy Process for consumer-directed diagnostic and health care information
KR20020050249A (en) * 1999-10-29 2002-06-26 스튜어트 알. 수터, 스티븐 베네티아너, 피터 존 기딩스 Method for administering a phosphodiesterase 4 inhibitor
US6985188B1 (en) 1999-11-30 2006-01-10 Thomson Licensing Video decoding and channel acquisition system
US6258833B1 (en) 1999-12-23 2001-07-10 Icos Corporation Cyclic AMP-specific phosphodiesterase inhibitors
US6348602B1 (en) * 1999-12-23 2002-02-19 Icos Corporation Cyclic AMP-specific phosphodiesterase inhibitors
AP2002002590A0 (en) 2000-01-31 2002-09-30 Pfizer Prod Inc Pyrimidinyl carboxamides useful as inhibitors of PDE4 isozymes.
US6379682B1 (en) * 2000-02-07 2002-04-30 Color Access, Inc. Clear water-in-oil emulsions
US20030207845A1 (en) * 2001-02-08 2003-11-06 Keating Elizabeth T. Method and compositions for treating an inflammatory disease
WO2001060358A1 (en) * 2000-02-16 2001-08-23 University Of Nebraska Medical Center Method and compositions for treating fibrotic diseases
KR20030019333A (en) * 2000-03-27 2003-03-06 쇼트 그라스 New cosmetic, personal care, cleaning agent, and nutritional supplement compositions comprising bioactive glass and methods of making and using the same
GB0011802D0 (en) * 2000-05-16 2000-07-05 Smithkline Beecham Method for enhancing cognitive function
JP2003534328A (en) * 2000-05-25 2003-11-18 メルク フロスト カナダ アンド カンパニー Fluoroalkoxy-substituted benzamide dichloropyridinyl N-oxide PDE4 inhibitors
EP1161950A1 (en) * 2000-06-09 2001-12-12 Warner-Lambert Company Use of diazepinoindoles for the treatment of chronic obstructive pulmonary disease
AR029984A1 (en) 2000-07-27 2003-07-23 Smithkline Beecham Corp METHOD FOR REDUCING ASSOCIATED EXCERBATIONS COPD AMBITO
US20040005995A1 (en) * 2001-07-26 2004-01-08 Edelson Jeffrey D Method for reducing exacerbations associated with copd
EP1199074A1 (en) 2000-09-15 2002-04-24 Warner-Lambert Company Pharmaceutical composition for preventing or treating a disease associated with an excess of il-12 production
US20020193393A1 (en) * 2001-03-07 2002-12-19 Michel Pairet Pharmaceutical compositions based on anticholinergics and PDE-IV inhibitors
AU2002227123A1 (en) 2000-11-07 2002-05-21 Merck And Co., Inc. Method of treatment with a combination of a pde4 inhibitor and a leukotriene antagonist
US20100310477A1 (en) * 2000-11-28 2010-12-09 Boehringer Ingelheim Pharma Gmbh & Co. Kg. Pharmaceutical compositions based on anticholingerics and additional active ingredients
CN100335040C (en) * 2000-12-07 2007-09-05 奥坦纳医药公司 Rapidly disintegrating tablet comprising acid-sensitive active ingredient
WO2002045692A1 (en) * 2000-12-07 2002-06-13 Altana Pharma Ag Pharmaceutical preparation in the form of a suspension comprising an acid-labile active ingredient
SI1341524T1 (en) * 2000-12-07 2012-02-29 Nycomed Gmbh Pharmaceutical preparation in the form of a paste comprising an acid-labile active ingredient
DE10061137B4 (en) 2000-12-07 2016-10-06 Takeda Gmbh New pharmaceutical preparation
UA80393C2 (en) * 2000-12-07 2007-09-25 Алтана Фарма Аг Pharmaceutical preparation comprising an pde inhibitor dispersed on a matrix
EP1366760B1 (en) 2001-02-15 2010-06-30 Mitsubishi Tanabe Pharma Corporation Tablets quickly disintegrated in oral cavity
CA2440069A1 (en) * 2001-03-05 2002-09-12 Ortho-Mcneil Pharmaceutical, Inc. Taste masked pharmaceutical compositions
