US20080299203A1 - Solid Pharmaceutical Dosage Formulation - Google Patents

Solid Pharmaceutical Dosage Formulation Download PDF

Info

Publication number
US20080299203A1
US20080299203A1 US12/190,252 US19025208A US2008299203A1 US 20080299203 A1 US20080299203 A1 US 20080299203A1 US 19025208 A US19025208 A US 19025208A US 2008299203 A1 US2008299203 A1 US 2008299203A1
Authority
US
United States
Prior art keywords
dosage form
lopinavir
study population
auc
ritonavir
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/190,252
Inventor
Joerg Rosenberg
Ulrich Reinhold
Bernd Liepold
Gunther Berndl
Jorg Breitenbach
Laman L. Alani
Soumojeet Ghosh
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.)
AbbVie Inc
Original Assignee
Abbott Laboratories
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=36678380&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080299203(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US10/925,442 external-priority patent/US8025899B2/en
Application filed by Abbott Laboratories filed Critical Abbott Laboratories
Priority to US12/190,252 priority Critical patent/US20080299203A1/en
Assigned to ABBOTT LABORATORIES reassignment ABBOTT LABORATORIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BERNDL, GUNTHER, BREITENBACH, JORG, LIEPOLD, BERND, ROSENBERG, JOERG, REINHOLD, ULRICH, ALANI, LAMAN L., GHOSH, SOUMOJEET
Publication of US20080299203A1 publication Critical patent/US20080299203A1/en
Assigned to ABBVIE INC. reassignment ABBVIE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABBOTT LABORATORIES
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
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1682Processes
    • A61K9/1694Processes resulting in granules or microspheres of the matrix type containing more than 5% of excipient
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • 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
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • 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
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • A61K9/204Polyesters, e.g. poly(lactide-co-glycolide)
    • 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
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • 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
    • A61K9/2095Tabletting processes; Dosage units made by direct compression of powders or specially processed granules, by eliminating solvents, by melt-extrusion, by injection molding, by 3D printing
    • 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
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/286Polysaccharides, e.g. gums; Cyclodextrin
    • A61K9/2866Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1617Organic 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
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1635Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates

Definitions

  • the present invention relates to a pharmaceutical dosage formulation, and more particularly, relates to a pharmaceutical dosage formulation comprising an HIV protease inhibitor.
  • PIs HTV protease inhibitors
  • NRTIs nucleoside/nucleotide reverse transcriptase inhibitors
  • NRTIs non-nucleoside reverse transcriptase inhibitors
  • PIs are poorly soluble and are very difficult to formulate. Originally, PIs were provided as liquid formulations in which the PI component was dissolved. Currently the most widely used PI dosage forms are gelatin capsules containing a fill solution in which the active ingredient is dissolved. The fill solutions required to dissolve the PI often contain excipients that cause discomfort or irritate the gastrointestinal system. Furthermore, only a limited amount of the PI can be dissolved in these dosage forms which therefore limits the amount of the PI loaded in each gelatin capsule.
  • a patient In order to obtain the necessary dose of an individual PI, a patient must take several gelatin capsules at any given dosing period, which is repeated several times in a day.
  • therapy for HIV patients includes multiple medications that commonly include a PI.
  • these patients often times require additional medications such as antibiotics and lipid lowering agents to control opportunistic infections and other diseases or conditions they may be afflicted with. Consequently, these patients can take an extraordinary number of medications in a variety of different dosage forms over the course of a given day.
  • a PI dosage form that reduces or eliminates gastrointestinal adverse events. It is also desirable to have such a dosage form that can be loaded with more active ingredient to reduce the pill burden on patients. Furthermore, it is desirable to have a dosage form that provides little variability in the blood levels of the PI within a subject and throughout a patient population. Another desirable feature would be a dosage form that provides similar blood levels of a PI regardless of whether or not a patient takes the medication following a meal. Yet another desirable feature would be a dosage form that does not have to be refrigerated to prevent degradation of the PI.
  • these consistent blood levels can be achieved with the formulation provided herein without regard to whether or not the patient has eaten or what type of meal was eaten. It is believed that this is the first time that an undissolved form of lopinavir has been formulated in a solid dosage form. Given the advantages attendant to such formulation, this represents the next breakthrough in HIV therapy which will help ease the complicated treatment regimens currently prescribed for HIV patients.
  • FIG. 1 shows Box (lower and upper quartiles) and Whiskers (5 th and 95 th percentiles) Plots for Lopinavir AUC Under Various Meal Conditions;
  • FIG. 2 shows Box (lower and upper quartiles) and Whiskers (5 th and 95 th percentiles) Plots for Lopinavir Cmax Under Various Meal Conditions.
  • AUC ⁇ is the area under the concentration time curve (AUC) extrapolated to infinity or the AUC to the last measured time point+(last measured concentration/elimination rate constant).
  • Cmax is defined as the observed maximum plasma concentration of an active ingredient.
  • “Pharmaceutically acceptable” as used herein means moieties or compounds that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio.
  • weight percent or “percent by weight” or “wt %” is defined as the weight of the individual component in the formulation divided by the total weight of all components of the formulation and then multiplied by 100. In some cases where a formulation has an outer coating, then weight of the coating can either be included or excluded in the total weight.
  • fasting/fasted state or condition generally is defined as 10 hours of abstinence from eating prior to dosing and 4 hours post-dosing, although those skilled in the art will recognize various other timings that would also qualify as a fasting or fasted state.
  • Moderate-fat meal condition is defined as receiving a meal that is approximately 500-600 KCal wherein 20-30% of the calories are from fat served approximately 30 minutes prior to dosing.
  • high-fat meal condition is defined as receiving a meal that is approximately 1000 Kcal wherein 50-55% of the calories are from fat served approximately 30 minutes prior to dosing and is used herein to refer to a “fed state” although those skilled in the art will recognize various meal conditions that would also qualify as a fed state.
  • solid solution is defined as a system in a solid state wherein the drug is molecularly dispersed throughout a matrix such that the system is chemically and physically uniform or homogenous throughout.
  • solid dispersion is defined as a system having small particles, typically of less than 400 ⁇ m in size, more typically less than 100 ⁇ m in size, and most typically less than 10 ⁇ m in size, of one phase dispersed in another phase (the carrier phase).
  • Suitable PI's for use in accordance with the present invention include but are not limited to (2S,3S,5S)-5-(N-(N-((N-methyl-N-((2-isopropyl-4-thiazolyl)-methyl)amino)carbonyl)-L-valinyl)amino-2-(N-(5-thiazolyl)methoxy-carbonyl)-amino)-amino-1,6-diphenyl-3hydroxyhexane (ritonavir); (2S,3S,5S)-2-(2,6-Dimethylphenoxyacetyl)amino-3-hydroxy-5-[2S-(1-tetrahydro-pyrimid-2-onyl)-3-methylbutanoyl]-amino-1,6-diphenylhexane (ABT-378; lopinavir); N-(2(R)-hydroxy-1(S)-indanyl)-2(R)-phenylmethyl
  • dosage forms of the present invention will comprise a therapeutically effective amount of at least one PI.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and other factors known to those of ordinary skill in the medical arts. For example, it is well within the skill of the art to start doses of the compound at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.
  • a pharmaceutical dosage form of the present invention will comprise from about 5 to about 30% by weight of the total dosage form, preferably from about 10 to about 25% by weight of the total dosage form, of an HIV protease inhibitor or a combination of HIV protease inhibitors.
  • the dosage form will contain between about 10 mg to about 1500 mg of a PI.
  • the dosage form will comprise lopinavir and ritonavir in a ratio of about 4:1 respectively.
  • the preferred dose of lopinavir and ritonavir is 400 mg and 100 mg respectively which can be divided evenly between multiple dosage forms, preferably two. It will be understood that multiple doses, typically two, can be given in a given day.
  • undissolved PI Pharmaceutical dosage forms provided herein generally will comprise an “undissolved” PI.
  • undissolved PI's as used herein means that the PI is in a solid form and not dissolved in a liquid carrier in its final dosage form.
  • Solid forms of a PI may include, for example, crystalline, micronized crystalline, crystalline nanoparticulates, amorphous, micronized amorphous, amorphous nanoparticulates, or preferably amorphous solid forms of a PI.
  • orally administered solid dosage forms include but are not limited to capsules, dragees, granules, pills, powders, and tablets.
  • Excipients commonly used to formulate such dosage forms include encapsulating materials or formulation additives such as absorption accelerators, antioxidants, binders, buffers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, and mixtures thereof.
  • absorption accelerators such as absorption accelerators, antioxidants, binders, buffers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, and mixtures thereof.
  • Excipients for orally administered compounds in solid dosage forms include agar, alginic acid, aluminum hydroxide, benzyl benzoate, 1,3-butylene glycol, castor oil, cellulose, cellulose acetate, cocoa butter, corn starch, corn oil, cottonseed oil, ethanol, ethyl acetate, ethyl carbonate, ethyl cellulose, ethyl laureate, ethyl oleate, gelatin, germ oil, glucose, glycerol, groundnut oil, isopropanol, isotonic saline, lactose, magnesium hydroxide, magnesium stearate, malt, olive oil, peanut oil, potassium phosphate salts, potato starch, propylene glycol, talc, tragacanth, water, safflower oil, sesame oil, sodium carboxymethyl cellulose, sodium lauryl sulfate, sodium phosphate salts, soybean oil, sucrose, t
  • a preferred dosage form will generally comprise at least one HIV protease inhibitor in a therapeutically effective amount, at least one pharmaceutically acceptable water-soluble polymer and at least one pharmaceutically acceptable surfactant.
  • a solid solution or solid dispersion can be formed into one of the above pharmaceutical dosage forms.
  • Such solutions or dispersions can be manufactured with suitable pharmaceutically acceptable water-soluble polymers including but not limited to water-soluble polymers having a Tg of at least about 50° C., preferably at least about 60° C., most preferred from about 80° C. to about 180° C. Methods for determining Tg values of the organic polymers are described in “Introduction to Physical Polymer Science”, 2nd Edition by L. H. Sperling, published by John Wiley & Sons, Inc., 1992.
  • Tg values for the homopolymers may be taken from “Polymer Handbook”, 2nd Edition by J. Brandrup and E. H. Immergut, Editors, published by John Wiley & Sons, Inc., 1975.
  • Water-soluble polymers having a Tg as defined above allow for the preparation of solid solutions or solid dispersions that are mechanically stable and, within ordinary temperature ranges, sufficiently temperature stable so that the solid solutions or solid dispersions may be used as dosage forms without further processing or be compacted to tablets with only a small amount of tableting aids.
  • the water-soluble polymer comprised in the preferred dosage form is a polymer that preferably has an apparent viscosity, when dissolved at 20° C. in an aqueous solution at 2% (w/v), of about 1 to about 5000 mPa ⁇ s, and more preferably of about 1 to about 700 mPa ⁇ s, and most preferred of about 5 to about 100 mPa ⁇ s.
  • Water-soluble polymers suitable for use in the preferred dosage form of the present invention include but are not limited to homopolymers and copolymers of N-vinyl lactams, especially homopolymers and copolymers of N-vinyl pyrrolidone, e.g.
  • polyvinylpyrrolidone (PVP), copolymers of N-vinyl pyrrolidone and vinyl acetate or vinyl propionate, cellulose esters and cellulose ethers, in particular methylcellulose and ethylcellulose, hydroxyalkylcelluloses, in particular hydroxypropylcellulose, hydroxyalkylalkylcelluloses, in particular hydroxypropylmethylcellulose, cellulose phthalates or succinates, in particular cellulose acetate phthalate and hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate or hydroxypropylmethylcellulose acetate succinate; high molecular polyalkylene oxides such as polyethylene oxide and polypropylene oxide and copolymers of ethylene oxide and propylene oxide, polyacrylates and polymethacrylates such as methacrylic acid/ethyl acrylate copolymers, methacrylic acid/methyl methacrylate copolymers, butyl methacrylate/2-dimethylaminoe
  • homopolymers or copolymers of N-vinyl pyrrolidone in particular a copolymer of N-vinyl pyrrolidone and vinyl acetate, are preferred.
  • a particularly preferred polymer is a copolymer of about 60% by weight of the copolymer, N-vinyl pyrrolidone and about 40% by weight of the copolymer, vinyl acetate.
  • the pharmaceutical dosage form comprises from about 50 to about 85% by weight of the total dosage form, preferably from about 60 to about 80% by weight of the total dosage form, of a water-soluble polymer or any combination of such polymers.
  • the term “pharmaceutically acceptable surfactant” as used herein refers to a pharmaceutically acceptable non-ionic surfactant.
  • the present invention provides a dosage form comprising at least one surfactant having an hydrophilic lipophilic balance (HLB) value of from about 4 to about 10, preferably from about 7 to about 9.
  • HLB hydrophilic lipophilic balance
  • the HLB system (Fiedler, H. B., Encylopedia of Excipients, 5 th ed., Aulendorf: ECV-Editio-Cantor-Verlag (2002)) attributes numeric values to surfactants, with lipophilic substances receiving lower HLB values and hydrophilic substances receiving higher HLB values.
  • Surfactants having an HLB value of from about 4 to about 10 suitable for use in the present invention include but are not limited to polyoxyethylene alkyl ethers, e.g. polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether; polyoxyethylene alkylaryl ethers, e.g. polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether, polyoxyethylene (3) octylphenyl ether; polyethylene glycol fatty acid esters, e.g.
  • sorbitan mono laurate Span® 20
  • sorbitan monooleate sorbitan monopalmitate
  • sorbitan stearate or mixtures of one or more thereof.
  • sorbitan mono fatty acid esters are preferred, with sorbitan mono laurate and sorbitan monopalmitate being particularly preferred.
  • a preferred dosage form of the present invention comprises from about 2 to about 20% by weight of the total dosage form, preferably from about 3 to about 15% by weight of the total dosage form, of the surfactant or combination of surfactants.
  • the preferred dosage form may comprise additional pharmaceutically acceptable surfactants such as polyoxyethylene castor oil derivates, e.g. polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate such as polyethylenglycol 40 hydrogenated castor oil (Cremophor® RH 40) or polyethylenglycol 60 hydrogenated castor oil (Cremophor® RH 60); or block copolymers of ethylene oxide and propylene oxide, also known as polyoxyethylene polyoxypropylene block copolymers or polyoxyethylene polypropyleneglycol, such as Poloxamer® 124, Poloxamer® 188, Poloxamer® 237, Poloxamer® 388, Poloxamer® 407 (BASF Wyandotte Corp.); or a mono fatty acid ester of polyoxyethylene castor oil derivates, e.g. polyoxyethylene
  • polyoxyethylene (20) sorbitan monooleate Tween® 80
  • polyoxyethylene (20) sorbitan monostearate Tween® 60
  • polyoxyethylene (20) sorbitan monopalmitate Tween® 40
  • polyoxyethylene (20) sorbitan monolaurate Tween® 20
  • the surfactant having an HLB value of from about 4 to about 10 generally accounts for at least about 50% by weight, preferably at least about 60% by weight, of the total amount of surfactant used.
  • the dosage form of the present invention can include additional excipients or additives such as, for example, flow regulators, lubricants, bulking agents (fillers) and disintegrants.
  • additional excipients may comprise from about 0 to about 15% by weight of the total dosage form.
  • the preferred solid dispersion or solid solution based dosage form of the present invention can be produced by preparing a solid solution or solid dispersion of the HIV protease inhibitor, or the combination of HIV protease inhibitors, in a matrix of a water-soluble polymer and a surfactant, and then shaping into the required tablet form.
  • the solid solution or solid dispersion product can be subdivided to granules, e.g. by grinding or milling, and the granules may subsequently be compacted to tablets.
  • melt-extrusion Various techniques exist for preparing solid solutions or solid dispersions including melt-extrusion, spray-drying and solution-evaporation with melt-extrusion being preferred.
  • the melt-extrusion process comprises the steps of preparing a homogeneous melt of the HIV protease inhibitor or the combination of HIV protease inhibitors, the water-soluble polymer and the surfactant, and cooling the melt until it solidifies.
  • Melting means a transition into a liquid or rubbery state in which it is possible for one component to get embedded homogeneously in the other. Typically, one component will melt and the other components will dissolve in the melt thus forming a solution. Melting usually involves heating above the softening point of the water-soluble polymer.
  • the preparation of the melt can take place in a variety of ways. The mixing of the components can take place before, during or after the formation of the melt.
  • the components can be mixed first and then melted or be simultaneously mixed and melted.
  • the melt is homogenized in order to disperse the active ingredients efficiently.
  • it may be convenient first to melt the water-soluble polymer and then to mix in and homogenize the active ingredients.
  • the melt temperature is in the range of about 70 to about 250° C., preferably from about 80 to about 180° C., most preferred from about 100 to about 140° C.
  • the active ingredients can be employed as such or as a solution or dispersion in a suitable solvent such as alcohols, aliphatic hydrocarbons or esters.
  • a suitable solvent such as alcohols, aliphatic hydrocarbons or esters.
  • Another solvent which can be used is liquid carbon dioxide. The solvent is removed, e.g. evaporated, upon preparation of the melt.
  • additives may be included in the melt, for example flow regulators such as colloidal silica; lubricants, fillers, disintegrants, plasticizers, stabilizers such as antioxidants, light stabilizers, radical scavengers, stabilizers against microbial attack.
  • extruders or kneaders include single screw extruders, intermeshing screw extruders or else multiscrew extruders, preferably twin screw extruders, which can be corotating or counterrotating and, optionally, be equipped with kneading disks.
  • working temperatures will also be determined by the kind of extruder or the kind of configuration within the extruder that is used. Part of the energy needed to melt, mix and dissolve the components in the extruder can be provided by heating elements. However, the friction and shearing of the material in the extruder may also provide a substantial amount of energy to the mixture and aid in the formation of a homogeneous melt of the components.
  • the melt ranges from pasty to viscous. Shaping of the extrudate conveniently is carried out by a calender with two counter-rotating rollers with mutually matching depressions on their surface. A broad range of tablet forms can be attained by using rollers with different forms of depressions. Alternatively, the extrudate is cut into pieces, either before (hot-cut) or after solidification (cold-cut).
  • the resulting solid solution or solid dispersion product is milled or ground to granules.
  • the granules may then be compacted.
  • Compacting means a process whereby a powder mass comprising the granules is densified under high pressure in order to obtain a compact with low porosity, e.g. a tablet. Compression of the powder mass is usually done in a tablet press, more specifically in a steel die between two moving punches.
  • a solid dosage form of the invention comprises a combination of more than one HIV protease inhibitor (or a combination of an HIV protease inhibitor with one or more other active ingredients) it is of course possible to separately prepare solid solution or solid dispersion products of the individual active ingredients and to blend the milled or ground products before compacting.
  • At least one additive selected from flow regulators, disintegrants, bulking agents (fillers) and lubricants is preferably used in compacting the granules.
  • Disintegrants promote a rapid disintegration of the compact in the stomach and keeps the granules which are liberated separate from one another.
  • Suitable disintegrants are crosslinked polymers such as crosslinked polyvinyl pyrrolidone and crosslinked sodium carboxymethylcellulose.
  • Suitable bulking agents also referred to as “fillers” are selected from lactose, calcium hydrogenphosphate, microcrystalline cellulose (Avicell®), silicates, in particular silicium dioxide, magnesium oxide, talc, potato or corn starch, isomalt, polyvinyl alcohol.
  • Suitable flow regulators are selected from highly dispersed silica (Aerosil®), and animal or vegetable fats or waxes.
  • a lubricant is preferably used in compacting the granules.
  • Suitable lubricants are selected from polyethylene glycol (e.g., having a Mw of from 1000 to 6000), magnesium and calcium stearates, sodium stearyl fumarate, and the like.
  • additives for example dyes such as azo dyes, organic or inorganic pigments such as aluminium oxide or titanium dioxide, or dyes of natural origin; stabilizers such as antioxidants, light stabilizers, radical scavengers, stabilizers against microbial attack.
  • dyes such as azo dyes, organic or inorganic pigments such as aluminium oxide or titanium dioxide, or dyes of natural origin
  • stabilizers such as antioxidants, light stabilizers, radical scavengers, stabilizers against microbial attack.
  • Dosage forms according to the invention may be provided as dosage forms consisting of several layers, for example laminated or multilayer tablets. They can be in open or closed form. “Closed dosage forms” are those in which one layer is completely surrounded by at least one other layer. Multilayer forms have the advantage that two active ingredients which are incompatible with one another can be processed, or that the release characteristics of the active ingredient(s) can be controlled. For example, it is possible to provide an initial dose by including an active ingredient in one of the outer layers, and a maintenance dose by including the active ingredient in the inner layer(s). Multilayer tablets types may be produced by compressing two or more layers of granules. Alternatively, multilayer dosage forms may be produced by a process known as “coextrusion”.
  • the process comprises preparation of at least two different melt compositions as explained above, and passing these molten compositions into a joint coextrusion die.
  • the shape of the coextrusion die depends on the required drug form. For example, dies with a plain die gap, called slot dies, and dies with an annular slit are suitable.
  • the dosage form In order to facilitate the intake of such a dosage form by a mammal, it is advantageous to give the dosage form an appropriate shape. Large tablets that can be swallowed comfortably are therefore preferably elongated rather than round in shape.
  • a film coat on the tablet further contributes to the ease with which it can be swallowed.
  • a film coat also improves taste and provides an elegant appearance.
  • the film-coat may be an enteric coat.
  • the film-coat usually includes a polymeric film-forming material such as hydroxypropyl methylcellulose, hydroxypropylcellulose, and acrylate or methacrylate copolymers.
  • the film-coat may further comprise a plasticizer, e.g. polyethylene glycol, a surfactant, e.g. a Tween® type, and optionally a pigment, e.g. titanium dioxide or iron oxides.
  • the film-coating may also comprise talc as anti-adhesive.
  • the film coat usually accounts for less than about 5% by weight of the dosage form.
  • Pharmacokinetic properties are generally understood to mean the manner and extent to which a drug is absorbed.
  • Common pK parameters include AUC (or “area under the curve”), which typically refers to the amount of drug that is measurable in blood or blood products of a person taking the drug over time.
  • AUC is variously referred to as a patients exposure to a drug.
  • Cmax is another pK term which refers to the maximum blood (or blood product) level over the course of a given regimen of a drug.
  • Drug regimens for which pK parameters are measured include “clinical studies.” Some clinical studies are performed in a finite population of healthy volunteer patients and are designed to determine the pK parameters of a drug (such as those mentioned above), and not to treat a patient. Each patient is thus called a member of the study population. While such clinical studies are carefully controlled and monitored, pK parameters can vary between clinical studies in large measure because different clinical studies are performed on different populations of patients. Although variances exist between clinical studies, those skilled in the art readily recognize that once a particular set of pK parameters is generally known, it is a matter of routine to formulate a drug to achieve a similar set of pK parameters.
  • the present invention provides a dosage form that can be taken without regard to whether a patient has eaten, sometimes referred to as “without regard to meals”, “can be taken with or without food”, “no food effect” or similar phrases.
  • the Cmax of the drug and AUC of the drug is similar in patients that have eaten (“fed state”) as compared to patients that have not eaten (“fasted state”).
  • the dosage form provided herein advantageously can be taken at any time regardless of whether or not patients have recently eaten.
  • a fasted state refers to the fact that a patient has not eaten for a given amount of time before taking a dose of medication, as well as not eating for a given amount of time after taking the dosage form. These time periods before and after dosing are a matter of choice, and can range between, for example 2 hours to 24 hours.
  • a fed state generally refers to the fact that a patient has eaten within a given time period of taking a particular medication. This time period is variable but may constitute, for example, a meal just before, during, or just after taking the medication, typically a meal is eaten within about an hour of dosing.
  • the quantity of food eaten that will qualify as a fed state is also variable but generally can comprise between about 500 to about 1500 Kcal of food.
  • the dosage forms provided herein will have substantially the same Cmax and AUC ⁇ values in patients in a fasted state as well as in a fed state, regardless of the dose given.
  • the mean of the individual patient ratios in a patient population for either the Cmax or AUC ⁇ in the fed state to fasted state will be in the range of about 0.7 to about 1.43; more preferably between about 0.75 and about 1.35; and most preferably between about 0.8 and about 1.25.
  • each patient is given a dose of drug in a fed state and, after an appropriate time period, a dose of the drug in a fasted state.
  • the AUC ⁇ and Cmax for both meal conditions are calculated for each patient.
  • the AUC ⁇ value for the fed state is then divided by the AUC ⁇ for the fasted state for each patient.
  • the individual patient values are then added together and then divided by the number of patients completing the study to arrive at a mean AUC ⁇ value for all patients completing the study.
  • the mean Cmax value is calculated in a similar manner. If the mean value of the fed to fasted ratio for all patients' Cmax or AUC ⁇ values in a given study is within 0.7 to 1.43, for example, then the dosage form provided to the patients would be considered to capable of administration without regard to whether or not the patient was in a fed or fasted state.
  • the dosage forms provided herein have less variability than other gelatin capsule based formulations containing a dissolved form of the drug or drugs. This lack of variability is evidenced in FIG. 1 and FIG. 2 which compare AUC ⁇ and Cmax data of an embodiment of the present invention and the data from a marketed gelatin capsule containing a dissolved PI. As shown by the Figures, the AUC ⁇ and Cmax data associated with an embodiment of the present invention shows less variation.
  • the graphs are a “box and whiskers” plot of the data comparing the two formulations wherein the bottom of any given “whisker” (labeled A in the first box and whisker plot of FIG.