FR2821745B1 (en) 2001-03-09 2004-07-02 Ethypharm Lab Prod Ethiques GRANULES AND GRANULES COATED WITH MASK TASTE
US6702796B2 (en) 2001-03-26 2004-03-09 The Procter & Gamble Company Absorbent interlabial device having an improved tab
UA77656C2 (en) * 2001-04-07 2007-01-15 Glaxo Group Ltd S-fluoromethyl ester of 6-alpha, 9-alpha-difluoro-17-alpha-[(2-furanylcarbonyl)oxy]-11-beta-hydroxy-16- alpha-methyl-3-oxoandrosta-1,4-dien-17-beta-carbothioacid as anti-inflammatory agent
US6872382B1 (en) * 2001-05-21 2005-03-29 Alcon, Inc. Use of selective PDE IV inhibitors to treat dry eye disorders
WO2002096423A2 (en) 2001-05-25 2002-12-05 Boehringer Ingelheim Pharma Gmbh & Co. Kg Combination of a pde4 inhibitor and tiotropium or derivate thereof for treating obstructive airways
CN1471538A (en) 2001-06-27 2004-01-28 前体生物药物股份有限公司 Peptide structures useful for competitive modulation of dipeptidyl peptidase IV catalysis
US20030092706A1 (en) 2001-11-09 2003-05-15 Johannes Barsig Combination
US6723348B2 (en) * 2001-11-16 2004-04-20 Ethypharm Orodispersible tablets containing fexofenadine
US6756392B2 (en) * 2002-02-11 2004-06-29 Pfizer Inc Nicotinamide derivatives useful as PDE4 inhibitors
PT1478358E (en) 2002-02-11 2013-09-11 Bayer Healthcare Llc Sorafenib tosylate for the treatment of diseases characterized by abnormal angiogenesis
MY140561A (en) 2002-02-20 2009-12-31 Nycomed Gmbh Dosage form containing pde 4 inhibitor as active ingredient
AU2003232148A1 (en) 2002-05-16 2003-12-02 Pharmacia Corporation A selective inos inhibitor and a pde inhibitor in combination for the treatment of respiratory diseases
US20030215498A1 (en) 2002-05-17 2003-11-20 Harland Ronald S. Rapidly disintegrating comressed tablets comprising biologically active compounds
DK2020243T3 (en) 2002-05-28 2018-11-19 Astrazeneca Ab Pharmaceutical composition which can be applied topically
US20030235631A1 (en) 2002-06-17 2003-12-25 Pfizer Inc. Combination treatment for depression and anxiety
US20040058950A1 (en) * 2002-07-09 2004-03-25 Boehringer Ingelheim Pharma Gmbh & Co. Kg Pharmaceutical compositions based on anticholinergics and PDE-IV inhibitors
PL213925B1 (en) * 2002-07-19 2013-05-31 Abbott Biotech Ltd Treatment of tnf ó related disorders
US20030018071A1 (en) * 2002-08-09 2003-01-23 Rennard Stephen I. Method and compositions for treating fibrotic diseases
ES2286491T3 (en) 2002-08-10 2007-12-01 Nycomed Gmbh PIPERIDINA-FTALAZONAS REPLACED WITH PIRROLIDINADIONA, AS INHIBITING AGENTS OF THE PDE4.
WO2004019944A1 (en) 2002-08-29 2004-03-11 Altana Pharma Ag 2-hydroxy-6-phenylphenanthridines as pde-4 inhibitors
CA2497176A1 (en) 2002-09-04 2004-03-18 Ranbaxy Laboratories Limited Taste masked dosage forms and processes for their preparation
US6822114B1 (en) * 2002-10-08 2004-11-23 Albemarle Corporation Process for production of fluoroalkoxy-substituted benzamides and their intermediates
AU2003288169B8 (en) * 2002-11-27 2010-08-26 Astrazeneca Ab Synergistic combination comprising roflumilast and (R,R) -formoterol
DE60321953D1 (en) * 2002-11-27 2008-08-14 Nycomed Gmbh NEW SYNERGISTIC COMPOSITION CONTAINING ROFLUMILAST AND FORMOTEROL
EP1581197A1 (en) * 2002-12-11 2005-10-05 Ranbaxy Laboratories, Limited Coating composition for taste masking coating and methods for their application and use
ES2334028T3 (en) 2003-01-30 2010-03-04 Ethypharm FLAVOR COVERED PARTICLES OF FLAVOR, PROCEDURE FOR THE PREPARATION OF THE SAME AND SPONSIBLE TABLETS THAT CONTAIN SUCH COVERED PARTICLES.