  • any particular box and whisker plot of FIG. 1 is called the “5 th percentile”, meaning that 5% of the patients in the study fell below the designated AUC ⁇ or Cmax value for the particular whisker.
  • the top of the whisker (labeled D in first box and whisker plot of FIG. 1 ) represents the “95 th percentile”, meaning that 5% of the patients in the study had a AUC ⁇ or Cmax value above the value designated by the top of any particular whisker.
  • the bottom of any particular box (labeled B in first box and whisker plot of FIG. 1 ) represents the 25 th percentile and the top of any particular box (labeled C in first box and whisker plot of FIG. 1 ) represent the 75 th percentile.
  • the line running through any particular box is the 50 th percentile or median of any particular study population.
  • the data generally demonstrates that the variability associated with the embodiment of the present invention is less than that associated with the existing gelatin capsule formulation.
  • the difference between 95 th percentile and 5 th percentile of the gelatin capsule is greater than the difference between 95 th percentile and 5 th percentile of the embodiment of the present invention. This translates into the fact that a greater portion of the study population is getting a therapeutic benefit from the PI without experiencing adverse events do to overexposure of the drug.
  • the difference between the 95 th percentile of AUC ⁇ and 5 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein is less than about 180, more preferably less than about 175, even more preferably less than about 165, and most preferably less than about 160.
  • the difference between the 95 th percentile of AUC ⁇ and 5 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein is less than about 170, more preferably less than about 160, and most preferably less than about 150.
  • the difference between the 95 th percentile of AUC ⁇ and 5 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein is less than about 130, more preferably less than about 120, and most preferably less than about 110.
  • the difference between the 75 th percentile and 25 th percentile of the AUC data in FIG. 1 is also very important in demonstrating the lack of variability in dosage forms of the present invention. It is generally preferred that the difference between the 75 th percentile of AUC ⁇ and 25 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein (regardless of whether the population is fed or fasted) is less than about 60, more preferably less than about 55, even more preferably less than about 50.
  • the difference between the 75 th percentile of AUC ⁇ and 25 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein is less than about 65, more preferably less than about 60, and most preferably less than about 55.
  • the difference between the 75 th percentile of AUC ⁇ and 25 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein is less than about 60, more preferably less than about 50, and most preferably less than about 40.
  • the 5 th percentile to the 95 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein ranges between about 33 ⁇ g ⁇ h/mL and about 175 ⁇ g ⁇ h/mL; and the 25 th percentile to the 75 th percentile of AUC ⁇ of any given study population taking a dosage form as provided ranges between about 54 ⁇ g ⁇ h/mL and about 107 ⁇ g ⁇ h/mL.
  • the 5 th percentile to the 95 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein ranges between about 57 ⁇ g ⁇ h/mL and about 142 ⁇ g ⁇ h/mL; and the 25 th percentile to the 75 th percentile of AUC ⁇ of any given study population taking a dosage form as provided herein ranges between about 75 ⁇ g ⁇ h/mL and about 109 ⁇ g ⁇ g/mL. It is also preferred that the 5 th percentile of the AUC ⁇ of any given study population taking a dosage form as provided herein greater than about 30 ⁇ g ⁇ h/mL under fasted conditions, and greater than about 50 ⁇ g ⁇ g/mL under fed conditions. Finally with respect to AUC ⁇ , it is preferred that under fasting conditions the mean AUC ⁇ is between about 60 ⁇ g ⁇ h/mL and about 95 ⁇ g ⁇ h/mL. for any given study population taking a dosage form provided.
  • the Cmax parameters shown in FIG. 2 also demonstrates lack of variability associated with dosage forms provided herein.
  • difference between the 95 th percentile and the 5 th percentile is less than about 15, more preferably less than about 13, and most preferably less than about 11.
  • the 5 th percentile of Cmax of a given study population taking a dose of active ingredient formulated according to the present invention is greater that about 2.5 ⁇ g/mL.
  • difference between the 95 th percentile and the 5 th percentile is less than about 12, more preferably less than about 11.
  • the so called dosage form contained 400 mg of lopinavir and 100 mg of ritonovir evenly divided between two dosage forms.
  • Lopinavir was the only drug measured in these studies due to the fact that ritonavir is supplied not for its action as a PI but as a pharmacokinetic enhancer or booster (ritonavir inhibits the metabolism of lopinavir).
  • ritonavir inhibits the metabolism of lopinavir.
  • the present invention provides methods of reducing the side effects associated with HIV therapy, methods of increasing the bioavailability of a PI, methods of decreasing the pill burden of an HIV/AIDs patient, methods of decreasing the variability of blood levels of a PI in a patient taking PI therapy, and methods of providing a PI to a patient taking PI therapy. All of these methods comprise the step of providing a pharmaceutical dosage form comprising a therapeutically effective amount of an undissolved form of a PI to a patient.
  • the PI is (2S,3 S,5S)-2-(2,6-Dimethylphenoxyacetyl)amino-3-hydroxy-5-[2S-(1-tetrahydro-pyrimid-2-onyl)-3-methylbutanoyl]-amino-1,6-diphenylhexane (ABT-378; lopinavir).
  • the dosage form will comprise (2S,3S,5S)-5-(N-(N-((N-methyl-N-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)-L-valinyl)amino-2-(N-((5-thiazolyl)-methoxy-carbonyl)-amino)-amino-1,6-diphenyl-3hydroxyhexane (ritonavir).
  • Copovidone N-vinyl pyrrolidone/vinyl acetate copolymer 60:40 was mixed with ritonavir (4.17 parts by weight), lopinavir (16.67 parts by weight) and colloidal silica (1.0 part by weight). The powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.0 kg/h and a melt temperature of 133° C. The clear, fully transparent melt was fed to a calender with two counter-rotating rollers having mutually matching cavities on their surfaces. Tablets of 1080 mg were thus obtained. DSC and WAXS analysis did not reveal any evidence of crystalline drug material in the formulation.
  • the bioavailability of the formulation was assessed using beagle dogs (mixed sexes, weighing approximately 10 kg) which received a balanced diet with 27% fat and were permitted water ad libitum. Each dog received a 100 ⁇ g/kg subcutaneous dose of histamine approximately 30 minutes prior to dosing. A single dose corresponding to about 200 mg lopinavir, about 50 mg ritonavir, or about 200 mg lopinavir and about 50 mg ritonavir, respectively, was administered to each dog. The dose was followed by approximately 10 milliliters of water. Blood samples were obtained from each animal prior to dosing and 0.25, 0.5, 1.0, 1.5, 2, 3, 4, 6, 8, 10, 12 and 24 hours after drug administration.
  • the plasma was separated from the red cells by centrifugation and frozen ( ⁇ 30° C.) until analysis. Concentrations of HIV protease inhibitors were determined by reverse phase HPLC with low wavelength UV detection following liquid-liquid extraction of the plasma samples. The area under the curve (AUC) was calculated by the trapezoidal method over the time course of the study. Each dosage form was evaluated in a group containing 8 dogs; the values reported are averages for each group of dogs.
  • the dose-adjusted AUC in dogs was 0.52 ⁇ g.h/ml/100 mg for ritonavir and 4.54 ⁇ g.h/m/100 mg for lopinavir.
  • This example shows that solid solutions or solid dispersions of HIV protease inhibitors without added surfactant yield a very poor bioavailability.
  • the above composition is processed by melt extrusion.
  • the resulting extrudate can be used as such or milled and compressed into tablets, preferably by the use of suitable tabletting aids such as sodium stearyl fumarate, colloidal silica, lactose, isomalt, calcium silicate, and magnesium stearate, cellulose or calcium hydrogenphosphate.
  • Copovidone N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 78.17 parts by weight
  • ritonavir 4.16 parts by weight
  • lopinavir 16.67 parts by weight
  • colloidal silica 1.0 part by weight
  • the powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.0 kg/h and a melt temperature of 133° C.
  • the clear, fully transparent melt was fed to a calender with two counter-rotating rollers having mutually matching cavities on their surfaces. Tablets of 1080 mg were thus obtained.
  • DSC and WAXS analysis did not reveal any evidence of crystalline drug material in the formulation.
  • Copovidone N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 68.17 parts by weight
  • Cremophor RH40 polyoxyethyleneglycerol oxystearate; 10.00 parts by weight
  • the resulting granules were mixed with ritonavir (4.17 parts by weight), lopinavir (16.67 parts by weight) and colloidal silica (1.00 parts by weight).
  • the powdery mixture was then fed into a Leistritz Micro 18 twin-screw extruder at a rate of 2.3 kg/h and a melt temperature of 126° C. The extrudate was cut into pieces and allowed to solidify.
  • the extruded pieces were milled using a high impact universal mill.
  • the milled material (86.49 parts by weight) was blended in a bin blender with lactose monohydrate (6.00 parts by weight), crosslinked PVP (6.00 parts by weight), colloidal silica (1.00 part by weight) and magnesium stearate (0.51 parts by weight).
  • the powdery blend was compressed to tablets of 1378.0 mg on a Fette E 1 single punch tablet press.
  • the tablets were then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • the bioavailability of the formulation was assessed using beagle dogs as in Example 1.
  • the dose-adjusted AUC in dogs was 0.60 ⁇ g.h/ml/100 mg for ritonavir and 7.43 ⁇ g.h/ml/100 mg for lopinavir.
  • This example shows that inclusion of a surfactant into solid solutions or solid dispersions of HIV protease inhibitors improves the bioavailability attained.
  • Copovidone N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 853.8 parts by weight
  • Span 20 Sorbitan monolaurate; 83.9 parts by weight
  • the resulting granules were mixed with ritonavir (50 parts by weight), lopinavir (200 parts by weight) and colloidal silica (12 parts by weight).
  • the powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.1 kg/h and a melt temperature of 119° C.
  • the extrudate was fed to a calender with two counter-rotating rollers having mutually matching cavities on their surfaces. Tablets of 1120 mg were thus obtained.
  • the bioavailability of the formulation was assessed using beagle dogs as in Example 1.
  • the dose-adjusted AUC in dogs was 10.88 ⁇ g.h/ml/100 mg for ritonavir and 51.2 ⁇ g.h/ml/100 mg for lopinavir.
  • This example shows that inclusion of a surfactant having an HLB of 4 to 10 into solid solutions or solid dispersions of HIV protease inhibitors markedly improves the bioavailability attained.
  • Example 5 was repeated, however, the extrudate was cut into pieces and allowed to solidify.
  • the extruded pieces were milled to a particle size of about 250 ⁇ m, using a high impact universal mill.
  • the milled material was blended in a bin blender with sodium stearyl fumarate (12.3 parts by weight) and colloidal silica (8.0 parts by weight) for 20 min.
  • the powdery blend was compressed on a rotary tablet machine with 3 punches (6500 tablets/h).
  • the tablets were then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • Opadry available from Colorcon
  • the bioavailability of the formulation was assessed using beagle dogs as in Example 1.
  • the dose-adjusted AUC in dogs was 14.24 ⁇ g.h/ml/100 mg for ritonavir and 52.2 ⁇ g.h/ml/100 mg for lopinavir.
  • Copovidone N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 841.3 parts by weight
  • Cremophor RH40 polyoxyethyleneglycerol oxystearate; 36.2 parts by weight
  • Span 20 Sorbitan monolaurate; 60.2 parts by weight
  • the resulting granules were mixed with ritonavir (50 parts by weight), lopinavir (200 parts by weight) and colloidal silica (12 parts by weight).
  • the powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.1 kg/h and a melt temperature of 114° C.
  • the extrudate was fed to a calender with two counter-rotating rollers having mutually matching cavities on their surfaces. Tablets of 1120 mg were thus obtained.
  • the bioavailability of the formulation was assessed using beagle dogs as in Example 1.
  • the dose-adjusted AUC in dogs was 10.96 ⁇ g.h/ml/100 mg for ritonavir and 46.5 ⁇ g.h/ml/100 mg for lopinavir.
  • This example shows that a combination of a surfactant having an HLB of 4 to 10 and a further surfactant can successfully be used.
  • Example 7 was repeated, however, the extrudate was cut into pieces and allowed to solidify.
  • the extruded pieces were milled to a particle size of about 250 ⁇ M, using a high impact universal mill.
  • the milled material was blended in a bin blender with sodium stearylfumarate (13.9 parts by weight), colloidal silica (7.0 parts by weight), isomalt DC100 (159.4 parts by weight) and calcium silicate (7.0 parts by weight) for 20 min.
  • the blend was compressed and then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • the bioavailability of the formulation was assessed using beagle dogs as in Example 1.
  • the dose-adjusted AUC in dogs was 10.38 ⁇ g.h/ml/100 mg for ritonavir and 42.7 ⁇ g.h/mV/100 mg for lopinavir.
  • Copovidone N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 683.3 parts by weight
  • Span 40 sorbitan monopalmitate; 67.2 parts by weight
  • the resulting granules were mixed with lopinavir (200 parts by weight) and colloidal silica (9.6 parts by weight).
  • the powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.1 kg/h and a melt temperature of 119° C.
  • the extrudate was cut into pieces and allowed to solidify.
  • the extruded pieces were milled using a high impact universal mill.
  • the milled material was blended in a bin blender with sodium stearylfumarate (7.9 parts by weight), colloidal silica (11.3 parts by weight), isomalt DC100 (129.1 parts by weight) and sodium dodecyl sulfate (15.6 parts by weight).
  • the blend was compressed and then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • the bioavailability of the formulation was assessed using beagle dogs as in Example 1. Tablets corresponding to 200 mg lopinavir were coadministered to dogs together with 50 mg ritonavir. The dose-adjusted AUC of lopinavir was 38.8 ⁇ g.h/ml/100 mg.
  • Copovidone N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 151.5 parts by weight
  • Cremophor RH40 24 parts by weight
  • PEG 6000 12 parts by weight
  • the resulting granules were mixed with ritonavir (50 parts by weight) and colloidal silica (2.4 parts by weight).
  • the powdery mixture was then fed into a twin-screw extruder and was melt-extruded.
  • the extrudate was cut into pieces and allowed to solidify. The extruded pieces were milled using a high impact universal mill.
  • the milled material was blended in a bin blender with colloidal silica (1.4 parts by weight), isomalt DC 100 (31.9 parts by weight) and calcium silicate (4.2 parts by weight).
  • the blend was compressed and then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • the extruded material was milled, compressed with tableting excipients, and coated.
  • the formulation consisted of lopinavir (200 mg/tablet), ritonavir (50 mg/tablet), copovidone as the carrier polymer, and polyoxyl 40 hydrogenated castor oil as the surfactant.
  • lopinavir 200 mg/tablet
  • ritonavir 50 mg/tablet
  • copovidone as the carrier polymer
  • polyoxyl 40 hydrogenated castor oil as the surfactant.
  • outer phase excipients were added to the milled extrudate.
  • the surfactant was incorporated prior to extrusion by granulation with a portion of the polymer.
  • the tablet formulation was compressed from separately extruded lopinavir and ritonavir powder mixtures.
  • the surfactant was incorporated prior to extrusion by granulation with a portion of the polymer.
  • the formulation was prepared by milling the extrudate, mixing with tableting excipients and compressing into tablets. An aqueous, hydroxypropyl methylcellulose based film coating was applied to the compressed tablets to enhance pharmaceutical elegance. The surfactant was incorporated prior to extrusion by granulation with a portion of the polymer.
  • the formulation was prepared by milling the extrudate, mixing with tableting excipients and compressing into tablets. An aqueous, hydroxypropyl methylcellulose based film coating was applied to the compressed tablets to enhance pharmaceutical elegance. The surfactant was incorporated prior to extrusion by granulation with a portion of the polymer.
  • the formulation was extruded in the shape of a tablet without the additional processing steps of milling, compression and coating.
  • the formulation composition included ritonavir, lopinavir, copovidone, surfactant, and colloidal silicon dioxide with the two formulations differing in the type of surfactant used.
  • the extruded tablet formulation contained sorbitan monolaurate as the surfactant that was incorporated prior to extrusion by granulation with a portion of the polymer.
  • the formulation was extruded in the shape of a tablet without the additional processing steps of milling, compression and coating.
  • the formulation composition included ritonavir, lopinavir, copovidone, surfactant, and colloidal silicon dioxide with the two formulations differing in the type of surfactant used.
  • the extruded tablet formulation contained both polyoxyl 40 hydrogenated castor oil and sorbitan monolaurate as the surfactants. The surfactants were incorporated prior to extrusion by granulation with a portion of the polymer.
  • This dosage form was characterized by an excellent stability and, in particular, exhibit high resistance against recrystallization or decomposition of the active ingredient(s).
  • the dosage forms according to the present invention did not exhibit any sign of crystallinity (as evidenced by DSC or WAXS analysis) and contained at least about 98% of the initial active ingredient content (as evidenced by HPLC analysis).
  • Table 1 shows the mean % lopinavir releaseded in minutes for the formulations disclosed in Examples 9-10 and 12-16.
  • Table 2 shows the mean % ritonavir dissolved in minutes for the formulations disclosed in Examples 9-10 and 12-16.
  • the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 20% to about 30% of lopinavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount
  • said dosage form providing an in vitro dissolution profile wherein about 20% to about 30% of lopinavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 20% to about 30% of lopinavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount
  • said dosage form providing an in vitro dissolution profile wherein about 20% to about 30% of lopinavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 43% to about 63% of lopinavir is released from about 15 to about 30 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount
  • said dosage form providing an in vitro dissolution profile wherein about 43% to about 63% of lopinavir is released from about 15 to about 30 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 61.3% to about 81.7% of lopinavir is released from about 30 to about 45 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 61.3% to about 81.7% of lopinavir is released from about 30 to about 45 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 75.4% to about 93.2% of lopinavir is released from about 45 to about 60 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 75.4% to about 93.2% of lopinavir is released from about 45 to about 60 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 19.8% to about 34.4% of ritonavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount
  • said dosage form providing an in vitro dissolution profile wherein about 19.8% to about 34.4% of ritonavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 41.6% to about 76.5% of ritonavir is released from about 15 to about 30 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount
  • said dosage form providing an in vitro dissolution profile wherein about 41.6% to about 76.5% of ritonavir is released from about 15 to about 30 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 59.4% to about 91.1% of ritonavir is released from about 30 to about 45 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount
  • said dosage form providing an in vitro dissolution profile wherein about 59.4% to about 91.1% of ritonavir is released from about 30 to about 45 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 73.4% to about 95% of ritonavir is released from about 45 to about 60 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount
  • said dosage form providing an in vitro dissolution profile wherein about 73.4% to about 95% of ritonavir is released from about 45 to about 60 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C. wherein:
  • the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C. wherein:
  • the present invention provides for example, a pharmaceutical dosage form comprising ritonavir and lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C. wherein:
  • the mean values for lopinavir Cmax and AUC ⁇ under moderate-fat meal condition were obtained from the central values in a cross-study meta-analysis of bioequivalence as generally known by those having ordinary skill in the art.
  • the variance values for the distribution were obtained from the between-subject variability estimated for the dosage form of the present invention and the currently marketed Kaletra gelatin capsule using the SAS Procedure Mixed as generally known by those having ordinary skill in the art.
  • the probability distributions of lopinavir Cmax and AUC ⁇ under fasting and high-fat meal conditions were adjusted using the point estimates from Studies C and A described below for the dosage form of the present invention and currently marketed Kaletra gelatin capsule.
  • the variance for each of fasting and high-fat meal conditions were projected according to the magnitude of the variability relative to that of the moderate-fat meal conditions using data from studies A, B and C described below in more detail.
  • Study A was a single-dose (lopinavir/ritonavir 400/100 mg), five-period, randomized, open-label, pivotal bioavailability study in 63 healthy subjects. The first four periods were conducted according to a complete-crossover design. Subjects were equally randomized to four sequences of Regimens A, B, C and D for Periods 1 through 4. Five subjects from each sequence group who completed Periods 1 through 4 were randomly chosen to participate in Period 5 and received Regimen E. A washout interval of at least 7 days separated the doses of the five study periods. The five regimens were:
  • Study B was a single-dose (lopinavir/ritonavir 400/100 mg), non-fasting, moderate-fat, open-label, four-period, randomized, complete-crossover, pivotal bioavailability study in 48 healthy subjects. Subjects were randomly assigned in equal numbers to receive one of four sequences of Regimens A, B, C and D defined as follows:
  • the single doses were administered in the morning on Study Day 1 of each period following a moderate-fat breakfast.
  • a washout interval of 7 days separated the doses of the four study periods.
  • Study C was a Phase 1, single-dose, fasting and non-fasting, open-label, randomized, five-period, partial crossover, single-center study in 56 healthy subjects.
  • the currently marketed Kaletra liquid and gelatin capsule formulations were administered to provide a single dose of lopinavir/ritonavir 400/100 mg. Both formulations were given under fasting conditions and following moderate and high-fat meals.
  • the dosage form of the present invention provides a substantially lower variation in Cmax and AUC ⁇ from the 5 th to the 95 th percentiles for lopinavir when administered to a subject whether fed or fasted than the gelatin capsule formulation. That is, the dosage form of the present invention provides a smaller ⁇ Cmax and ⁇ AUC ⁇ from the 5 th to the 95 th percentiles for lopinavir than the Kaletra gelatin capsule formulation. This is shown both graphically in FIGS. 1 and 2 as well as numerically in Tables 3-5.
  • the dosage form of the present invention also provides a substantially lower variation in Cmax and AUC ⁇ from the 25 th to the 75 th percentiles for lopinavir when administered to a subject whether fed or fasted than the gelatin capsule formulation. That is, the dosage form of the present invention provides a smaller ⁇ Cmax and ⁇ AUC ⁇ from the 25 th to the 75 th percentiles for lopinavir than the Kaletra gelatin capsule formulation. This is shown both graphically in FIGS. 1 and 2 as well as numerically in Tables 3-5.
  • the Kaletra gelatin capsule formulation provides a ⁇ AUC ⁇ of 257.9 ⁇ g ⁇ h/mL from the 5 th to the 95 th percentile, and ⁇ Cmax of 20.21 ⁇ g/mL from the 5 th to the 95 th percentile.
  • the dosage form of the present invention provides a ⁇ AUC ⁇ of 141.15 ⁇ g ⁇ h/mL from the 5 th to the 95 th percentile, and ⁇ Cmax of 11.98 ⁇ g/mL from the 5 th to the 95 th percentile.
  • the Kaletra gelatin capsule formulation provides a ⁇ AUC ⁇ of 75.53 ⁇ g ⁇ h/mL and ⁇ Cmax of 6.36 ⁇ g/mL for 50% of the study subjects.
  • the dosage form of the present invention provides a ⁇ AUC ⁇ of 52.71 ⁇ g ⁇ h/mL and ⁇ Cmax of 4.5 ⁇ g/mL for 50% of the study subjects.
  • the dosage form of the present invention demonstrates no food effect.
  • the ratio “X” of AUC ⁇ fed to AUC ⁇ fasted for lopinavir is calculated using the formula below,
  • the calculation is performed for each individual member of a study population in a given trial.
  • the mean value is calculated by adding up the “X” values of every subject and then dividing the total by the number of subjects in the trial.
  • the “X” value is in the range of about 0.7 to about 1.43, it is determined that the dosage form has no food effect. That is, the dosage form will have substantially the same bioavailability whether it is administered on a full or empty stomach.
  • the calculation is performed for each individual member of a study population in a given trial.
  • the mean value is calculated by adding up the “Y” values of every subject and then dividing the total by the number of subjects in the trial.
  • the “Y” value is in the range of about 0.7 to about 1.43, it is determined that the dosage form has no food effect. That is, the dosage form will have substantially the same bioavailability whether it is administered on a full or empty stomach.
  • Table 6 below better illustrates how “X” and “Y” values are calculated from individual members of a study population totaling 20 subjects.
  • Table 6 shows the mean Cmax value is 1.04 and the mean AUC ⁇ value is 1.28. These values are both individually within the range of about 0.7 to about 1.43 and show that the dosage form of the present invention has no food effect.

Abstract

The present invention provides a pharmaceutical dosage formulation, and more particularly, a pharmaceutical dosage formulation comprising an HIV protease inhibitor.

Description

  • This application is a continuation of U.S. patent application Ser. No. 11/064,467 filed on Feb. 23, 2005, which is incorporated herein by reference in its entirety. This application is also a continuation-in-part of U.S. patent application Ser. No. 10/925,442 filed on Aug. 25, 2004, which claims priority to U.S. Provisional Patent Application Ser. No. 60/498,412 filed on Aug. 28, 2003.
  • FIELD OF THE INVENTION
  • The present invention relates to a pharmaceutical dosage formulation, and more particularly, relates to a pharmaceutical dosage formulation comprising an HIV protease inhibitor.
  • BACKGROUND OF THE INVENTION
  • Millions of people around the world are suffering from HIV/AIDS, and millions more are likely to become infected each year. Many medications are currently available for the treatment of HIV/AIDS including HTV protease inhibitors (PIs), nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs). Most current treatment regimens require a combination of at least three medications, most commonly two NRTIs and either a PI or a NNRTI.
  • PIs are poorly soluble and are very difficult to formulate. Originally, PIs were provided as liquid formulations in which the PI component was dissolved. Currently the most widely used PI dosage forms are gelatin capsules containing a fill solution in which the active ingredient is dissolved. The fill solutions required to dissolve the PI often contain excipients that cause discomfort or irritate the gastrointestinal system. Furthermore, only a limited amount of the PI can be dissolved in these dosage forms which therefore limits the amount of the PI loaded in each gelatin capsule.
  • In order to obtain the necessary dose of an individual PI, a patient must take several gelatin capsules at any given dosing period, which is repeated several times in a day. As mentioned above, therapy for HIV patients includes multiple medications that commonly include a PI. Moreover, these patients often times require additional medications such as antibiotics and lipid lowering agents to control opportunistic infections and other diseases or conditions they may be afflicted with. Consequently, these patients can take an extraordinary number of medications in a variety of different dosage forms over the course of a given day.
  • Such treatment regimens are further complicated by the fact that some of the dosage forms (including some PI's) require refrigerated storage conditions to prevent degradation of the active ingredients. For subjects residing in economically challenged or developing countries where refrigerators are not as common in households, such storage conditions represent a particularly challenging dilemma.
  • It has also been observed that upon administration of a PI from gelatin capsules there is variability in the blood levels of the active ingredient from subject to subject and even within the same subject. That is, some patients receiving treatment can have very high or very low blood levels of the PI. In turn, this can lead to unwanted adverse events in those patients experiencing high blood levels of the drug or rendering the treatment less effective or ineffective in those patients experiencing low blood levels of the drug.
  • In order to increase the bioavailability of PI's it is recommended that patients take the gelatin capsule formulation following a meal to increase the overall bioavailability of the active ingredient. Bioavailability can further vary depending on fat content in each meal. Unfortunately, many patients do not always adhere to this routine due to the complexity of their treatment regimens or otherwise. Often patients will take the medication on an empty stomach that leads to low bioavailability of the drug, and perhaps ineffective treatment.
  • Therefore, it is desirable to have a PI dosage form that reduces or eliminates gastrointestinal adverse events. It is also desirable to have such a dosage form that can be loaded with more active ingredient to reduce the pill burden on patients. Furthermore, it is desirable to have a dosage form that provides little variability in the blood levels of the PI within a subject and throughout a patient population. Another desirable feature would be a dosage form that provides similar blood levels of a PI regardless of whether or not a patient takes the medication following a meal. Yet another desirable feature would be a dosage form that does not have to be refrigerated to prevent degradation of the PI.
  • SUMMARY OF THE INVENTION
  • Surprisingly, it has been discovered that by formulating an undissolved form of a PI (in particular lopinavir and a lopinavir/ritonavir combination) in a pharmaceutical dosage form, all of the aforementioned disadvantages associated with dosage forms containing a dissolved PI can be overcome. In particular, pharmaceutical dosage forms containing the undissolved PI reduce pill burdens on HIV patients, in large measure because the drug load in these formulations can be increased. Additionally, such formulations can be stored at room temperature and do not require refrigeration. Moreover, these formulations provide a more consistent blood level of the PI among patients taking such therapy which helps insure an effective therapeutic benefit and less adverse events. Further, these consistent blood levels can be achieved with the formulation provided herein without regard to whether or not the patient has eaten or what type of meal was eaten. It is believed that this is the first time that an undissolved form of lopinavir has been formulated in a solid dosage form. Given the advantages attendant to such formulation, this represents the next breakthrough in HIV therapy which will help ease the complicated treatment regimens currently prescribed for HIV patients.
  • IN THE DRAWINGS
  • In the drawings,
  • FIG. 1 shows Box (lower and upper quartiles) and Whiskers (5th and 95th percentiles) Plots for Lopinavir AUC Under Various Meal Conditions; and
  • FIG. 2 shows Box (lower and upper quartiles) and Whiskers (5th and 95th percentiles) Plots for Lopinavir Cmax Under Various Meal Conditions.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • The term “AUC∞” is the area under the concentration time curve (AUC) extrapolated to infinity or the AUC to the last measured time point+(last measured concentration/elimination rate constant).
  • The term “Cmax” is defined as the observed maximum plasma concentration of an active ingredient. “Pharmaceutically acceptable” as used herein means moieties or compounds that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio.
  • The term “weight percent” or “percent by weight” or “wt %” is defined as the weight of the individual component in the formulation divided by the total weight of all components of the formulation and then multiplied by 100. In some cases where a formulation has an outer coating, then weight of the coating can either be included or excluded in the total weight.
  • The phrase “fasting/fasted state or condition” generally is defined as 10 hours of abstinence from eating prior to dosing and 4 hours post-dosing, although those skilled in the art will recognize various other timings that would also qualify as a fasting or fasted state.
  • The phrase “moderate-fat meal condition” is defined as receiving a meal that is approximately 500-600 KCal wherein 20-30% of the calories are from fat served approximately 30 minutes prior to dosing.
  • The phrase “high-fat meal condition” is defined as receiving a meal that is approximately 1000 Kcal wherein 50-55% of the calories are from fat served approximately 30 minutes prior to dosing and is used herein to refer to a “fed state” although those skilled in the art will recognize various meal conditions that would also qualify as a fed state.
  • The term “solid solution” is defined as a system in a solid state wherein the drug is molecularly dispersed throughout a matrix such that the system is chemically and physically uniform or homogenous throughout.
  • The term “solid dispersion” is defined as a system having small particles, typically of less than 400 μm in size, more typically less than 100 μm in size, and most typically less than 10 μm in size, of one phase dispersed in another phase (the carrier phase).
  • Suitable PI's for use in accordance with the present invention include but are not limited to (2S,3S,5S)-5-(N-(N-((N-methyl-N-((2-isopropyl-4-thiazolyl)-methyl)amino)carbonyl)-L-valinyl)amino-2-(N-(5-thiazolyl)methoxy-carbonyl)-amino)-amino-1,6-diphenyl-3hydroxyhexane (ritonavir); (2S,3S,5S)-2-(2,6-Dimethylphenoxyacetyl)amino-3-hydroxy-5-[2S-(1-tetrahydro-pyrimid-2-onyl)-3-methylbutanoyl]-amino-1,6-diphenylhexane (ABT-378; lopinavir); N-(2(R)-hydroxy-1(S)-indanyl)-2(R)-phenylmethyl-4(S)-hydroxy-5-(1-(4-(3-pyridylmethyl)-2(S)-N′-(t-butylcarboxamido)-piperazinyl))-pentaneamide (indinavir); N-tert-butyl-decahydro-2-[2(R)-hydroxy-4-phenyl-3(S)-[[N-(2-quinolylcarbonyl)-L-asparaginyl]amino]butyl]-(4aS,8aS)-isoquinoline-3(S)-carboxamide (saquinavir); 5(S)-Boc-amino-4(S)-hydroxy-6-phenyl-2(R)phenylmethylhexanoyl-(L)-Val-(L)-Phe-morpholin-4-ylamide; 1-Naphthoxyacetyl-beta-methylthio-Ala-(2S,3S)3-amino-2-hydroxy-4-butanoyl-1,3-thiazolidine-4t-butylamide; 5-isoquinolinoxyacetyl-beta-methylthio-Ala-(2S,3S)-3amino-2-hydroxy-4-butanoyl-1,3-thiazolidine-4-tbutylamide; [1S-[1R-(R-),2S*])-N1 [3-[[[(1,1-dimethylethyl)amino]carbonyl](2-methylpropyl)amino]-2hydroxy-1-(phenylmethyl)propyl]-2-[(2-quinolinylcarbonyl)amino]-butanediamide; amprenavir (VX-478); DMP-323; DMP-450; AG1343 (nelfinavir); atazanavir (BMS 232,632); tipranavir; palinavir; TMC-114; RO033-4649; fosamprenavir (GW433908); P-1946; BMS 186,318; SC-55389a; BILA 1096 BS; and U-140690, or any combinations thereof, whether used for PI activity or otherwise, such as with the case of ritonavir that can sometimes be employed as a cytochrome P450 monooxygenase inhibitor (variously referred to as a “pK booster). Preferred PIs are lopinavir and ritonavir alone, or in combination.
  • Generally, dosage forms of the present invention will comprise a therapeutically effective amount of at least one PI. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and other factors known to those of ordinary skill in the medical arts. For example, it is well within the skill of the art to start doses of the compound at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. Typically, however, a pharmaceutical dosage form of the present invention will comprise from about 5 to about 30% by weight of the total dosage form, preferably from about 10 to about 25% by weight of the total dosage form, of an HIV protease inhibitor or a combination of HIV protease inhibitors. Preferably, the dosage form will contain between about 10 mg to about 1500 mg of a PI. Most preferably, the dosage form will comprise lopinavir and ritonavir in a ratio of about 4:1 respectively. The preferred dose of lopinavir and ritonavir is 400 mg and 100 mg respectively which can be divided evenly between multiple dosage forms, preferably two. It will be understood that multiple doses, typically two, can be given in a given day.
  • Pharmaceutical dosage forms provided herein generally will comprise an “undissolved” PI. In contradistinction to existing gelatin capsules filled with a PI dissolved in a solvent, undissolved PI's as used herein means that the PI is in a solid form and not dissolved in a liquid carrier in its final dosage form. Solid forms of a PI may include, for example, crystalline, micronized crystalline, crystalline nanoparticulates, amorphous, micronized amorphous, amorphous nanoparticulates, or preferably amorphous solid forms of a PI.
  • Many pharmaceutical dosage forms are acceptable for use in accordance with the present invention; the choice of which is well within the skill of a person of ordinary skill in this art based upon the properties of the dosage forms provided herein. For example, orally administered solid dosage forms include but are not limited to capsules, dragees, granules, pills, powders, and tablets. Excipients commonly used to formulate such dosage forms include encapsulating materials or formulation additives such as absorption accelerators, antioxidants, binders, buffers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, and mixtures thereof. Excipients for orally administered compounds in solid dosage forms include agar, alginic acid, aluminum hydroxide, benzyl benzoate, 1,3-butylene glycol, castor oil, cellulose, cellulose acetate, cocoa butter, corn starch, corn oil, cottonseed oil, ethanol, ethyl acetate, ethyl carbonate, ethyl cellulose, ethyl laureate, ethyl oleate, gelatin, germ oil, glucose, glycerol, groundnut oil, isopropanol, isotonic saline, lactose, magnesium hydroxide, magnesium stearate, malt, olive oil, peanut oil, potassium phosphate salts, potato starch, propylene glycol, talc, tragacanth, water, safflower oil, sesame oil, sodium carboxymethyl cellulose, sodium lauryl sulfate, sodium phosphate salts, soybean oil, sucrose, tetrahydrofurfuryl alcohol, and mixtures thereof.
  • A preferred dosage form, will generally comprise at least one HIV protease inhibitor in a therapeutically effective amount, at least one pharmaceutically acceptable water-soluble polymer and at least one pharmaceutically acceptable surfactant.
  • More preferably, a solid solution or solid dispersion can be formed into one of the above pharmaceutical dosage forms. Such solutions or dispersions can be manufactured with suitable pharmaceutically acceptable water-soluble polymers including but not limited to water-soluble polymers having a Tg of at least about 50° C., preferably at least about 60° C., most preferred from about 80° C. to about 180° C. Methods for determining Tg values of the organic polymers are described in “Introduction to Physical Polymer Science”, 2nd Edition by L. H. Sperling, published by John Wiley & Sons, Inc., 1992. The Tg value can be calculated as the weighted sum of the Tg values for homopolymers derived from each of the individual monomers, i.e., that make up the polymer: Tg=ΣWiXi where W is the weight percent of monomer i in the organic polymer, and X is the Tg value for the homopolymer derived from monomer i. Tg values for the homopolymers may be taken from “Polymer Handbook”, 2nd Edition by J. Brandrup and E. H. Immergut, Editors, published by John Wiley & Sons, Inc., 1975.
  • Water-soluble polymers having a Tg as defined above allow for the preparation of solid solutions or solid dispersions that are mechanically stable and, within ordinary temperature ranges, sufficiently temperature stable so that the solid solutions or solid dispersions may be used as dosage forms without further processing or be compacted to tablets with only a small amount of tableting aids.
  • The water-soluble polymer comprised in the preferred dosage form is a polymer that preferably has an apparent viscosity, when dissolved at 20° C. in an aqueous solution at 2% (w/v), of about 1 to about 5000 mPa·s, and more preferably of about 1 to about 700 mPa·s, and most preferred of about 5 to about 100 mPa·s.
  • Water-soluble polymers suitable for use in the preferred dosage form of the present invention include but are not limited to homopolymers and copolymers of N-vinyl lactams, especially homopolymers and copolymers of N-vinyl pyrrolidone, e.g. polyvinylpyrrolidone (PVP), copolymers of N-vinyl pyrrolidone and vinyl acetate or vinyl propionate, cellulose esters and cellulose ethers, in particular methylcellulose and ethylcellulose, hydroxyalkylcelluloses, in particular hydroxypropylcellulose, hydroxyalkylalkylcelluloses, in particular hydroxypropylmethylcellulose, cellulose phthalates or succinates, in particular cellulose acetate phthalate and hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate or hydroxypropylmethylcellulose acetate succinate; high molecular polyalkylene oxides such as polyethylene oxide and polypropylene oxide and copolymers of ethylene oxide and propylene oxide, polyacrylates and polymethacrylates such as methacrylic acid/ethyl acrylate copolymers, methacrylic acid/methyl methacrylate copolymers, butyl methacrylate/2-dimethylaminoethyl methacrylate copolymers, poly(hydroxyalkyl acrylates), poly(hydroxyalkyl methacrylates), polyacrylamides, vinyl acetate polymers such as copolymers of vinyl acetate and crotonic acid, partially hydrolyzed polyvinyl acetate (also referred to as partially saponified “polyvinyl alcohol”), polyvinyl alcohol, oligo- and polysaccharides such as carrageenans, galactomannans and xanthan gum, or mixtures of one or more thereof.
  • Of these, homopolymers or copolymers of N-vinyl pyrrolidone, in particular a copolymer of N-vinyl pyrrolidone and vinyl acetate, are preferred. A particularly preferred polymer is a copolymer of about 60% by weight of the copolymer, N-vinyl pyrrolidone and about 40% by weight of the copolymer, vinyl acetate.
  • According to the preferred dosage form of the present invention, the pharmaceutical dosage form comprises from about 50 to about 85% by weight of the total dosage form, preferably from about 60 to about 80% by weight of the total dosage form, of a water-soluble polymer or any combination of such polymers.
  • The term “pharmaceutically acceptable surfactant” as used herein refers to a pharmaceutically acceptable non-ionic surfactant. In one embodiment, the present invention provides a dosage form comprising at least one surfactant having an hydrophilic lipophilic balance (HLB) value of from about 4 to about 10, preferably from about 7 to about 9. The HLB system (Fiedler, H. B., Encylopedia of Excipients, 5th ed., Aulendorf: ECV-Editio-Cantor-Verlag (2002)) attributes numeric values to surfactants, with lipophilic substances receiving lower HLB values and hydrophilic substances receiving higher HLB values.
  • Surfactants having an HLB value of from about 4 to about 10 suitable for use in the present invention include but are not limited to polyoxyethylene alkyl ethers, e.g. polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether; polyoxyethylene alkylaryl ethers, e.g. polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether, polyoxyethylene (3) octylphenyl ether; polyethylene glycol fatty acid esters, e.g. PEG-200 monolaurate, PEG-200 dilaurate, PEG-300 dilaurate, PEG400 dilaurate, PEG-300 distearate, PEG-300 dioleate; alkylene glycol fatty acid mono esters, e.g. propylene glycol monolaurate (Lauroglycol®); sucrose fatty acid esters, e.g. sucrose monostearate, sucrose distearate, sucrose monolaurate, sucrose dilaurate; or sorbitan fatty acid mono esters such as sorbitan mono laurate (Span® 20), sorbitan monooleate, sorbitan monopalmitate (Span® 40), or sorbitan stearate, or mixtures of one or more thereof.
  • The sorbitan mono fatty acid esters are preferred, with sorbitan mono laurate and sorbitan monopalmitate being particularly preferred.
  • A preferred dosage form of the present invention comprises from about 2 to about 20% by weight of the total dosage form, preferably from about 3 to about 15% by weight of the total dosage form, of the surfactant or combination of surfactants.
  • Besides the surfactant having an HLB value of from about 4 to about 10, the preferred dosage form may comprise additional pharmaceutically acceptable surfactants such as polyoxyethylene castor oil derivates, e.g. polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate such as polyethylenglycol 40 hydrogenated castor oil (Cremophor® RH 40) or polyethylenglycol 60 hydrogenated castor oil (Cremophor® RH 60); or block copolymers of ethylene oxide and propylene oxide, also known as polyoxyethylene polyoxypropylene block copolymers or polyoxyethylene polypropyleneglycol, such as Poloxamer® 124, Poloxamer® 188, Poloxamer® 237, Poloxamer® 388, Poloxamer® 407 (BASF Wyandotte Corp.); or a mono fatty acid ester of polyoxyethylene (20) sorbitan, e.g. polyoxyethylene (20) sorbitan monooleate (Tween® 80), polyoxyethylene (20) sorbitan monostearate (Tween® 60), polyoxyethylene (20) sorbitan monopalmitate (Tween® 40), polyoxyethylene (20) sorbitan monolaurate (Tween® 20).
  • Where such additional surfactants are used, the surfactant having an HLB value of from about 4 to about 10 generally accounts for at least about 50% by weight, preferably at least about 60% by weight, of the total amount of surfactant used.
  • The dosage form of the present invention can include additional excipients or additives such as, for example, flow regulators, lubricants, bulking agents (fillers) and disintegrants. Such additional excipients may comprise from about 0 to about 15% by weight of the total dosage form.
  • The preferred solid dispersion or solid solution based dosage form of the present invention can be produced by preparing a solid solution or solid dispersion of the HIV protease inhibitor, or the combination of HIV protease inhibitors, in a matrix of a water-soluble polymer and a surfactant, and then shaping into the required tablet form. Alternatively, the solid solution or solid dispersion product can be subdivided to granules, e.g. by grinding or milling, and the granules may subsequently be compacted to tablets.
  • Various techniques exist for preparing solid solutions or solid dispersions including melt-extrusion, spray-drying and solution-evaporation with melt-extrusion being preferred.
  • The melt-extrusion process comprises the steps of preparing a homogeneous melt of the HIV protease inhibitor or the combination of HIV protease inhibitors, the water-soluble polymer and the surfactant, and cooling the melt until it solidifies. “Melting” means a transition into a liquid or rubbery state in which it is possible for one component to get embedded homogeneously in the other. Typically, one component will melt and the other components will dissolve in the melt thus forming a solution. Melting usually involves heating above the softening point of the water-soluble polymer. The preparation of the melt can take place in a variety of ways. The mixing of the components can take place before, during or after the formation of the melt. For example, the components can be mixed first and then melted or be simultaneously mixed and melted. Usually, the melt is homogenized in order to disperse the active ingredients efficiently. Also, it may be convenient first to melt the water-soluble polymer and then to mix in and homogenize the active ingredients.
  • Usually, the melt temperature is in the range of about 70 to about 250° C., preferably from about 80 to about 180° C., most preferred from about 100 to about 140° C.
  • The active ingredients can be employed as such or as a solution or dispersion in a suitable solvent such as alcohols, aliphatic hydrocarbons or esters. Another solvent which can be used is liquid carbon dioxide. The solvent is removed, e.g. evaporated, upon preparation of the melt.
  • Various additives may be included in the melt, for example flow regulators such as colloidal silica; lubricants, fillers, disintegrants, plasticizers, stabilizers such as antioxidants, light stabilizers, radical scavengers, stabilizers against microbial attack.
  • The melting and/or mixing takes place in an apparatus customary for this purpose. Particularly suitable ones are extruders or kneaders. Suitable extruders include single screw extruders, intermeshing screw extruders or else multiscrew extruders, preferably twin screw extruders, which can be corotating or counterrotating and, optionally, be equipped with kneading disks. It will be appreciated that the working temperatures will also be determined by the kind of extruder or the kind of configuration within the extruder that is used. Part of the energy needed to melt, mix and dissolve the components in the extruder can be provided by heating elements. However, the friction and shearing of the material in the extruder may also provide a substantial amount of energy to the mixture and aid in the formation of a homogeneous melt of the components.
  • The melt ranges from pasty to viscous. Shaping of the extrudate conveniently is carried out by a calender with two counter-rotating rollers with mutually matching depressions on their surface. A broad range of tablet forms can be attained by using rollers with different forms of depressions. Alternatively, the extrudate is cut into pieces, either before (hot-cut) or after solidification (cold-cut).
  • Optionally, the resulting solid solution or solid dispersion product is milled or ground to granules. The granules may then be compacted. Compacting means a process whereby a powder mass comprising the granules is densified under high pressure in order to obtain a compact with low porosity, e.g. a tablet. Compression of the powder mass is usually done in a tablet press, more specifically in a steel die between two moving punches. Where a solid dosage form of the invention comprises a combination of more than one HIV protease inhibitor (or a combination of an HIV protease inhibitor with one or more other active ingredients) it is of course possible to separately prepare solid solution or solid dispersion products of the individual active ingredients and to blend the milled or ground products before compacting.
  • At least one additive selected from flow regulators, disintegrants, bulking agents (fillers) and lubricants is preferably used in compacting the granules. Disintegrants promote a rapid disintegration of the compact in the stomach and keeps the granules which are liberated separate from one another. Suitable disintegrants are crosslinked polymers such as crosslinked polyvinyl pyrrolidone and crosslinked sodium carboxymethylcellulose. Suitable bulking agents (also referred to as “fillers”) are selected from lactose, calcium hydrogenphosphate, microcrystalline cellulose (Avicell®), silicates, in particular silicium dioxide, magnesium oxide, talc, potato or corn starch, isomalt, polyvinyl alcohol.
  • Suitable flow regulators are selected from highly dispersed silica (Aerosil®), and animal or vegetable fats or waxes.
  • A lubricant is preferably used in compacting the granules. Suitable lubricants are selected from polyethylene glycol (e.g., having a Mw of from 1000 to 6000), magnesium and calcium stearates, sodium stearyl fumarate, and the like.
  • Various other additives may be used, for example dyes such as azo dyes, organic or inorganic pigments such as aluminium oxide or titanium dioxide, or dyes of natural origin; stabilizers such as antioxidants, light stabilizers, radical scavengers, stabilizers against microbial attack.
  • Dosage forms according to the invention may be provided as dosage forms consisting of several layers, for example laminated or multilayer tablets. They can be in open or closed form. “Closed dosage forms” are those in which one layer is completely surrounded by at least one other layer. Multilayer forms have the advantage that two active ingredients which are incompatible with one another can be processed, or that the release characteristics of the active ingredient(s) can be controlled. For example, it is possible to provide an initial dose by including an active ingredient in one of the outer layers, and a maintenance dose by including the active ingredient in the inner layer(s). Multilayer tablets types may be produced by compressing two or more layers of granules. Alternatively, multilayer dosage forms may be produced by a process known as “coextrusion”. In essence, the process comprises preparation of at least two different melt compositions as explained above, and passing these molten compositions into a joint coextrusion die. The shape of the coextrusion die depends on the required drug form. For example, dies with a plain die gap, called slot dies, and dies with an annular slit are suitable.
  • In order to facilitate the intake of such a dosage form by a mammal, it is advantageous to give the dosage form an appropriate shape. Large tablets that can be swallowed comfortably are therefore preferably elongated rather than round in shape.
  • A film coat on the tablet further contributes to the ease with which it can be swallowed. A film coat also improves taste and provides an elegant appearance. If desired, the film-coat may be an enteric coat. The film-coat usually includes a polymeric film-forming material such as hydroxypropyl methylcellulose, hydroxypropylcellulose, and acrylate or methacrylate copolymers. Besides a film-forming polymer, the film-coat may further comprise a plasticizer, e.g. polyethylene glycol, a surfactant, e.g. a Tween® type, and optionally a pigment, e.g. titanium dioxide or iron oxides. The film-coating may also comprise talc as anti-adhesive. The film coat usually accounts for less than about 5% by weight of the dosage form.
  • The benefits provided by the present invention are presently believed to be attributable to the pharmacokinetic (pK) properties of the dosage form. Pharmacokinetic properties are generally understood to mean the manner and extent to which a drug is absorbed. Common pK parameters include AUC (or “area under the curve”), which typically refers to the amount of drug that is measurable in blood or blood products of a person taking the drug over time. AUC is variously referred to as a patients exposure to a drug. Cmax is another pK term which refers to the maximum blood (or blood product) level over the course of a given regimen of a drug. Drug regimens for which pK parameters are measured include “clinical studies.” Some clinical studies are performed in a finite population of healthy volunteer patients and are designed to determine the pK parameters of a drug (such as those mentioned above), and not to treat a patient. Each patient is thus called a member of the study population. While such clinical studies are carefully controlled and monitored, pK parameters can vary between clinical studies in large measure because different clinical studies are performed on different populations of patients. Although variances exist between clinical studies, those skilled in the art readily recognize that once a particular set of pK parameters is generally known, it is a matter of routine to formulate a drug to achieve a similar set of pK parameters.
  • As previously mentioned, the present invention provides a dosage form that can be taken without regard to whether a patient has eaten, sometimes referred to as “without regard to meals”, “can be taken with or without food”, “no food effect” or similar phrases. In particular, the Cmax of the drug and AUC of the drug is similar in patients that have eaten (“fed state”) as compared to patients that have not eaten (“fasted state”). Hence, the dosage form provided herein advantageously can be taken at any time regardless of whether or not patients have recently eaten.
  • Notwithstanding the previous definition, there is no completely standard definitions for fed and fasted states. Generally, however, a fasted state refers to the fact that a patient has not eaten for a given amount of time before taking a dose of medication, as well as not eating for a given amount of time after taking the dosage form. These time periods before and after dosing are a matter of choice, and can range between, for example 2 hours to 24 hours. A fed state generally refers to the fact that a patient has eaten within a given time period of taking a particular medication. This time period is variable but may constitute, for example, a meal just before, during, or just after taking the medication, typically a meal is eaten within about an hour of dosing. The quantity of food eaten that will qualify as a fed state is also variable but generally can comprise between about 500 to about 1500 Kcal of food.
  • The dosage forms provided herein will have substantially the same Cmax and AUC∞ values in patients in a fasted state as well as in a fed state, regardless of the dose given. In particular, the mean of the individual patient ratios in a patient population for either the Cmax or AUC∞ in the fed state to fasted state will be in the range of about 0.7 to about 1.43; more preferably between about 0.75 and about 1.35; and most preferably between about 0.8 and about 1.25. Thus for example, in study population of 30 individuals each patient is given a dose of drug in a fed state and, after an appropriate time period, a dose of the drug in a fasted state. The AUC∞ and Cmax for both meal conditions are calculated for each patient. The AUC∞ value for the fed state is then divided by the AUC∞ for the fasted state for each patient. The individual patient values are then added together and then divided by the number of patients completing the study to arrive at a mean AUC∞ value for all patients completing the study. The mean Cmax value is calculated in a similar manner. If the mean value of the fed to fasted ratio for all patients' Cmax or AUC∞ values in a given study is within 0.7 to 1.43, for example, then the dosage form provided to the patients would be considered to capable of administration without regard to whether or not the patient was in a fed or fasted state.
  • As also previously mentioned, the dosage forms provided herein have less variability than other gelatin capsule based formulations containing a dissolved form of the drug or drugs. This lack of variability is evidenced in FIG. 1 and FIG. 2 which compare AUC∞ and Cmax data of an embodiment of the present invention and the data from a marketed gelatin capsule containing a dissolved PI. As shown by the Figures, the AUC∞ and Cmax data associated with an embodiment of the present invention shows less variation. In particular, the graphs are a “box and whiskers” plot of the data comparing the two formulations wherein the bottom of any given “whisker” (labeled A in the first box and whisker plot of FIG. 1) is called the “5th percentile”, meaning that 5% of the patients in the study fell below the designated AUC∞ or Cmax value for the particular whisker. The top of the whisker (labeled D in first box and whisker plot of FIG. 1) represents the “95th percentile”, meaning that 5% of the patients in the study had a AUC∞ or Cmax value above the value designated by the top of any particular whisker. Similarly, the bottom of any particular box (labeled B in first box and whisker plot of FIG. 1) represents the 25th percentile and the top of any particular box (labeled C in first box and whisker plot of FIG. 1) represent the 75th percentile. The line running through any particular box is the 50th percentile or median of any particular study population.
  • As seen by the Figures, the data generally demonstrates that the variability associated with the embodiment of the present invention is less than that associated with the existing gelatin capsule formulation. Looking at the dosage forms given under fasting conditions of FIG. 1 (for example), the difference between 95th percentile and 5th percentile of the gelatin capsule is greater than the difference between 95th percentile and 5th percentile of the embodiment of the present invention. This translates into the fact that a greater portion of the study population is getting a therapeutic benefit from the PI without experiencing adverse events do to overexposure of the drug. For purposes of, for example, reducing side effects and achieving therapeutic levels, it is generally preferred that the difference between the 95th percentile of AUC∞ and 5th percentile of AUC∞ of any given study population taking a dosage form as provided herein (regardless of whether the population is fed or fasted) is less than about 180, more preferably less than about 175, even more preferably less than about 165, and most preferably less than about 160. Under fasting conditions, it is preferable that the difference between the 95th percentile of AUC∞ and 5th percentile of AUC∞ of any given study population taking a dosage form as provided herein is less than about 170, more preferably less than about 160, and most preferably less than about 150. Under fed conditions, it is preferable that the difference between the 95th percentile of AUC∞ and 5th percentile of AUC∞ of any given study population taking a dosage form as provided herein is less than about 130, more preferably less than about 120, and most preferably less than about 110.
  • Similarly to the differences between the 95th and 5th percentiles provided above, the difference between the 75th percentile and 25th percentile of the AUC data in FIG. 1 is also very important in demonstrating the lack of variability in dosage forms of the present invention. It is generally preferred that the difference between the 75th percentile of AUC∞ and 25th percentile of AUC∞ of any given study population taking a dosage form as provided herein (regardless of whether the population is fed or fasted) is less than about 60, more preferably less than about 55, even more preferably less than about 50. Under fasting conditions, it is preferable that the difference between the 75th percentile of AUC∞ and 25th percentile of AUC∞ of any given study population taking a dosage form as provided herein is less than about 65, more preferably less than about 60, and most preferably less than about 55. Under fed conditions, it is preferable that the difference between the 75th percentile of AUC∞ and 25th percentile of AUC∞ of any given study population taking a dosage form as provided herein is less than about 60, more preferably less than about 50, and most preferably less than about 40.