EP1606261B1 (en) 2003-03-10 2009-11-04 Nycomed GmbH Novel process for the preparation of roflumilast
DK1610787T3 (en) * 2003-03-28 2008-06-02 Nycomed Gmbh Synergistic combination comprising roflumilast and an anticholinergic agent selected from tiotropium salts for the treatment of respiratory diseases
EP1628682B1 (en) 2003-05-22 2013-09-25 Takeda GmbH Composition comprising a pde4 inhibitor and a pde5 inhibitor for the treatment of copd
US20050026883A1 (en) 2003-07-31 2005-02-03 Robinson Cynthia B. Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a PDE-4 inhibitor for treatment of asthma or chronic obstructive pulmonary disease
EP1656117A1 (en) * 2003-08-11 2006-05-17 Merck Frosst Canada Ltd. Flavored taste-masked pharmaceutical formulation made using a one-step coating process
EP1673064A1 (en) 2003-08-28 2006-06-28 Sandoz AG Pharmaceutical composition comprising anticonvulsant with taste mask coating
WO2005026095A1 (en) 2003-09-12 2005-03-24 Ranbaxy Laboratories Limited Process for the preparation of roflumilast
WO2005034871A2 (en) 2003-10-09 2005-04-21 Inverseon, Inc. Methods for treating diseases and conditions with inverse agonists
EP1691797A2 (en) 2003-10-21 2006-08-23 Pharmacia Corporation Combination of cyclooxygenase-2 inhibitor and phosphodiesterase 4 inhibitor and method
WO2005058892A1 (en) 2003-12-19 2005-06-30 Glaxo Group Limited Pyrazolo [3,4-b] pyridine compounds, and their use as phosphodiesterase inhibitors
JP2007536350A (en) * 2004-05-10 2007-12-13 ニコメッド ゲゼルシャフト ミット ベシュレンクテル ハフツング Use of roflumilast for the prevention or treatment of emphysema
US7562356B2 (en) * 2004-09-22 2009-07-14 Hewlett-Packard Development Company, L.P. Automatically resolving patch dependencies for a path bundle
DE102004046235A1 (en) 2004-09-22 2006-03-30 Altana Pharma Ag drug preparation
US7884059B2 (en) * 2004-10-20 2011-02-08 Afton Chemical Corporation Oil-soluble molybdenum derivatives derived from hydroxyethyl-substituted Mannich bases
US20060105038A1 (en) * 2004-11-12 2006-05-18 Eurand Pharmaceuticals Limited Taste-masked pharmaceutical compositions prepared by coacervation
WO2006094640A2 (en) * 2005-03-04 2006-09-14 F.Hoffmann-La Roche Ag Roflumilast and integrin inhibitor combination and method of treatment
JP5383183B2 (en) * 2005-03-16 2014-01-08 タケダ ゲゼルシャフト ミット ベシュレンクテル ハフツング A tasted dosage form containing roflumilast
US20060293343A1 (en) 2005-05-18 2006-12-28 Asahi Kasei Pharma Corporation Pyrimidine derivatives
JP5280357B2 (en) 2006-07-07 2013-09-04 スティーブン・ピー・ガベック Bicyclic heteroaryl inhibitors of PDE4
USD580547S1 (en) * 2007-07-31 2008-11-11 Nycomed Gmbh Tablet
JP5271070B2 (en) 2008-12-24 2013-08-21 エスアイアイ・プリンテック株式会社 Head chip, liquid ejecting head, and liquid ejecting apparatus
US20120052122A1 (en) 2010-08-26 2012-03-01 Nycomed Gmbh Treatment Of Chronic Obstructive Pulmonary Disease With Phosphodiesterase-4 Inhibitor
AU2012253834B2 (en) 2011-05-06 2016-06-16 Glaxosmithkline Consumer Healthcare (Uk) Ip Limited Sustained release paracetamol formulations
AP2014007875A0 (en) 2012-03-05 2014-08-31 Gilead Calistoga Llc Polymorphic forms of (S)-2-(1-(9H-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3H)-one
US8921307B2 (en) 2012-11-20 2014-12-30 Novartis Ag Synthetic linear apelin mimetics for the treatment of heart failure