  • In terms of ranges of AUC∞ values, it is preferred that under fasted conditions the 5th percentile to the 95th percentile of AUC∞ of any given study population taking a dosage form as provided herein ranges between about 33 μg·h/mL and about 175 μg·h/mL; and the 25th percentile to the 75th percentile of AUC∞ of any given study population taking a dosage form as provided ranges between about 54 μg·h/mL and about 107 μg·h/mL. Under fed conditions it is preferred that the 5th percentile to the 95th percentile of AUC∞ of any given study population taking a dosage form as provided herein ranges between about 57 μg·h/mL and about 142 μg·h/mL; and the 25th percentile to the 75th percentile of AUC∞ of any given study population taking a dosage form as provided herein ranges between about 75 μg·h/mL and about 109 μg·g/mL. It is also preferred that the 5th percentile of the AUC∞ of any given study population taking a dosage form as provided herein greater than about 30 μg·h/mL under fasted conditions, and greater than about 50 μg·g/mL under fed conditions. Finally with respect to AUC∞, it is preferred that under fasting conditions the mean AUC∞ is between about 60 μg·h/mL and about 95 μg·h/mL. for any given study population taking a dosage form provided.
  • Similarly to the AUC parameters associated with FIG. 1, the Cmax parameters shown in FIG. 2 also demonstrates lack of variability associated with dosage forms provided herein. For example, looking at the box and whiskers plot of FIG. 2 for patients under fasting conditions taking a dose of PI formulated according to the present invention, it is preferred that difference between the 95th percentile and the 5th percentile is less than about 15, more preferably less than about 13, and most preferably less than about 11. Under fasted conditions it is also preferable that the 5th percentile of Cmax of a given study population taking a dose of active ingredient formulated according to the present invention is greater that about 2.5 μg/mL. Turning to the box and whiskers plot of FIG. 2 for fed conditions taking a dosage form of the present invention, it is preferred that difference between the 95th percentile and the 5th percentile is less than about 12, more preferably less than about 11.
  • With respect to the description of the figures provided above, it should be pointed out that when a patient is referred to as taking a dosage form of the present invention, they received a dose of a PI in multiple dosage forms. Specifically, the so called dosage form contained 400 mg of lopinavir and 100 mg of ritonovir evenly divided between two dosage forms. Lopinavir was the only drug measured in these studies due to the fact that ritonavir is supplied not for its action as a PI but as a pharmacokinetic enhancer or booster (ritonavir inhibits the metabolism of lopinavir). Further, it will be understood that the when ritonavir is employed it can be separately dosed instead of part of a combination dosage form. Moreover, it will be understood that the values given can vary due to, for example, changes in meal timings and quantities, as well as the constitution of the study population. It is well known that study populations from different nationalities may have different drug metabolism rates. Accordingly, in cases where study data is taken from such populations, the data may have to be normalized as is well known in the art. Moreover, in cases where an increase in the dose or a decrease in the dose of lopinovir, for example, is provided to a study population, the data resulting from such dosing may require normalization using appropriate modeling as is well known in the art. Last, with respect to the above discussion concerning the figures, a “High Fat Meal” as described in the figures is considered to be a fed state.
  • In addition to providing methods of treating a human patient afflicted with HIV/AIDs, the present invention provides methods of reducing the side effects associated with HIV therapy, methods of increasing the bioavailability of a PI, methods of decreasing the pill burden of an HIV/AIDs patient, methods of decreasing the variability of blood levels of a PI in a patient taking PI therapy, and methods of providing a PI to a patient taking PI therapy. All of these methods comprise the step of providing a pharmaceutical dosage form comprising a therapeutically effective amount of an undissolved form of a PI to a patient. Preferably, the PI is (2S,3 S,5S)-2-(2,6-Dimethylphenoxyacetyl)amino-3-hydroxy-5-[2S-(1-tetrahydro-pyrimid-2-onyl)-3-methylbutanoyl]-amino-1,6-diphenylhexane (ABT-378; lopinavir). More preferably, the dosage form will comprise (2S,3S,5S)-5-(N-(N-((N-methyl-N-((2-isopropyl-4-thiazolyl)methyl)amino)carbonyl)-L-valinyl)amino-2-(N-((5-thiazolyl)-methoxy-carbonyl)-amino)-amino-1,6-diphenyl-3hydroxyhexane (ritonavir).
  • EXAMPLES
  • The following examples are provided to further understand and illustrate the present invention and not to limit the spirit and scope of the present invention as it is defined in the appended claims.
  • Example 1
  • Component Weight % Weight % Weight %
    Ritonavir  18-22.5 4.17 4.17
    Lopinavir in total 16.67 16.67
    Copovidone (N- 65-75  71.16 70.12
    vinyl
    pyrrolidone/vinyl
    acetate copolymer
    60:40)
    Span 20 (Sorbitan 4-10 7.0 5.02
    monolaurate)
    Cremophor RH40 0-10 3.02
    (polyoxyethylene-
    glycerol
    oxystearate)
    Colloidal silica 0-3  1.0 1.0
  • Copovidone (N-vinyl pyrrolidone/vinyl acetate copolymer 60:40) was mixed with ritonavir (4.17 parts by weight), lopinavir (16.67 parts by weight) and colloidal silica (1.0 part by weight). The powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.0 kg/h and a melt temperature of 133° C. The clear, fully transparent melt was fed to a calender with two counter-rotating rollers having mutually matching cavities on their surfaces. Tablets of 1080 mg were thus obtained. DSC and WAXS analysis did not reveal any evidence of crystalline drug material in the formulation.
  • The bioavailability of the formulation was assessed using beagle dogs (mixed sexes, weighing approximately 10 kg) which received a balanced diet with 27% fat and were permitted water ad libitum. Each dog received a 100 μg/kg subcutaneous dose of histamine approximately 30 minutes prior to dosing. A single dose corresponding to about 200 mg lopinavir, about 50 mg ritonavir, or about 200 mg lopinavir and about 50 mg ritonavir, respectively, was administered to each dog. The dose was followed by approximately 10 milliliters of water. Blood samples were obtained from each animal prior to dosing and 0.25, 0.5, 1.0, 1.5, 2, 3, 4, 6, 8, 10, 12 and 24 hours after drug administration. The plasma was separated from the red cells by centrifugation and frozen (−30° C.) until analysis. Concentrations of HIV protease inhibitors were determined by reverse phase HPLC with low wavelength UV detection following liquid-liquid extraction of the plasma samples. The area under the curve (AUC) was calculated by the trapezoidal method over the time course of the study. Each dosage form was evaluated in a group containing 8 dogs; the values reported are averages for each group of dogs.
  • The dose-adjusted AUC in dogs was 0.52 μg.h/ml/100 mg for ritonavir and 4.54 μg.h/m/100 mg for lopinavir. This example shows that solid solutions or solid dispersions of HIV protease inhibitors without added surfactant yield a very poor bioavailability.
  • Example 2
  • Component Weight % Weight %
    Ritonavir   18-22.5 20.8
    Lopinavir
    Copovidone (N-vinyl 60-75 63.15
    pyrrolidone/vinyl acetate
    copolymer 60:40)
    Span 20 (Sorbitan  5-15
    monolaurate) in total
    Cremophor RH40 10.00
    (polyoxyethyleneglycerol
    oxystearate)
    PEG 6000 0-8 5.00
    Colloidal silica 0-3 1.04
  • The above composition is processed by melt extrusion. The resulting extrudate can be used as such or milled and compressed into tablets, preferably by the use of suitable tabletting aids such as sodium stearyl fumarate, colloidal silica, lactose, isomalt, calcium silicate, and magnesium stearate, cellulose or calcium hydrogenphosphate.
  • Example 3
  • Component Weight %
    Ritonavir 4.16
    Lopinavir 16.67
    Copovidone (N-vinyl pyrrolidone/vinyl 78.17
    acetate copolymer 60:40)
    Colloidal silica 1.0
  • Copovidone (N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 78.17 parts by weight) was mixed with ritonavir (4.16 parts by weight), lopinavir (16.67 parts by weight) and colloidal silica (1.0 part by weight). The powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.0 kg/h and a melt temperature of 133° C. The clear, fully transparent melt was fed to a calender with two counter-rotating rollers having mutually matching cavities on their surfaces. Tablets of 1080 mg were thus obtained. DSC and WAXS analysis did not reveal any evidence of crystalline drug material in the formulation.
  • Example 4
  • Component Weight %
    Ritonavir 4.17
    Lopinavir 16.67
    Copovidone 68.17
    Cremophor RH40 10.00
    colloidal silica 1.0
    lactose monohydrate 6.0
    crosslinked PVP 6.0
    colloidal silica 1.0
    magnesium stearate 0.51
  • Copovidone (N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 68.17 parts by weight) was blended with Cremophor RH40 (polyoxyethyleneglycerol oxystearate; 10.00 parts by weight) in a Diosna high-shear mixer. The resulting granules were mixed with ritonavir (4.17 parts by weight), lopinavir (16.67 parts by weight) and colloidal silica (1.00 parts by weight). The powdery mixture was then fed into a Leistritz Micro 18 twin-screw extruder at a rate of 2.3 kg/h and a melt temperature of 126° C. The extrudate was cut into pieces and allowed to solidify. The extruded pieces were milled using a high impact universal mill. The milled material (86.49 parts by weight) was blended in a bin blender with lactose monohydrate (6.00 parts by weight), crosslinked PVP (6.00 parts by weight), colloidal silica (1.00 part by weight) and magnesium stearate (0.51 parts by weight). The powdery blend was compressed to tablets of 1378.0 mg on a Fette E 1 single punch tablet press. The tablets were then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • The bioavailability of the formulation was assessed using beagle dogs as in Example 1. The dose-adjusted AUC in dogs was 0.60 μg.h/ml/100 mg for ritonavir and 7.43 μg.h/ml/100 mg for lopinavir. This example shows that inclusion of a surfactant into solid solutions or solid dispersions of HIV protease inhibitors improves the bioavailability attained.
  • Example 5
  • Component Weight (mg)
    Ritonavir 50
    Lopinavir 200
    Copovidone 853.8
    Span 20 83.9
    colloidal silica 12
  • Copovidone (N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 853.8 parts by weight) was blended with Span 20 (Sorbitan monolaurate; 83.9 parts by weight) in a Diosna high-shear mixer. The resulting granules were mixed with ritonavir (50 parts by weight), lopinavir (200 parts by weight) and colloidal silica (12 parts by weight). The powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.1 kg/h and a melt temperature of 119° C. The extrudate was fed to a calender with two counter-rotating rollers having mutually matching cavities on their surfaces. Tablets of 1120 mg were thus obtained.
  • The bioavailability of the formulation was assessed using beagle dogs as in Example 1. The dose-adjusted AUC in dogs was 10.88 μg.h/ml/100 mg for ritonavir and 51.2 μg.h/ml/100 mg for lopinavir. This example shows that inclusion of a surfactant having an HLB of 4 to 10 into solid solutions or solid dispersions of HIV protease inhibitors markedly improves the bioavailability attained.
  • Example 6
  • Example 5 was repeated, however, the extrudate was cut into pieces and allowed to solidify. The extruded pieces were milled to a particle size of about 250 μm, using a high impact universal mill. The milled material was blended in a bin blender with sodium stearyl fumarate (12.3 parts by weight) and colloidal silica (8.0 parts by weight) for 20 min. The powdery blend was compressed on a rotary tablet machine with 3 punches (6500 tablets/h). The tablets were then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • The bioavailability of the formulation was assessed using beagle dogs as in Example 1. The dose-adjusted AUC in dogs was 14.24 μg.h/ml/100 mg for ritonavir and 52.2 μg.h/ml/100 mg for lopinavir.
  • Example 7
  • Component Weight (mg)
    Ritonavir 50
    Lopinavir 200
    Copovidone 841.3
    Span 20 60.2
    Cremophor RH40 36.2
    colloidal silica 12
  • Copovidone (N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 841.3 parts by weight) was blended with Cremophor RH40 (polyoxyethyleneglycerol oxystearate; 36.2 parts by weight), Span 20 (Sorbitan monolaurate; 60.2 parts by weight) in a Diosna high-shear mixer. The resulting granules were mixed with ritonavir (50 parts by weight), lopinavir (200 parts by weight) and colloidal silica (12 parts by weight). The powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.1 kg/h and a melt temperature of 114° C. The extrudate was fed to a calender with two counter-rotating rollers having mutually matching cavities on their surfaces. Tablets of 1120 mg were thus obtained.
  • The bioavailability of the formulation was assessed using beagle dogs as in Example 1. The dose-adjusted AUC in dogs was 10.96 μg.h/ml/100 mg for ritonavir and 46.5 μg.h/ml/100 mg for lopinavir. This example shows that a combination of a surfactant having an HLB of 4 to 10 and a further surfactant can successfully be used.
  • Example 8
  • Example 7 was repeated, however, the extrudate was cut into pieces and allowed to solidify. The extruded pieces were milled to a particle size of about 250 μM, using a high impact universal mill. The milled material was blended in a bin blender with sodium stearylfumarate (13.9 parts by weight), colloidal silica (7.0 parts by weight), isomalt DC100 (159.4 parts by weight) and calcium silicate (7.0 parts by weight) for 20 min. The blend was compressed and then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • The bioavailability of the formulation was assessed using beagle dogs as in Example 1. The dose-adjusted AUC in dogs was 10.38 μg.h/ml/100 mg for ritonavir and 42.7 μg.h/mV/100 mg for lopinavir.
  • Example 9
  • Component Weight (mg)
    Lopinavir 200
    Copovidone 683.3
    Span40 67.2
    colloidal silica 9.6
    Sodium stearylfumarate 7.9
    colloidal silica 11.3
    Isomalt DC100 129.1
    Sodium dodecyl sulfate 15.6
  • Copovidone (N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 683.3 parts by weight) was blended with Span 40 (sorbitan monopalmitate; 67.2 parts by weight) in a Diosna high-shear mixer. The resulting granules were mixed with lopinavir (200 parts by weight) and colloidal silica (9.6 parts by weight). The powdery mixture was then fed into a twin-screw extruder (screw diameter 18 mm) at a rate of 2.1 kg/h and a melt temperature of 119° C. The extrudate was cut into pieces and allowed to solidify. The extruded pieces were milled using a high impact universal mill. The milled material was blended in a bin blender with sodium stearylfumarate (7.9 parts by weight), colloidal silica (11.3 parts by weight), isomalt DC100 (129.1 parts by weight) and sodium dodecyl sulfate (15.6 parts by weight). The blend was compressed and then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • The bioavailability of the formulation was assessed using beagle dogs as in Example 1. Tablets corresponding to 200 mg lopinavir were coadministered to dogs together with 50 mg ritonavir. The dose-adjusted AUC of lopinavir was 38.8 μg.h/ml/100 mg.
  • Example 10
  • Component Weight (mg)
    Ritonavir 50
    Copovidone 151.5
    Cremophor RH40 24
    colloidal silica 3.8
    PEG 6000 12
    Isomalt DC100 31.9
    Calcium silicate 4.2
  • Copovidone (N-vinyl pyrrolidone/vinyl acetate copolymer 60:40; 151.5 parts by weight) was blended with Cremophor RH40 (24 parts by weight) and PEG 6000 (12 parts by weight) in a Diosna high-shear mixer. The resulting granules were mixed with ritonavir (50 parts by weight) and colloidal silica (2.4 parts by weight). The powdery mixture was then fed into a twin-screw extruder and was melt-extruded. The extrudate was cut into pieces and allowed to solidify. The extruded pieces were milled using a high impact universal mill. The milled material was blended in a bin blender with colloidal silica (1.4 parts by weight), isomalt DC 100 (31.9 parts by weight) and calcium silicate (4.2 parts by weight). The blend was compressed and then film-coated in a coating pan by spraying an aqueous dispersion for film coating (Opadry, available from Colorcon) at a temperature of 60° C.
  • Example 11
  • Component Weight %
    Extrusion
    Ritonavir 3.53
    Lopinavir 14.11
    Copovidone 57.71
    Polyoxyl 40 hydrogenated castor oil 8.47
    (Cremophor RH 40)
    Colloidal silicon dioxide 1.28
    Post extrusion
    Lactose 5.88
    Crospovidone 5.88
    Magnesium stearate 0.49
    Colloidal silicon dioxide 0.55
    Film coating 2.12
  • The extruded material was milled, compressed with tableting excipients, and coated. The formulation consisted of lopinavir (200 mg/tablet), ritonavir (50 mg/tablet), copovidone as the carrier polymer, and polyoxyl 40 hydrogenated castor oil as the surfactant. For compression, outer phase excipients were added to the milled extrudate. The surfactant was incorporated prior to extrusion by granulation with a portion of the polymer.
  • Example 12
  • Component Weight %
    Extrusion
    Ritonavir 3.48
    Lopinavir 13.91
    Copovidone 58.06
    Polyoxyl 40 hydrogenated castor oil 1.67
    (Cremophor RH 40)
    Sorbitan monopalmitate (Span 40) 4.67
    PEG 6000 0.83
    Colloidal silicon dioxide 0.84
    Post extrusion
    Isomalt 11.29
    Calcium silicate 1.47
    Sodium stearyl fumarate 0.59
    Sodium lauryl sulfate 0.88
    Colloidal silicon dioxide 0.49
    Film coating 1.81
  • The tablet formulation was compressed from separately extruded lopinavir and ritonavir powder mixtures. The surfactant was incorporated prior to extrusion by granulation with a portion of the polymer.
  • Example 13
  • Component Weight %
    Extrusion
    Ritonavir 4.03
    Lopinavir 16.10
    Copovidone 68.74
    Sorbitan monolaurate (Span 20) 6.76
    Colloidal silicon dioxide 0.97
    Post extrusion
    Sodium stearyl fumarate 0.99
    Colloidal silicon dioxide 0.64
    Film coating 1.77
  • The formulation was prepared by milling the extrudate, mixing with tableting excipients and compressing into tablets. An aqueous, hydroxypropyl methylcellulose based film coating was applied to the compressed tablets to enhance pharmaceutical elegance. The surfactant was incorporated prior to extrusion by granulation with a portion of the polymer.
  • Example 14
  • Component Weight %
    Extrusion
    Ritonavir 3.54
    Lopinavir 14.15
    Copovidone 59.54
    Polyoxyl 40 hydrogenated castor oil 2.56
    (Cremophor RH 40)
    Sorbitan monolaurate (span 20) 4.26
    Colloidal silicon dioxide 0.85
    Post extrusion
    Isomalt 11.28
    Calcium silicate 0.50
    Sodium stearyl fumarate 0.98
    Colloidal silicon dioxide 0.50
    Film coating 1.84
  • The formulation was prepared by milling the extrudate, mixing with tableting excipients and compressing into tablets. An aqueous, hydroxypropyl methylcellulose based film coating was applied to the compressed tablets to enhance pharmaceutical elegance. The surfactant was incorporated prior to extrusion by granulation with a portion of the polymer.
  • Example 15
  • Component Weight %
    Extrusion
    Ritonavir 4.17
    Lopinavir 16.67
    Copovidone 71.17
    Sorbitan monolaurate (span 20) 6.99
    Colloidal silicon dioxide 1.00
  • The formulation was extruded in the shape of a tablet without the additional processing steps of milling, compression and coating. The formulation composition included ritonavir, lopinavir, copovidone, surfactant, and colloidal silicon dioxide with the two formulations differing in the type of surfactant used. The extruded tablet formulation contained sorbitan monolaurate as the surfactant that was incorporated prior to extrusion by granulation with a portion of the polymer.
  • Example 16
  • Component Weight %
    Extrusion
    Ritonavir 4.17
    Lopinavir 16.67
    Copovidone 70.13
    Polyoxyl 40 hydrogenated castor oil 3.02
    (Cremophor RH 40)
    Sorbitan monolaurate (span 20) 5.02
    Colloidal silicon dioxide 1.00
  • The formulation was extruded in the shape of a tablet without the additional processing steps of milling, compression and coating. The formulation composition included ritonavir, lopinavir, copovidone, surfactant, and colloidal silicon dioxide with the two formulations differing in the type of surfactant used. The extruded tablet formulation contained both polyoxyl 40 hydrogenated castor oil and sorbitan monolaurate as the surfactants. The surfactants were incorporated prior to extrusion by granulation with a portion of the polymer.
  • This dosage form was characterized by an excellent stability and, in particular, exhibit high resistance against recrystallization or decomposition of the active ingredient(s). Thus, upon storage for 6 weeks at 40° C. and 75% humidity (e.g., when kept in high density polyethylene (HDPE) bottles without desiccant), the dosage forms according to the present invention did not exhibit any sign of crystallinity (as evidenced by DSC or WAXS analysis) and contained at least about 98% of the initial active ingredient content (as evidenced by HPLC analysis).
  • In vitro dissolution tests were performed on several of the formulation disclosed in the Examples above. The testing method and conditions are shown in the table below.
  • Apparatus: USP Apparatus 2 (paddle)
    Agitation: 75 rpm
    Medium: 0.06 M POE10LE (Polyoxyethylene 10
    Lauryl Ether)
    Temperature: 37° C.
    Profile Times: 15, 30, 60, 90, 120 and 150 minutes with
    medium replacement
    Proposed Specification: Q = 80% in 120 minutes
  • The results are shown below. Table 1 shows the mean % lopinavir releaseded in minutes for the formulations disclosed in Examples 9-10 and 12-16.
  • TABLE 1
    Mean % lopinavir dissolved in minutes.
    Example Mean % Lopinavir Dissolved (minutes)
    No. 15 30 45 60 90 120 150
    9 30.4 56.0 75.1 88.7 100.6 101.1 100.9
    10
    12 21.6 47.3 67.1 82.0 96.0 100.8 101.1
    13 20.6 43.0 61.3 75.4 92.2 98.1 99.2
    13 23.1 47.3 80.0 93.9 98.1 98.8
    14 21.0 47.6 69.9 85.6 98.5 101.1 101.7
    15 36.9 63.0 81.7 93.2 102.0 103.0 103.1
    16 32.1 57.0 74.9 86.5 95.9 99.2 99.6
  • Table 2 shows the mean % ritonavir dissolved in minutes for the formulations disclosed in Examples 9-10 and 12-16.
  • TABLE 2
    Mean % ritonavir dissolved in minutes
    Example Mean % Ritonavir Dissolved (minutes)
    No. 15 30 45 60 90 120 150
    9
    10 76.5 91.1 95.0 96.9
    12 21.8 46.4 65.6 79.8 93.3 98.1 98.3
    13 19.8 41.6 59.4 73.4 90.0 96.2 97.5
    13 23.1 46.0 78.0 92.0 96.3 96.9
    14 21.0 45.4 66.5 82.3 95.1 100.1 98.2
    15 34.4 59.1 76.9 88.0 96.6 67.6 97.7
    16 30.5 54.4 71.7 83.1 92.3 95.4 96.0
  • Therefore, in one embodiment the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 20% to about 30% of lopinavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 20% to about 30% of lopinavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 43% to about 63% of lopinavir is released from about 15 to about 30 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 61.3% to about 81.7% of lopinavir is released from about 30 to about 45 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 75.4% to about 93.2% of lopinavir is released from about 45 to about 60 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 19.8% to about 34.4% of ritonavir is released from about 0 to about 15 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 41.6% to about 76.5% of ritonavir is released from about 15 to about 30 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 59.4% to about 91.1% of ritonavir is released from about 30 to about 45 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile wherein about 73.4% to about 95% of ritonavir is released from about 45 to about 60 minutes using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C. wherein:
    • about 20% to about 30% of lopinavir is released from about 0 to about 15 minutes;
    • about 43% to about 63% of lopinavir is released from about 15 to about 30 minutes;
    • about 61.3% to about 81.7% of lopinavir is released from about 30 to about 45 minutes; and
    • about 75.4% to about 93.2% of lopinavir is released from about 45 to about 60 minutes.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising ritonavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C. wherein:
    • about 19.8% to about 34.4% of ritonavir is released from about 0 to about 15 minutes;
    • about 41.6% to about 76.5% of ritonavir is released from about 15 to about 30 minutes;
    • about 59.4% to about 91.1% of ritonavir is released from about 30 to about 45 minutes; and
    • about 73.4% to about 95% of ritonavir is released from about 45 to about 60 minutes.
  • In one embodiment the present invention provides for example, a pharmaceutical dosage form comprising ritonavir and lopinavir in a therapeutically effective amount, said dosage form providing an in vitro dissolution profile using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C. wherein:
    • about 19.8% to about 34.4% of ritonavir is released and about 20% to about 30% of lopinavir is released from about 0 to about 15 minutes;
    • about 41.6% to about 76.5% of ritonavir and about 43% to about 63% of lopinavir is released from about 15 to about 30 minutes;
    • about 59.4% to about 91.1% of ritonavir and about 61.3% to about 81.7% of lopinavir is released from about 30 to about 45 minutes; and
    • about 73.4% to about 95% of ritonavir and about 75.4% to about 93.2% of lopinavir is released from about 45 to about 60 minutes.
  • In order to understand lopinavir exposure among humans receiving the dosage form of the present invention and currently marketed Kaletra gelatin capsule, probability distributions were constructed from studies described below. It was assumed that the natural logarithms of lopinavir Cmax and AUC∞ followed normal distributions with mean (μ) and variance (σ2) for each formulation. These values were taken from single 400/100 mg lopinavir/ritonavir dose, 4 or 5 period, randomized, open-label cross-over studies in healthy human volunteers under controlled meal conditions (either fasting, moderate-fat, or high-fat). Each study had between 48 and 63 subjects with a washout between periods of at least 7 days. The mean values for lopinavir Cmax and AUC∞ under moderate-fat meal condition were obtained from the central values in a cross-study meta-analysis of bioequivalence as generally known by those having ordinary skill in the art. The variance values for the distribution were obtained from the between-subject variability estimated for the dosage form of the present invention and the currently marketed Kaletra gelatin capsule using the SAS Procedure Mixed as generally known by those having ordinary skill in the art.
  • The probability distributions of lopinavir Cmax and AUC∞ under fasting and high-fat meal conditions were adjusted using the point estimates from Studies C and A described below for the dosage form of the present invention and currently marketed Kaletra gelatin capsule. The variance for each of fasting and high-fat meal conditions were projected according to the magnitude of the variability relative to that of the moderate-fat meal conditions using data from studies A, B and C described below in more detail.
  • The probability density in relation to AUC∞ for each formulation was calculated based on the mean and variance using the following formula:
  • 1 2 π σ - ( log x - μ ) 2 / ( 2 σ 2 ) AUC
  • The probability distribution of lopinavir Cmax was constructed in the same manner.
  • Study A was a single-dose (lopinavir/ritonavir 400/100 mg), five-period, randomized, open-label, pivotal bioavailability study in 63 healthy subjects. The first four periods were conducted according to a complete-crossover design. Subjects were equally randomized to four sequences of Regimens A, B, C and D for Periods 1 through 4. Five subjects from each sequence group who completed Periods 1 through 4 were randomly chosen to participate in Period 5 and received Regimen E. A washout interval of at least 7 days separated the doses of the five study periods. The five regimens were:
    • Regimen A: Three lopinavir/ritonavir 133.3/33.3 mg currently marketed Kaletra gelatin capsules following a moderate-fat breakfast;
    • Regimen B: Three lopinavir/ritonavir 133.3/33.3 mg currently marketed Kaletra gelatin capsules under fasting conditions;
    • Regimen C: Two lopinavir/ritonavir 200/50 mg dosage forms of the present invention following a moderate-fat breakfast;
    • Regimen D: Two lopinavir/ritonavir 200/50 mg dosage forms of the present invention under fasting conditions; and
    • Regimen E: Two lopinavir/ritonavir 200/50 mg dosage forms of the present invention following a high-fat breakfast.
  • Study B was a single-dose (lopinavir/ritonavir 400/100 mg), non-fasting, moderate-fat, open-label, four-period, randomized, complete-crossover, pivotal bioavailability study in 48 healthy subjects. Subjects were randomly assigned in equal numbers to receive one of four sequences of Regimens A, B, C and D defined as follows:
    • Regimen A: Two lopinavir/ritonavir 200/50 mg dosage forms of the present invention (Lot 1);
    • Regimen B: Two lopinavir/ritonavir 200/50 mg dosage forms of the present invention (Lot 2);
    • Regimen C: Two lopinavir/ritonavir 200/50 mg dosage forms of the present invention (Lot 3);
    • Regimen D: Three lopinavir/ritonavir 133.3/33.3 mg currently marketed Kaletra gelatin capsules.
  • The single doses were administered in the morning on Study Day 1 of each period following a moderate-fat breakfast. A washout interval of 7 days separated the doses of the four study periods.
  • Study C was a Phase 1, single-dose, fasting and non-fasting, open-label, randomized, five-period, partial crossover, single-center study in 56 healthy subjects. The currently marketed Kaletra liquid and gelatin capsule formulations were administered to provide a single dose of lopinavir/ritonavir 400/100 mg. Both formulations were given under fasting conditions and following moderate and high-fat meals.
  • It has been discovered that the dosage form of the present invention provides a substantially lower variation in Cmax and AUC∞ from the 5th to the 95th percentiles for lopinavir when administered to a subject whether fed or fasted than the gelatin capsule formulation. That is, the dosage form of the present invention provides a smaller Δ Cmax and Δ AUC∞ from the 5th to the 95th percentiles for lopinavir than the Kaletra gelatin capsule formulation. This is shown both graphically in FIGS. 1 and 2 as well as numerically in Tables 3-5.
  • The dosage form of the present invention also provides a substantially lower variation in Cmax and AUC∞ from the 25th to the 75th percentiles for lopinavir when administered to a subject whether fed or fasted than the gelatin capsule formulation. That is, the dosage form of the present invention provides a smaller Δ Cmax and Δ AUC∞ from the 25th to the 75th percentiles for lopinavir than the Kaletra gelatin capsule formulation. This is shown both graphically in FIGS. 1 and 2 as well as numerically in Tables 3-5.
  • TABLE 3
    Lopinavir bioavailability from Kaletra ® Gelatin
    Capsule v. Claimed Dosage Form under Fasted Conditions.
    AUC∞ Cmax
    Dosage Form Percentile (μg · h/mL) (μg/mL)
    Gelatin Capsule 5 10.6 1.31
    25 26.67 2.698
    50 52.22 4.946
    75 102.2 9.057
    95 268.5 21.52
    Solid Dosage Form 5 33.15 3.051
    25 54.09 4.882
    50 76.02 6.809
    75 106.8 9.379
    95 174.3 15.03
  • TABLE 4
    Lopinavir bioavailability from Kaletra ® Gelatin Capsule v.
    Claimed Dosage Form under Moderate-Fat Meal Conditions.
    AUC∞ Cmax
    Dosage Form Percentile (μg · h/mL) (μg/mL)
    Gelatin Capsule 5 28.43 2.615
    25 52.9 4.433
    50 81.45 6.424
    75 125.41 9.314
    95 233.5 16.316
    Solid Dosage Form 5 46.06 3.829
    25 71.27 5.91
    50 96.54 8.004
    75 130.8 10.89
    95 202.3 16.77
  • TABLE 5
    Lopinavir bioavailability from Kaletra ® Gelatin Capsule v.
    Claimed Dosage Form under High-Fat Meal Conditions.
    AUC∞ Cmax
    Dosage Form Percentile (μg · h/mL) (μg/mL)
    Gelatin Capsule 5 37.56 2.865
    25 68.05 4.882
    50 102.9 7.066
    75 155.5 10.28
    95 287.7 17.47
    Solid Dosage Form 5 57.77 3.302
    25 75.26 5.011
    50 90.46 6.713
    75 108.7 8.993
    95 141.67 13.683
  • For example, it is shown in Table 3 that the Kaletra gelatin capsule formulation provides a Δ AUC∞ of 257.9 μg·h/mL from the 5th to the 95th percentile, and Δ Cmax of 20.21 μg/mL from the 5th to the 95th percentile. In contrast, the dosage form of the present invention provides a Δ AUC∞ of 141.15 μg·h/mL from the 5th to the 95th percentile, and Δ Cmax of 11.98 μg/mL from the 5th to the 95th percentile.
  • In other words, 90% of the study subjects in Table 3 will have a Δ AUC∞ of 257.9 μg·g/mL and Δ Cmax of 20.21 μg/mL upon dosing of the Kaletra gelatin capsule formulation, while 90% of the study subjects will have a α AUC∞ of 141.15 μg·h/mL and Δ Cmax of 11.98 μg/mL upon dosing of dosage form of the present invention.
  • Again, looking at Table 3, this difference is even evident at the 25th to the 75th percentile wherein the Kaletra gelatin capsule formulation provides a Δ AUC∞ of 75.53 μg·h/mL and Δ Cmax of 6.36 μg/mL for 50% of the study subjects. In stark contrast, the dosage form of the present invention provides a Δ AUC∞ of 52.71 μg·h/mL and Δ Cmax of 4.5 μg/mL for 50% of the study subjects.
  • The dosage form of the present invention demonstrates no food effect. The ratio “X” of AUC∞ fed to AUC∞ fasted for lopinavir is calculated using the formula below,
  • AUC ( fed ) AUC ( fasted ) = X .
  • The calculation is performed for each individual member of a study population in a given trial. The mean value is calculated by adding up the “X” values of every subject and then dividing the total by the number of subjects in the trial. When the “X” value is in the range of about 0.7 to about 1.43, it is determined that the dosage form has no food effect. That is, the dosage form will have substantially the same bioavailability whether it is administered on a full or empty stomach.
  • The ratio “Y” of Cmax fed to Cmax fasted for lopinavir is calculated using the formula below,
  • C max ( fed ) C max ( fasted ) = X .
  • The calculation is performed for each individual member of a study population in a given trial. The mean value is calculated by adding up the “Y” values of every subject and then dividing the total by the number of subjects in the trial. When the “Y” value is in the range of about 0.7 to about 1.43, it is determined that the dosage form has no food effect. That is, the dosage form will have substantially the same bioavailability whether it is administered on a full or empty stomach.
  • Table 6 below better illustrates how “X” and “Y” values are calculated from individual members of a study population totaling 20 subjects.
  • TABLE 6
    Fed/Fasted Ratio of Cmax and AUC for individual subjects.
    AUC∞
    Subject Cmax (fed)/Cmax (fasted) (fed)/AUC∞ (fasted)
     1 1.10 0.93
     2 0.86 0.86
     3 0.74 1.25
     4 1.69 2.70
     5 0.89 1.07
     6 1.36 1.25
     7 0.97 1.25
     8 0.77 1.05
     9 1.30 1.77
    10 1.48 2.23
    11 1.12 1.45
    12 0.60 0.67
    13 0.94 0.75
    14 1.48 1.82
    15 1.19 1.32
    16 0.94 0.93
    17 0.41 0.62
    18 0.98 1.49
    19 0.95 1.01
    20 1.05 1.13
    Total number of 20 20
    subjects (N)
    Total value 20.82 25.55
    Mean value (20.82/20) = 1.04 (25.55/20) = 1.28
  • Table 6. shows the mean Cmax value is 1.04 and the mean AUC∞ value is 1.28. These values are both individually within the range of about 0.7 to about 1.43 and show that the dosage form of the present invention has no food effect.
  • In conducting several studies comparing the dosage form of the present invention to the currently marketed Kaletra gelatin capsule formulation it has also been found that the dosage form of the present invention minimizes or eliminates many adverse events. Particularly, it has been found that the dosage form of the present invention minimizes or eliminates gastrointestinal adverse events. Table 7. below compares the number and types of adverse events in terms of percentage of study populations when administered the dosage form of the present invention versus the currently marketed Kaletra gelatin capsule formulation.
  • TABLE 7
    Percentage of study population suffering adverse event by type.
    Presently Currently marketed Kaletra
    Type of claimed dosage form gelatin evapsule formulation
    adverse event (% of study subjects) (% of study subjects)
    Abdominal pain 13 20
    Asthenia 0 23
    Headache 13 23
    Diarrhea 17 50
    Flatulence 4 14
    Nausea 9 23
    Taste Perversion 4 11