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9468598B2 (en) 2002-02-20 2016-10-18 Astrazeneca Ab Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient

Also Published As

Publication number Publication date
NZ535197A (en) 2006-11-30
US20130345265A1 (en) 2013-12-26
GEP20074079B (en) 2007-03-26
SI1478399T1 (en) 2012-07-31
RS52548B (en) 2013-04-30
EP1478399A1 (en) 2004-11-24
PL226401B1 (en) 2017-07-31
TW200303768A (en) 2003-09-16
TWI363636B (en) 2012-05-11
HK1077752A1 (en) 2006-02-24
US20130131123A1 (en) 2013-05-23
PT1478399E (en) 2012-05-30
ATE550038T1 (en) 2012-04-15
EP2258394A1 (en) 2010-12-08
AU2003206924B2 (en) 2008-11-13
EA008219B1 (en) 2007-04-27
US20060269600A1 (en) 2006-11-30
CN1635909B (en) 2012-09-05
DK1478399T3 (en) 2012-06-25
PL370595A1 (en) 2005-05-30
KR20100103729A (en) 2010-09-27
US7951397B2 (en) 2011-05-31
ES2424634T3 (en) 2013-10-07
US8431154B2 (en) 2013-04-30
US20150290179A1 (en) 2015-10-15
KR101386843B1 (en) 2014-04-17
RS73604A (en) 2007-02-05
TNSN04124A1 (en) 2007-03-12
NO332844B1 (en) 2013-01-21
EP1478399B1 (en) 2012-03-21
IS7410A (en) 2004-08-18
CY1112811T1 (en) 2016-02-10
WO2003070279A1 (en) 2003-08-28
SI2258394T1 (en) 2013-09-30
EA200401019A1 (en) 2005-04-28
ME00566B (en) 2011-12-20
US20150290180A1 (en) 2015-10-15
KR101253033B1 (en) 2013-04-11
JP4163120B2 (en) 2008-10-08
CA2475923A1 (en) 2003-08-28
JP2005517724A (en) 2005-06-16
UA84266C2 (en) 2008-10-10
US20110251244A1 (en) 2011-10-13
CN102764242B (en) 2016-09-07
CN1635909A (en) 2005-07-06
NO20043904L (en) 2004-11-11
US20110060016A1 (en) 2011-03-10
CA2475923C (en) 2013-06-18
IL237196A0 (en) 2015-03-31
MA27100A1 (en) 2004-12-20
US20140031396A1 (en) 2014-01-30
MXPA04005759A (en) 2004-09-10
AR038527A1 (en) 2005-01-19
US9468598B2 (en) 2016-10-18
PE20030823A1 (en) 2003-10-11
DK2258394T3 (en) 2013-08-19
ME00566A (en) 2011-12-20
EP2258394B1 (en) 2013-05-15
KR20040084926A (en) 2004-10-06
IL162843A0 (en) 2005-11-20
CN102764242A (en) 2012-11-07
AU2003206924A1 (en) 2003-09-09
US20140031397A1 (en) 2014-01-30
ES2384378T3 (en) 2012-07-04
ECSP045238A (en) 2004-09-28
US20050159492A1 (en) 2005-07-21
CY1114210T1 (en) 2016-08-31
PT2258394E (en) 2013-07-23
MY140561A (en) 2009-12-31
IL162843A (en) 2015-03-31
BR0307739A (en) 2005-01-25

Similar Documents

Publication Publication Date Title
US9468598B2 (en) Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
JP5383183B2 (en) A tasted dosage form containing roflumilast
ZA200406644B (en) Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyroolidone as excipient
EP3843702B1 (en) Immediate release fixed-dose combination of memantine and donepezil
DE10207160A1 (en) Dosage form useful for treating diseases e.g. psoriasis, allergic contact eczema, atopic eczema, sunburn and pruritis comprises phosphodiesterase inhibitor and polyvinylpyrrolidone

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: ASTRAZENECA AB, SWEDEN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TAKEDA GMBH;REEL/FRAME:041328/0362

Effective date: 20160429