Claims (20)

1. A solid pharmaceutical dosage form comprising an undissolved form of lopinavir in a therapeutically effective amount wherein when said dosage form is dissolved in vitro using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POEIOLE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C., 20% to 30% of lopinavir in said dosage form is released from 0 to 15 minutes, or 43% to 63% of lopinavir in said dosage form is released from 15 to 30 minutes, or 61.3% to 81.7% of lopinavir in said dosage form is released from 30 to 45 minutes, or 75.4% to 93.2% of lopinavir in said dosage form is released from 45 to 60 minutes.
2. The dosage form of claim 1, wherein said undissolved form is an amorphous form.
3. The dosage form of claim 1, wherein upon administration of said dosage form to each member of a study population, the mean of lopinavir AUC∞ (fed) over lopinavir AUC∞ (fasted) ratios for all members of the study population is from 0.7 to 1.43.
4. The dosage form of claim 1, wherein upon administration of said dosage form to each member of a study population, the mean of lopinavir Cmax (fed) over lopinavir Cmax (fasted) ratios for all members of the study population is from 0.7 to 1.43.
5. The dosage form of claim 1, wherein upon administration of said dosage form to each member of a study population under fasting conditions, the 5th percentile of lopinavir AUC∞ in the study population is greater than 30 μg·h/mL; or the mean of lopinavir AUC∞ in the study population is between 60 μg·h/mL and 95 μg·h/mL.
6. The dosage form of claim 1, wherein upon administration of said dosage form to each member of a study population under fed conditions, the 5th percentile of lopinavir AUC∞ in the study population is greater than 50 μg·h/mL.
7. The dosage form of claim 1, wherein upon administration of said dosage form to each member of a study population under fasting conditions, the 5th percentile of lopinavir Cmax in the study population is greater than 2.5 μg/mL.
8. A solid pharmaceutical dosage form comprising a solid dispersion or solid solution of lopinavir in a matrix, said matrix comprising at least one pharmaceutically acceptable water-soluble polymer and at least one pharmaceutically acceptable surfactant, wherein when said dosage form is dissolved in vitro using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POEIOLE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C., 20% to 30% of lopinavir in said dosage form is released from 0 to 15 minutes, or 43% to 63% of lopinavir in said dosage form is released from 15 to 30 minutes, or 61.3% to 81.7% of lopinavir in said dosage form is released from 30 to 45 minutes, or 75.4% to 93.2% of lopinavir in said dosage form is released from 45 to 60 minutes.
9. The dosage form of claim 8, wherein said surfactant having an HLB value of from 4 to 10.
10. The dosage form of claim 8, wherein each of said at least one pharmaceutically acceptable water-soluble polymer has a Tg of at least 50° C., and each of said at least one pharmaceutically acceptable surfactant has an HLB value of from 4 to 10.
11. The dosage form of claim 10, wherein said at least one pharmaceutically acceptable surfactant accounts for at least 50% by weight of the total amount of all surfactants comprised in said dosage form.
12. The dosage form of claim 10, wherein said at least one pharmaceutically acceptable water-soluble polymer comprises a copolymer of N-vinyl pyrrolidone and vinyl acetate, and said at least one pharmaceutically acceptable surfactant comprises a sorbitan mono fatty acid ester.
13. The dosage form of claim 10, wherein said dosage form comprises from 50 to 85% by weight of the dosage form of said at least one pharmaceutically acceptable water-soluble polymer, and from 2 to 20% by weight of the dosage form of said at least one pharmaceutically acceptable surfactant.
14. The dosage form of claim 13, wherein said at least one pharmaceutically acceptable water-soluble polymer is copovidone, and said at least one pharmaceutically acceptable surfactant is sorbitan monolaurate.
15. The dosage form of claim 14, wherein said dosage form further comprises ritonavir, and said solid dispersion or solid solution is a solid solution.
16. The dosage form of claim 8, wherein upon administration of said dosage form to each member of a study population under fasting conditions, the 5th percentile of lopinavir AUC∞ in the study population is greater than 30 μg·h/mL; or the mean of lopinavir AUC∞ in the study population is between 60 μg·h/mL and 95 μg·h/mL.
17. The dosage form of claim 8, wherein upon administration of said dosage form to each member of a study population under fed conditions, the 5th percentile of lopinavir AUC∞ in the study population is greater than 50 μg·h/mL.
18. A solid pharmaceutical dosage form comprising an undissolved form of lopinavir or a solid dispersion or solid solution of lopinavir in a matrix, said matrix comprising at least one pharmaceutically acceptable water-soluble polymer and at least one pharmaceutically acceptable surfactant, wherein upon administration of said dosage form to each member of a study population under fasting conditions, the 5th percentile of lopinavir AUC∞ in the study population is greater than 30 μg·h/mL; or the mean of lopinavir AUC∞ in the study population is between 60 μg·h/mL and 95 μg·h/mL.
19. A solid pharmaceutical dosage form comprising an undissolved form of ritonavir or a solid dispersion or solid solution of ritonavir in a matrix, said matrix comprising at least one pharmaceutically acceptable water-soluble polymer and at least one pharmaceutically acceptable surfactant, wherein when said dosage form is dissolved in vitro using a USP apparatus 2 (paddle) at 75 rpm with a 0.06M POE10LE (Polyoxyethylene 10 Lauryl Ether) medium at 37° C., from 19.8% to 34.4% of ritonavir in said dosage form is released from 0 to 15 minutes, or from 41.6% to 76.5% of ritonavir in said dosage form is released from 15 to 30 minutes, or from 59.4% to 91.1% of ritonavir in said dosage form is released from 30 to 45 minutes, or from 73.4% to 95% of ritonavir in said dosage form is released from 45 to 60 minutes; or wherein upon administration of said dosage form to each member of a study population under fasting conditions, the 5th percentile of ritonavir AUC∞ in the study population is greater than 30 μg·h/mL; or the mean of ritonavir AUC∞ in the study population is between 60 μg·h/mL and 95 μg·h/mL.
20. The dosage form of claim 19, wherein said dosage form comprises said solid dispersion or solution, and said dosage form comprises from 50 to 85% by weight of the dosage form of said at least one pharmaceutically acceptable water-soluble polymer, and each of at least one pharmaceutically acceptable water-soluble polymer has a Tg of at least 50° C.
US12/190,252 2003-08-28 2008-08-12 Solid Pharmaceutical Dosage Formulation Abandoned US20080299203A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/190,252 US20080299203A1 (en) 2003-08-28 2008-08-12 Solid Pharmaceutical Dosage Formulation

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US49841203P 2003-08-28 2003-08-28
US10/925,442 US8025899B2 (en) 2003-08-28 2004-08-25 Solid pharmaceutical dosage form
US11/064,467 US8377952B2 (en) 2003-08-28 2005-02-23 Solid pharmaceutical dosage formulation
US12/190,252 US20080299203A1 (en) 2003-08-28 2008-08-12 Solid Pharmaceutical Dosage Formulation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/064,467 Continuation US8377952B2 (en) 2003-08-28 2005-02-23 Solid pharmaceutical dosage formulation

Publications (1)

Publication Number Publication Date
US20080299203A1 true US20080299203A1 (en) 2008-12-04

Family

ID=36678380

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/064,467 Expired - Fee Related US8377952B2 (en) 2003-08-28 2005-02-23 Solid pharmaceutical dosage formulation
US12/190,252 Abandoned US20080299203A1 (en) 2003-08-28 2008-08-12 Solid Pharmaceutical Dosage Formulation
US13/608,482 Abandoned US20130004578A1 (en) 2005-02-23 2012-09-10 Solid Pharmaceutical Dosage Formulation

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/064,467 Expired - Fee Related US8377952B2 (en) 2003-08-28 2005-02-23 Solid pharmaceutical dosage formulation

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/608,482 Abandoned US20130004578A1 (en) 2005-02-23 2012-09-10 Solid Pharmaceutical Dosage Formulation

Country Status (38)

Country Link
US (3) US8377952B2 (en)
EP (4) EP2206500B1 (en)
JP (1) JP5087409B2 (en)
KR (1) KR101429024B1 (en)
CN (1) CN101163479B (en)
AR (2) AR055734A1 (en)
AU (1) AU2006216856B2 (en)
BR (2) BR122012031169A2 (en)
CA (2) CA2821046C (en)
CL (3) CL2009001844A1 (en)
CR (1) CR9353A (en)
CY (2) CY1120377T1 (en)
DK (2) DK2283844T3 (en)
DO (2) DOP2006000050A (en)
EA (1) EA014446B1 (en)
ES (2) ES2694073T3 (en)
GE (1) GEP20105083B (en)
GT (2) GT200600295A (en)
HR (2) HRP20180999T1 (en)
HU (2) HUE041591T2 (en)
IL (1) IL185390A (en)
LT (2) LT2206500T (en)
MX (1) MX2007010275A (en)
MY (1) MY146247A (en)
NO (1) NO20074807L (en)
NZ (2) NZ560829A (en)
PE (2) PE20061016A1 (en)
PL (2) PL2283844T3 (en)
PT (2) PT2283844T (en)
RS (2) RS57938B1 (en)
SI (2) SI2283844T1 (en)
SV (1) SV2007002427A (en)
TR (2) TR201809084T4 (en)
TW (1) TWI381840B (en)
UA (1) UA89220C2 (en)
UY (2) UY29391A1 (en)
WO (1) WO2006091529A2 (en)
ZA (1) ZA200707022B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8691878B2 (en) 2003-08-28 2014-04-08 Abbvie Inc. Solid pharmaceutical dosage form

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1175205B1 (en) * 1999-11-12 2006-06-14 Abbott Laboratories Solid dispersion comprising ritonavir, fenofibrate or griseofulvin
US8377952B2 (en) * 2003-08-28 2013-02-19 Abbott Laboratories Solid pharmaceutical dosage formulation
US20050048112A1 (en) 2003-08-28 2005-03-03 Jorg Breitenbach Solid pharmaceutical dosage form
CA2568378C (en) * 2004-05-28 2013-03-19 Abbott Gmbh & Co. Kg Formulation obtained from a powder mixture comprising an inorganic pigment
AR045841A1 (en) * 2004-09-28 2005-11-16 Richmond Sa Com Ind Y Financie A SOLID PHARMACEUTICAL COMPOSITION THAT INCLUDES THE TIAZOLIL METHYL ESTER OF THE ACID [5S- (5R *, 8R *, 10R *, 11R *)] -10- HYDROXY-2-METHYL-5- (1-METHYTILE) -1- [2 - (1-METHYLE) -4-TIAZOLIL] -3,6-DIOXO-8,11-BIS (PHENYLMETIL) -2,4,7,12- TETRAAZATRIDECAN-13-OICO AND A PROCEDURE TO PREPARE IT.
US10532028B2 (en) * 2005-07-28 2020-01-14 Isp Investments Llc Method to improve characteristics of spray dried powders and granulated materials, and the products thereby produced
CL2007002331A1 (en) * 2006-08-10 2008-04-18 Cipla Ltd SOLID ORAL COMPOSITION THAT INCLUDES ONE OR MORE ANTIRRETROVIRAL PHARMACOS AND AT LEAST AN INSOLUBLE POLYMER IN WATER, IN RELATIONSHIP APPROXIMATELY 1: 1 TO 1: 6; ELABORATION PROCESS; AND USE FOR HIV TREATMENT.
US20090281132A1 (en) * 2006-09-04 2009-11-12 Kiran Kumar Narsaiah Velaveni Pharmaceutical formulation for use in hiv therapy
US20080085310A1 (en) * 2006-10-06 2008-04-10 Maria Oksana Bachynsky Capecitabine rapidly disintegrating tablets
WO2008067164A2 (en) * 2006-11-15 2008-06-05 Abbott Laboratories Solid pharmaceutical dosage formulations
WO2008076780A2 (en) * 2006-12-14 2008-06-26 Isp Investments Inc. Amorphous valsartan and the production thereof
WO2008080037A2 (en) * 2006-12-21 2008-07-03 Isp Investments Inc. Carotenoids of enhanced bioavailability
WO2008092046A2 (en) * 2007-01-26 2008-07-31 Isp Investments Inc. Amorphous oxcarbazepine and the production thereof
WO2008092057A2 (en) * 2007-01-26 2008-07-31 Isp Investments Inc. Formulation process method to produce spray dried products
US20100303901A1 (en) * 2007-04-26 2010-12-02 Eyal Shimoni Oral delivery of proteins and peptides
EP2188265A1 (en) * 2007-08-07 2010-05-26 Ranbaxy Laboratories Limited Process for preparation of amorphous lopinavir
US20100285115A1 (en) * 2007-10-29 2010-11-11 Cipla Limited Novel Antiretroviral Combination
WO2009084036A2 (en) * 2007-12-20 2009-07-09 Matrix Laboratories Limited Composition for treatment of viral infections
PA8809601A1 (en) * 2007-12-24 2009-07-23 Cipla Ltd ANTI-RETROVIRAL COMBINATION
EP3272337A3 (en) * 2008-02-28 2018-02-14 AbbVie Inc. Tablets
US20100222315A1 (en) * 2008-09-24 2010-09-02 Harbor BioSciencs, Inc. Patient populations and treatment methods
TWI471321B (en) 2009-06-08 2015-02-01 Abbott Gmbh & Co Kg Pharmaceutical dosage form for oral administration of a bcl-2 family inhibitor
WO2011013110A1 (en) * 2009-07-31 2011-02-03 Ranbaxy Laboratories Limited Unit dosage forms of hiv protease inhibitors
US20110034489A1 (en) 2009-07-31 2011-02-10 Ranbaxy Laboratories Limited Solid dosage forms of hiv protease inhibitors
US20130195978A1 (en) * 2010-05-10 2013-08-01 Hetero Research Foundation Darunavir Compositions
UA113500C2 (en) 2010-10-29 2017-02-10 MEL EXTRUSION SOLID DISPERSIONS CONTAINING AN APOPTOSIS-INDUCING AGENT
EP2564832A1 (en) * 2011-08-29 2013-03-06 Hexal AG Solid dosage form of HIV protease inhibitors
KR101794032B1 (en) * 2011-09-21 2017-11-07 (주)바이오시네틱스 Method for preparing nanoparticles
JP6314131B2 (en) * 2012-05-03 2018-04-18 シプラ・リミテッド Antiretroviral composition
CN103655571B (en) * 2012-09-11 2016-04-20 上海星泰医药科技有限公司 A kind of Lopinavir and ritonavir compound recipe high evenness nanometer are divided into prose style free from parallelism and preparation method thereof
US20140234415A1 (en) * 2013-02-20 2014-08-21 Abbvie Inc. Tablet Dosage Forms
US10034865B2 (en) 2015-09-10 2018-07-31 Kashiv Pharma, Llc Surfactant-free HIV protease inhibitor composition and method of manufacturing thereof
RU2619840C1 (en) * 2016-09-21 2017-05-18 Общество с ограниченной ответственностью "Изварино Фарма" Pharmaceutical composition for hiv infection treatment
CN108186578A (en) * 2018-03-27 2018-06-22 聊城大学 A kind of preparation method of Ritonavir solid dispersions
EP3569225A1 (en) 2018-05-18 2019-11-20 Pharmaceutical Oriented Services Ltd Solid dispersion containing ritonavir
GB201808564D0 (en) 2018-05-24 2018-07-11 Douglas Pharmaceuticals Ltd Treatments
EA202191371A1 (en) * 2018-12-14 2021-12-27 Фуджифилм Корпорэйшн PHARMACEUTICAL COMPOSITION AND METHOD FOR ITS OBTAINING
GB201907305D0 (en) * 2019-05-23 2019-07-10 Douglas Pharmaceuticals Ltd Treatment of conditions
CN112263554B (en) * 2020-10-22 2022-09-30 安徽贝克生物制药有限公司 Lopinavir ritonavir compound tablet and preparation method thereof

Citations (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4758427A (en) * 1985-08-08 1988-07-19 Ciba-Geigy Corporation Enhanced absorption of psychoactive 2-aryl-pyrazolo quinolines as a solid molecular dispersion in polyvinylpyrrolidone
US4801460A (en) * 1986-04-11 1989-01-31 Basf Aktiengesellschaft Preparation of solid pharmaceutical forms
US4804699A (en) * 1987-05-15 1989-02-14 Ici Americas Inc. Monomeric and oligomeric glutarate-based light stabilizers for plastics
US4851438A (en) * 1985-08-19 1989-07-25 S. C. Johnson & Son, Inc. Aqueous pyrethroid insecticidal formulations in polyvinyl chloride containers, and methods of producing such formulations that are stable
US4904699A (en) * 1986-12-18 1990-02-27 Bauer Kurt H Nifedipine concentrate stabilized against the influence of light and a process for its preparation
US4996058A (en) * 1987-09-18 1991-02-26 Ciba-Geigy Corporation Covered retard forms
US5405616A (en) * 1992-01-17 1995-04-11 Alfatec Pharma Gmbh Means for containing active substances, having a shell of hydrophilic macromolecules, active substances and process for preparation thereof
US5490990A (en) * 1992-06-25 1996-02-13 Basf Aktiengesellschaft Production of solid pharmaceutical depot forms
US5525628A (en) * 1992-06-12 1996-06-11 Edmond Pharma S.R.L. Salts of a glutathione alkylester and anaminoacids
US5541206A (en) * 1989-05-23 1996-07-30 Abbott Laboratories Retroviral protease inhibiting compounds
US5545628A (en) * 1995-01-10 1996-08-13 Galephar P.R. Inc. Pharmaceutical composition containing fenofibrate
US5610193A (en) * 1993-10-01 1997-03-11 Abbott Laboratories Pharmaceutical composition
US5641516A (en) * 1992-08-13 1997-06-24 Basf Aktiengesellschaft Compositions which contain active substances and are in the form of solid particles
US5648497A (en) * 1989-05-23 1997-07-15 Abbott Laboraotries Retroviral protease inhibiting compounds
US5707648A (en) * 1993-11-17 1998-01-13 Lds Technologies, Inc. Transparent liquid for encapsulated drug delivery
US5725878A (en) * 1993-09-13 1998-03-10 Abbott Laboratories Pharmaceutical composition comprising HIV protease inhibiting compounds
US5727878A (en) * 1996-10-17 1998-03-17 Cdf Corporation Liner for a mixing container and an assembly and method for mixing fluid components
US5741519A (en) * 1995-03-21 1998-04-21 Basf Aktiengesellschaft The production of active substance compositions in the form of a solid solution of the active substance in a polymer matrix, and active substance compositions produced by this process
US5773025A (en) * 1993-09-09 1998-06-30 Edward Mendell Co., Inc. Sustained release heterodisperse hydrogel systems--amorphous drugs
US5776495A (en) * 1994-07-26 1998-07-07 Laboratoires Effik Process for the production of dry pharmaceutical forms and the thus obtained pharmaceutical compositions
US5889051A (en) * 1997-07-15 1999-03-30 Development Center For Biotechnology Stabilization of prostaglandin drug
US5897910A (en) * 1994-12-23 1999-04-27 Basf Aktiengesellschaft Production of covered tablets
US5914332A (en) * 1995-12-13 1999-06-22 Abbott Laboratories Retroviral protease inhibiting compounds
US5939099A (en) * 1995-02-14 1999-08-17 Basf Aktiengesellschaft Solid active extrusion compound preparations containing low-substituted hydroxypropylcellulose
US5945127A (en) * 1995-03-21 1999-08-31 Basf Aktiengesellschaft Storage-stable drug form
US5945123A (en) * 1998-04-02 1999-08-31 K-V Pharmaceutical Company Maximizing effectiveness of substances used to improve health and well being
US6009690A (en) * 1994-12-23 2000-01-04 Basf Aktiengesellschaft Process and apparatus for the production of divisible tablets
US6027747A (en) * 1997-11-11 2000-02-22 Terracol; Didier Process for the production of dry pharmaceutical forms and the thus obtained pharmaceutical compositions
US6037157A (en) * 1995-06-29 2000-03-14 Abbott Laboratories Method for improving pharmacokinetics
US6042847A (en) * 1995-05-19 2000-03-28 Lek, Tovarna Farmacevtskih In Kemicnih Izdelkov, D.D. Three-phase pharmaceutical form with constant and controlled release of amorphous active ingredient for single daily application
US6045829A (en) * 1997-02-13 2000-04-04 Elan Pharma International Limited Nanocrystalline formulations of human immunodeficiency virus (HIV) protease inhibitors using cellulosic surface stabilizers
US6063821A (en) * 1995-03-21 2000-05-16 Basf Aktiengesellschaft Transparent rapid release compositions of non-steroidal analgesics
US6066334A (en) * 1997-03-10 2000-05-23 Basf Aktiengesellschaft Use of redispersible polymer powders or polymer granules as binders for producing solid pharmaceutical presentations
US6083518A (en) * 1995-06-20 2000-07-04 Bioglan Ab Composition comprising an active agent dissolved in a glass-forming carrier and a process for the preparation thereof
US6187342B1 (en) * 1995-09-29 2001-02-13 Basf Aktiengesellschaft Solid medicaments obtained by extrusion of an isomalt-containing polymer-active substance melt
US6190781B1 (en) * 1998-01-20 2001-02-20 Mitsubishi Paper Mills Limited Support for imaging material
US6221368B1 (en) * 1996-09-13 2001-04-24 Basf Aktiengesellschaft Process for producing solid dosage forms by extrusion
US6232333B1 (en) * 1996-11-21 2001-05-15 Abbott Laboratories Pharmaceutical composition
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6251434B1 (en) * 1996-01-23 2001-06-26 Basf Aktiengesellschaft Preparations of non-steroidal analgesics
US20010006650A1 (en) * 1997-09-19 2001-07-05 Beth A. Burnside Solid solution beadlet
US6274727B1 (en) * 1996-11-15 2001-08-14 Merck Patent Gesellschaft Mit Beschrankter Haftung Method for producing shaped and unshaped polyol masses
US6281282B1 (en) * 1996-05-03 2001-08-28 Basf Aktiengesellschaft Polymer powders redispersible in aqueous solution
US20020006443A1 (en) * 1999-12-23 2002-01-17 Curatolo William J. Pharmaceutical compositions providing enhanced drug concentrations
US20020015814A1 (en) * 1998-10-07 2002-02-07 Weinstein Larry J. Pre-cut fibrous insulation for custom fitting building cavities of different widths
US6350398B1 (en) * 1998-09-03 2002-02-26 Basf Aktiengesellschaft Process for producing coated solid dosage forms
US6372905B1 (en) * 2000-08-31 2002-04-16 Abbott Laboratories Processes and intermediates for preparing retroviral protease inhibitors
US6383471B1 (en) * 1999-04-06 2002-05-07 Lipocine, Inc. Compositions and methods for improved delivery of ionizable hydrophobic therapeutic agents
US6387401B2 (en) * 1995-08-25 2002-05-14 Basf Aktiengesellschaft Use of lipids as adjuvents in the production of solid medicinal forms by the melt extrusion process
US6423256B1 (en) * 1998-10-15 2002-07-23 Basf Aktiengesellschaft Process for producing solid dosage forms
US6436440B1 (en) * 1998-04-02 2002-08-20 Basf Aktiengesellschaft Use of N-vinyllactam-or-N-vinylamine-containing copolymers as matrix for producing solid pharmaceutical and cosmetic presentations
US20020114833A1 (en) * 2000-12-06 2002-08-22 Abu-Izza Khawla A. Fast dissolving tablet
US6440946B1 (en) * 1999-02-25 2002-08-27 Takeda Chemical Industries, Ltd. Multiple-agents-binding compound, production and use thereof
US20030021840A1 (en) * 2001-05-03 2003-01-30 Infeld Martin Howard High dose solid unit oral pharmaceutical dosage form of amorphous nelfinavir mesylate and process for making same
US20030039686A1 (en) * 2001-06-06 2003-02-27 Karsten Maeder Pharmaceutical compositions having depressed melting points
US20030054038A1 (en) * 2001-06-22 2003-03-20 Crew Marshall D. Pharmaceutical compositions of drugs and neutralized acidic polymers
US6547997B1 (en) * 1997-11-28 2003-04-15 Abbot Laboratories Method for producing solvent-free noncrystalline biologically active substances
US20030072801A1 (en) * 2001-06-22 2003-04-17 Pfizer Inc. Pharmaceutical compositions comprising drug and concentration-enhancing polymers
US20030091630A1 (en) * 2001-10-25 2003-05-15 Jenny Louie-Helm Formulation of an erodible, gastric retentive oral dosage form using in vitro disintegration test data
US20030091643A1 (en) * 2001-06-22 2003-05-15 Friesen Dwayne T. Pharmaceutical compositions of dispersions of drugs and neutral polymers
US20030096791A1 (en) * 2001-05-31 2003-05-22 Cima Labs Inc. Taste masking of highly water-soluble drugs
US20030104063A1 (en) * 2001-06-22 2003-06-05 Babcock Walter C. Pharmaceutical compositions of dispersions of amorphous drugs mixed with polymers
US6579521B2 (en) * 2000-10-20 2003-06-17 Chiron Corporation Methods of therapy for HIV infection
US6599528B1 (en) * 1999-03-25 2003-07-29 Abbott Gmbh & Co. Kg Mechanically stable pharmaceutical presentations form containing liquid or semisolid surface-active substances
US6599931B1 (en) * 1998-09-18 2003-07-29 Abbott Gmbh & Co. Kg Test system for characterizing the compatibility of bioactive substances and polyvinylpyrrolidone
US20030153608A1 (en) * 2000-03-17 2003-08-14 Markus Maegerlein Torasemide-containing pharmaceutical preparations
US6608198B2 (en) * 2000-03-30 2003-08-19 Abbott Laboratories Crystalline pharmaceutical
US20030161884A1 (en) * 2000-05-30 2003-08-28 Jorg Rosenberg Formulation based on heparin, glycosaminoglycan or heparinoid, use of the formulation and the formulation base
US20040001888A1 (en) * 2002-06-26 2004-01-01 Biopharm Solutions Inc. Solid dosage forms for rapid dissolution of poorly soluble drugs
US20040003004A1 (en) * 2002-06-28 2004-01-01 Microsoft Corporation Time-bound database tuning
US20040014817A1 (en) * 2000-09-19 2004-01-22 Joerg Rosenberg Stable dosage forms containing ubiquinones
US20040013697A1 (en) * 2000-05-30 2004-01-22 Gunther Berndl Self-emulsifying active substance formulation and use of this formulation
US20040013734A1 (en) * 1999-02-10 2004-01-22 Pfizer Inc. Pharmaceutical solid dispersions
US20040024031A1 (en) * 2002-05-03 2004-02-05 Transform Pharmaceuticals, Inc. Solvates and polymorphs of ritonavir and methods of making and using the same
US20040029892A1 (en) * 2000-08-03 2004-02-12 Joerg Rosenberg Compositions and dosage forms for application in the oral cavity in the treatment of mykoses
US20040062802A1 (en) * 1998-04-02 2004-04-01 Hermelin Victor M. Maximizing effectiveness of substances used to improve health and well being
US20040091529A1 (en) * 2002-06-26 2004-05-13 David Edgren Methods and dosage forms for increasing solubility of drug compositions for controlled delivery
US6737005B1 (en) * 1998-11-23 2004-05-18 Abbott Gmbh & Co. Kg Method of producing solid dosage forms
US20040096499A1 (en) * 2002-08-05 2004-05-20 Navin Vaya Novel dosage form
US6763607B2 (en) * 2002-02-01 2004-07-20 Pfizer Inc. Method for making homogeneous spray-dried solid amorphous drug dispersions utilizing modified spray-drying apparatus
US20050008706A1 (en) * 2001-07-06 2005-01-13 Per Holm Controlled agglomeration
US20050014304A1 (en) * 2000-01-25 2005-01-20 Kook-Chul Moon Low temperature polycrystalline silicon type thin film transistor and a method of the thin film transistor fabrication
US20050025791A1 (en) * 2002-06-21 2005-02-03 Julius Remenar Pharmaceutical compositions with improved dissolution
US20050031691A1 (en) * 2002-09-11 2005-02-10 Elan Pharma International Ltd. Gel stabilized nanoparticulate active agent compositions
US20050048112A1 (en) * 2003-08-28 2005-03-03 Jorg Breitenbach Solid pharmaceutical dosage form
US20050079138A1 (en) * 2002-12-19 2005-04-14 Chickering Donald E. Methods for making pharmaceutical formulations comprising microparticles with improved dispersibility, suspendability or wettability
US20050084529A1 (en) * 2003-08-28 2005-04-21 Joerg Rosenberg Solid pharmaceutical dosage form
US20050100586A1 (en) * 1999-05-19 2005-05-12 Michel Sournac Transdermal device for administering testosterone or one of the derivatives thereof
US6894171B1 (en) * 1998-07-20 2005-05-17 Abbott Laboratories Polymorph of a pharmaceutical
US20050143404A1 (en) * 2003-08-28 2005-06-30 Joerg Rosenberg Solid pharmaceutical dosage formulation
US20060003942A1 (en) * 2003-10-27 2006-01-05 Roger Tung Combinations for HCV treatment
US7014810B2 (en) * 1999-12-15 2006-03-21 Abbott Gmbh & Co. Kg Method of producing solid shape forms containing an active ingredient
US20060115539A1 (en) * 2003-06-07 2006-06-01 Armin Prasch Micropellets method for the production thereof, and use thereof
US7229641B2 (en) * 2000-07-05 2007-06-12 Capricorn Pharma, Inc. Rapid-melt compositions methods of making same and methods of using same
US7364752B1 (en) * 1999-11-12 2008-04-29 Abbott Laboratories Solid dispersion pharamaceutical formulations

Family Cites Families (310)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE973095C (en) 1955-01-19 1959-12-03 Kalle & Co Ag Process for converting oils into powders that can be sprinkled by spray-drying oil-in-water emulsions
HU176101B (en) 1977-12-29 1980-12-28 Gyogyszerkutato Intezet Process for producing bis-bracket-nitrozo-ureido-bracket closed-polyol derivatives
DE3013839A1 (en) 1979-04-13 1980-10-30 Freunt Ind Co Ltd METHOD FOR PRODUCING AN ACTIVATED PHARMACEUTICAL COMPOSITION
CA1146866A (en) 1979-07-05 1983-05-24 Yamanouchi Pharmaceutical Co. Ltd. Process for the production of sustained release pharmaceutical composition of solid medical material
FR2525108B1 (en) 1982-04-19 1989-05-12 Elan Corp Ltd HIGH-SOLUBILITY MEDICINES AND PROCESS FOR OBTAINING THEM
US4629621A (en) * 1984-07-23 1986-12-16 Zetachron, Inc. Erodible matrix for sustained release bioactive composition
JPS61205208A (en) 1985-03-08 1986-09-11 Yamanouchi Pharmaceut Co Ltd Rapidly solubilizing preparation for hard-soluble medicines
US4590065A (en) 1985-04-18 1986-05-20 Colgate-Palmolive Company Stable flavor-containing dentifrice
DE3612211A1 (en) 1986-04-11 1987-10-15 Basf Ag CONTINUOUS TABLET METHOD
WO1989005138A1 (en) 1987-12-08 1989-06-15 Mark Chasin Method of forming bioerodible implants for improved controlled drug release
DE3812567A1 (en) 1988-04-15 1989-10-26 Basf Ag METHOD FOR PRODUCING PHARMACEUTICAL MIXTURES
DE3830353A1 (en) 1988-09-07 1990-03-15 Basf Ag METHOD FOR THE CONTINUOUS PRODUCTION OF SOLID PHARMACEUTICAL FORMS
US5368864A (en) 1988-11-25 1994-11-29 Henning Berlin Gmbh Chemie- Und Pharmawerk Formulation of oxypurinol and/or its alkali and alkaline earth salts
GB8903328D0 (en) 1989-02-14 1989-04-05 Ethical Pharma Ltd Nifedipine-containing pharmaceutical compositions and process for the preparation thereof
HU204529B (en) 1989-08-10 1992-01-28 Richter Gedeon Vegyeszet Process for producing new 1-oxa-2-oxo-8-aza-spiro(4,5)decane derivatives and pharmaceutical compositions containing them
US5075291A (en) 1989-11-22 1991-12-24 Ici Americas Inc. Crystalline sugar alcohol containing uniformly dispersed particulate pharmaceutical compound
JPH07501259A (en) 1990-06-01 1995-02-09 リサーチ コーポレイション テクノロジーズ,インコーポレイテッド self-emulsifying glass
SE9003296L (en) 1990-10-16 1992-04-17 Kabi Pharmacia Ab PROCEDURE SHOULD FORMULATE MEDICINAL PRODUCTS
KR0166096B1 (en) 1990-11-28 1999-01-15 우에하라 아키라 6-o-methylerythromycin-a derivative
ATE159426T1 (en) 1991-04-16 1997-11-15 Nippon Shinyaku Co Ltd METHOD FOR PRODUCING A SOLID DISPERSION
WO1993007859A1 (en) 1991-10-23 1993-04-29 Warner-Lambert Company Novel pharmaceutical pellets and process for their production
DE4138513A1 (en) 1991-11-23 1993-05-27 Basf Ag SOLID PHARMACEUTICAL RETARD FORM
DK0617612T3 (en) * 1991-12-18 1998-04-14 Warner Lambert Co Process for preparing a solid dispersion
DE4200821A1 (en) 1992-01-15 1993-07-22 Bayer Ag TASTE-MASKED PHARMACEUTICAL AGENTS
EP0626850B1 (en) 1992-02-18 2002-05-15 Pharmos Corporation Dry compositions for preparing submicron emulsions
US5654003A (en) 1992-03-05 1997-08-05 Fuisz Technologies Ltd. Process and apparatus for making tablets and tablets made therefrom
ZA932268B (en) 1992-03-30 1993-11-11 Alza Corp Polymer system containing a partially soluble compound
CA2095776C (en) 1992-05-12 2007-07-10 Richard C. Fuisz Rapidly dispersable compositions containing polydextrose
US5968942A (en) 1992-08-25 1999-10-19 G. D. Searle & Co. α- and β-amino acid hydroxyethylamino sulfonamides useful as retroviral protease inhibitors
IS2334B (en) 1992-09-08 2008-02-15 Vertex Pharmaceuticals Inc., (A Massachusetts Corporation) Aspartyl protease inhibitor of a new class of sulfonamides
US5484926A (en) * 1993-10-07 1996-01-16 Agouron Pharmaceuticals, Inc. HIV protease inhibitors
GB9306887D0 (en) 1993-04-01 1993-05-26 Graham Neil B Random block copolymers
WO1995005809A1 (en) 1993-08-20 1995-03-02 Nippon Shinyaku Co., Ltd. Gastric remaining preparation, swollen molding, and production process
KR100354702B1 (en) 1993-11-23 2002-12-28 유로-셀티크 소시에떼 아노뉨 Manufacturing method and sustained release composition of pharmaceutical composition
IL111991A (en) 1994-01-28 2000-07-26 Abbott Lab Liquid pharmaceutical composition of HIV protease inhibitors in organic solvent
US6787156B1 (en) 1994-02-23 2004-09-07 Bm Research A/S Controlled release composition
DE4413350A1 (en) 1994-04-18 1995-10-19 Basf Ag Retard matrix pellets and process for their production
IL129871A (en) 1994-05-06 2003-11-23 Pharmacia & Upjohn Inc Process for preparing 4-phenyl-substituted octanoyl-oxazolidin-2-one intermediates that are useful for preparing pyran-2-ones useful for treating retroviral infections
US5443178A (en) 1994-06-23 1995-08-22 Allergan, Inc. Tablet dispensing system
US6113941A (en) 1994-09-30 2000-09-05 Takeda Chemical Industries, Ltd. Substained release microcapsule of physiologically active compound which is slightly water soluble at pH 6 to 8
US5965161A (en) 1994-11-04 1999-10-12 Euro-Celtique, S.A. Extruded multi-particulates
DE4446467A1 (en) 1994-12-23 1996-06-27 Basf Ag Process for the production of lenticular tablets by melt calendering
DE19500977C2 (en) 1995-01-14 1999-01-07 Lohmann Therapie Syst Lts Solid drug form with active ingredient distributed in polymeric material
NZ270439A (en) 1995-02-02 1996-04-26 Bernard Charles Sherman Solid slow release pharmaceutical composition: carrier is polyethylene glycol and hydrophilic gel-forming polymer
AU706195B2 (en) 1995-04-14 1999-06-10 Inhale Therapeutic Systems Powdered pharmaceutical formulations having improved dispersibility
US5567823A (en) 1995-06-06 1996-10-22 Abbott Laboratories Process for the preparation of an HIV protease inhibiting compound
BE1009856A5 (en) * 1995-07-14 1997-10-07 Sandoz Sa Pharmaceutical composition in the form of a solid release including macrolide and a vehicle.
TW487582B (en) 1995-08-11 2002-05-21 Nissan Chemical Ind Ltd Method for converting sparingly water-soluble medical substance to amorphous state
EP0844870B1 (en) 1995-08-17 2001-11-21 Csir Controlled release products
US6391338B1 (en) 1995-09-07 2002-05-21 Biovail Technologies Ltd. System for rendering substantially non-dissoluble bio-affecting agents bio-available
GB9519363D0 (en) 1995-09-22 1995-11-22 Euro Celtique Sa Pharmaceutical formulation
DE19536387A1 (en) 1995-09-29 1997-04-03 Basf Ag Process for the preparation of vitamin-containing solid preparations
WO1997013503A1 (en) 1995-10-13 1997-04-17 The Penn State Research Foundation Synthesis of drug nanoparticles by spray drying
DE19539361A1 (en) 1995-10-23 1997-04-24 Basf Ag Process for the preparation of multilayer, solid pharmaceutical forms for oral or rectal administration
DE19539574A1 (en) * 1995-10-25 1997-04-30 Boehringer Mannheim Gmbh Preparations and processes for stabilizing biological materials by means of drying processes without freezing
US5858401A (en) * 1996-01-22 1999-01-12 Sidmak Laboratories, Inc. Pharmaceutical composition for cyclosporines
US5762961A (en) 1996-02-09 1998-06-09 Quadrant Holdings Cambridge Ltd. Rapidly soluble oral solid dosage forms, methods of making same, and compositions thereof
US5958455A (en) 1996-02-09 1999-09-28 Quadrant Holdings Cambridge Ltd Oral solid dosage forms, methods of making same and compositions thereof
GB2311027B (en) 1996-03-15 1999-10-27 Johnson & Johnson Medical Coated bioabsorbable beads for wound treatment
US20030064108A1 (en) 1996-04-23 2003-04-03 Stefan Lukas Taste masked pharmaceutical compositions
US5834472A (en) 1996-05-24 1998-11-10 Schering Corporation Antifungal composition with enhanced bioavailability
US6764690B2 (en) 1996-05-29 2004-07-20 Delsitech Oy Dissolvable oxides for biological applications
WO1997046222A1 (en) 1996-06-03 1997-12-11 Merck & Co., Inc. Pediatric formulation for hiv protease inhibitors
US5935936A (en) 1996-06-03 1999-08-10 Genzyme Corporation Compositions comprising cationic amphiphiles and co-lipids for intracellular delivery of therapeutic molecules
DE19624607A1 (en) 1996-06-20 1998-01-02 Basf Ag Process for the preparation of salts of pharmaceutical active substances bearing acid groups
EP2272536B1 (en) 1996-06-26 2016-05-04 Board of Regents, The University of Texas System Hot-melt extrudable Pharmaceutical formulation
DE19629753A1 (en) 1996-07-23 1998-01-29 Basf Ag Process for the production of solid dosage forms
DE19635676A1 (en) 1996-09-03 1998-03-05 Basf Ag Solid foamed active ingredient preparations
DE19637479A1 (en) 1996-09-13 1998-03-19 Basf Ag Preparation of solid pharmaceutical forms
US6649186B1 (en) 1996-09-20 2003-11-18 Ethypharm Effervescent granules and methods for their preparation
US6071539A (en) 1996-09-20 2000-06-06 Ethypharm, Sa Effervescent granules and methods for their preparation
US6488961B1 (en) 1996-09-20 2002-12-03 Ethypharm, Inc. Effervescent granules and methods for their preparation
US8828432B2 (en) 1996-10-28 2014-09-09 General Mills, Inc. Embedding and encapsulation of sensitive components into a matrix to obtain discrete controlled release particles
US20010006677A1 (en) 1996-10-29 2001-07-05 Mcginity James W. Effervescence polymeric film drug delivery system
ZA9710071B (en) 1996-11-21 1998-05-25 Abbott Lab Pharmaceutical composition.
TW486370B (en) * 1996-12-25 2002-05-11 Yamanouchi Pharma Co Ltd Rapidly disintegrable pharmaceutical composition
US6197787B1 (en) * 1997-03-03 2001-03-06 Sanofi-Synthelabo Pharmaceutical formulations containing poorly soluble drug substances
DE19710009A1 (en) 1997-03-12 1998-09-24 Knoll Ag Multi-phase preparation forms containing active ingredients
DE19710213A1 (en) 1997-03-12 1998-09-17 Basf Ag Process for the manufacture of solid combination dosage forms
US5948426A (en) 1997-05-03 1999-09-07 Jefferies; Steven R. Method and article to induce hematopoietic expansion
US6610764B1 (en) 1997-05-12 2003-08-26 Metabolix, Inc. Polyhydroxyalkanoate compositions having controlled degradation rates
DE19721467A1 (en) 1997-05-22 1998-11-26 Basf Ag Process for the preparation of small-scale preparations of biologically active substances
GB9710699D0 (en) * 1997-05-24 1997-07-16 Danbiosyst Uk Gastro-retentive controlled release system
US5955475A (en) 1997-06-30 1999-09-21 Endo Pharmaceuticals Inc. Process for manufacturing paroxetine solid dispersions
JPH1125076A (en) 1997-06-30 1999-01-29 Fujitsu Ltd Document managing device and document management program storage medium
DE19733505A1 (en) 1997-08-01 1999-02-04 Knoll Ag Fast acting analgesic
EP0901786B1 (en) * 1997-08-11 2007-06-13 Pfizer Products Inc. Solid pharmaceutical dispersions with enhanced bioavailability
HN1998000115A (en) 1997-08-21 1999-06-02 Warner Lambert Co SOLID PHARMACEUTICAL DOSAGE FORMS
JP3460538B2 (en) 1997-10-08 2003-10-27 救急薬品工業株式会社 Fast dissolving film preparation
US20020164374A1 (en) * 1997-10-29 2002-11-07 John Jackson Polymeric systems for drug delivery and uses thereof
DE19753298A1 (en) * 1997-12-01 1999-06-02 Basf Ag Process for the preparation of solid dosage forms
US6632455B2 (en) 1997-12-22 2003-10-14 Schering Corporation Molecular dispersion composition with enhanced bioavailability
GB9802088D0 (en) 1998-01-30 1998-03-25 Scherer Ltd R P Pharmaceutical products
US6787157B1 (en) 1998-03-10 2004-09-07 Abbott Laboratories Multiphase active ingredient-containing formulations
WO1999047124A1 (en) 1998-03-16 1999-09-23 Yamanouchi Pharmaceutical Co., Ltd. Tablets quickly disintegrating in the oral cavity and process for producing the same
DE19812688A1 (en) 1998-03-23 1999-09-30 Basf Ag Solid dosage forms useful for slow release of drugs, fragrances, plant-treating agents, animal feed additives and food additives
DE19814739A1 (en) 1998-04-02 1999-10-07 Basf Ag Solubilizing agents useful in pharmaceutical, cosmetic and food compositions
AU763143B2 (en) 1998-04-08 2003-07-17 Kyowa Hakko Kirin Co., Ltd. Tablet manufacturing method and tablet
WO1999055774A1 (en) 1998-04-24 1999-11-04 Eastman Chemical Company Coprecipitation of cellulose esters with functional additives and compositions thus obtainable
JP4027535B2 (en) 1998-05-26 2007-12-26 エーザイ・アール・アンド・ディー・マネジメント株式会社 Powder containing fat-soluble drug
WO1999063841A1 (en) 1998-06-05 1999-12-16 Forbes Medi-Tech Inc. Compositions comprising phytosterol and/or phytostanol having enhanced solubility and dispersability
WO1999063971A1 (en) 1998-06-11 1999-12-16 Em Industries, Inc. Micro-osmotic controlled drug delivery systems
KR100336090B1 (en) 1998-06-27 2002-05-27 윤승원 Solid dispersed preparation of poorly water-soluble drug containing oil, fatty acid or mixture thereof
MY145265A (en) * 1998-07-20 2012-01-13 Abbott Lab Amorphous ritonavir
US6323180B1 (en) 1998-08-10 2001-11-27 Boehringer Ingelheim (Canada) Ltd Hepatitis C inhibitor tri-peptides
US20010021372A1 (en) 1998-08-18 2001-09-13 Tore Omtveit Apparatus having partially gold-plated surface
DE19839276A1 (en) 1998-08-28 2000-03-02 Basf Ag Process for the preparation of solid dosage forms
DE19841244A1 (en) 1998-09-09 2000-03-16 Knoll Ag Method and device for making tablets
DE19842753A1 (en) 1998-09-18 2000-03-23 Bayer Ag Multiple-unit retard oral dosage formulation having controlled release independent of agitation and food effect, containing particles of combination of drug and hydroxypropyl cellulose
DE19843904A1 (en) 1998-09-24 2000-03-30 Basf Ag Solid dosage form for prolonged slow release of e.g. drugs, plant treatment agents, or food or feed additives, containing copolymer of N-vinyl-lactam, methyl methacrylate and further monomer(s) as binder
US20040013613A1 (en) 2001-05-18 2004-01-22 Jain Rajeev A Rapidly disintegrating solid oral dosage form
US6502135B1 (en) 1998-10-30 2002-12-31 Science Applications International Corporation Agile network protocol for secure communications with assured system availability
DE19855440A1 (en) 1998-12-01 2000-06-08 Basf Ag Process for the production of solid dosage forms by melt extrusion
DE19856147A1 (en) 1998-12-04 2000-06-08 Knoll Ag Divisible solid dosage forms and methods for their preparation
DE19856432A1 (en) * 1998-12-08 2000-06-15 Basf Ag Nanoparticulate core-shell systems and their use in pharmaceutical and cosmetic preparations
AT407111B (en) 1998-12-22 2000-12-27 Gergely Dr & Co SUGAR- AND / OR SUGAR-ALCOHOLIC MATRIX MATERIAL AND METHOD FOR THE PRODUCTION THEREOF
JP2002534371A (en) 1999-01-06 2002-10-15 コリア リサーチ インスティテュート オブ ケミカル テクノロジー Method for producing pharmaceutically active ingredient containing water-insoluble drug and pharmaceutical composition for oral administration containing the same
DE19901383A1 (en) 1999-01-15 2000-07-20 Knoll Ag Process for the preparation of different solid dosage forms
US6706283B1 (en) 1999-02-10 2004-03-16 Pfizer Inc Controlled release by extrusion of solid amorphous dispersions of drugs
ES2310164T3 (en) 1999-02-10 2009-01-01 Pfizer Products Inc. LIBERATION DEVICE CONTROLLED BY THE MATRIX.
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
DE19911097A1 (en) 1999-03-12 2000-09-14 Basf Ag Process for the preparation of solid dosage forms containing cyclodextrin
ATE307808T1 (en) * 1999-03-24 2005-11-15 Scherer Technologies Inc R P PHARMACEUTICAL FORMULATIONS WITH IMPROVED SOLUBILITY IN WATER
DE19913606A1 (en) 1999-03-25 2000-09-28 Basf Ag Powdery solubilization aids for solid pharmaceutical dosage forms
US7919119B2 (en) * 1999-05-27 2011-04-05 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US6465011B2 (en) 1999-05-29 2002-10-15 Abbott Laboratories Formulations comprising lipid-regulating agents
NL1012300C2 (en) 1999-06-11 2000-12-12 Rijksuniversiteit Stabilizer for pharmaceuticals.
FR2795326B1 (en) 1999-06-28 2001-08-31 Adir SOLID THERMOFORMABLE PHARMACEUTICAL COMPOSITION WITH CONTROLLED RELEASE
DE19930454A1 (en) 1999-07-02 2001-01-04 Knoll Ag Solid paroxetine-containing preparations
DE19934610A1 (en) 1999-07-23 2001-01-25 Bayer Ag Rapid-release extrudates containing low viscosity hydroxypropylcellulose, useful for formulating plant protecting agents and oral pharmaceutical and veterinary compositions
US6284270B1 (en) 1999-08-04 2001-09-04 Drugtech Corporation Means for creating a mass having structural integrity
SE9903236D0 (en) 1999-09-10 1999-09-10 Astra Ab Method of obtaining microparticles
DE19943501A1 (en) 1999-09-10 2001-03-15 Basf Ag Underwater granulation of melts containing active ingredients
DK1233755T3 (en) 1999-09-29 2007-10-29 Scherer Technologies Inc R P Hydrolyzed cellulose granulations of drug salts
DE19949897A1 (en) 1999-10-15 2001-04-19 Rainer Rogasch Shaped article, e.g. in stick form, for controlled release of drugs in wounds or body cavities, comprising hydrophilic outer phase, lipophilic inner phase and drug, e.g. antibiotic
US6264981B1 (en) 1999-10-27 2001-07-24 Anesta Corporation Oral transmucosal drug dosage using solid solution
US6372259B1 (en) 1999-11-10 2002-04-16 University Of Iowa Research Foundation Palatable, sustained release drug granules
EP1175205B1 (en) 1999-11-12 2006-06-14 Abbott Laboratories Solid dispersion comprising ritonavir, fenofibrate or griseofulvin
PT1227797E (en) 1999-11-12 2005-05-31 Abbott Lab PHARMACEUTICAL FORMULATIONS OF SOLID DISPERSION
DE19958007A1 (en) 1999-12-02 2001-06-07 Roehm Gmbh Injection molding process for (meth) acrylate copolymers with tertiary ammonium groups
DE19961334A1 (en) 1999-12-17 2001-06-21 Roehm Gmbh Injection molding process for neutral and acid group-containing (meth) acrylate copolymers
KR100523127B1 (en) * 1999-12-23 2005-10-20 화이자 프로덕츠 인코포레이티드 Hydrogel-driven drug dosage form
EP2415462A1 (en) 1999-12-23 2012-02-08 Mayne Pharma International Pty Ltd. Improved pharmaceutical compositions for poorly soluble drugs
DE10000792A1 (en) 2000-01-11 2001-07-19 Bernhard C Lippold Formulations of active substances in the form of a solid dispersion
US7029700B2 (en) 2000-01-14 2006-04-18 Brown University Research Foundation Micronized freeze-dried particles
GB0003782D0 (en) * 2000-02-17 2000-04-05 Dumex Ltd As Process
GB0005016D0 (en) 2000-03-01 2000-04-26 Jumik Technologies Limited PVA-Containing compositions
DE60119905T2 (en) 2000-03-09 2007-01-11 Ohio State University Research Foundation, Columbus Process for the preparation of solid dispersions
UY26615A1 (en) 2000-03-16 2001-10-25 Pfizer Prod Inc GLUCOGEN PHOSPHORYLASE INHIBITOR.
DE10015479A1 (en) 2000-03-29 2001-10-11 Basf Ag Solid oral dosage forms with delayed release of active ingredient and high mechanical stability
GB0008785D0 (en) 2000-04-10 2000-05-31 Novartis Ag Organic compounds
DE10021539C1 (en) 2000-05-03 2002-02-28 Henkel Kgaa Device for spray drying solvent-containing compositions
US20020001617A1 (en) * 2000-05-26 2002-01-03 Chang-Hyun Lee Rapidly disintegrating tablet and process for the manufacture thereof
DE10029201A1 (en) 2000-06-19 2001-12-20 Basf Ag Retarded release oral dosage form, obtained by granulating mixture containing active agent and polyvinyl acetate-polyvinyl pyrrolidone mixture below the melting temperature
US20100010101A1 (en) * 2000-07-05 2010-01-14 Capricorn Pharma, Inc. Rapid-Melt Compositions and Methods of Making Same
IT1318618B1 (en) 2000-07-10 2003-08-27 A C R Applied Coating Res S A QUICK RELEASE BIOADHESIVE MICROSPHERES FOR SUBLINGUAL ADMINISTRATION OF ACTIVE INGREDIENTS.
WO2002005788A1 (en) 2000-07-14 2002-01-24 Universiteit Gent Composite solid shaped articles for the controlled delivery of biologically active ingredients
WO2002013792A1 (en) 2000-08-11 2002-02-21 Eisai Co., Ltd. Drug-containing solid dispersion having improved solubility
FR2814366A1 (en) * 2000-09-22 2002-03-29 Rhodia Chimie Sa PROCESS FOR GRANULATING ACTIVE MATERIALS BY LOW PRESSURE EXTRUSION FOR OBTAINING DIRECTLY COMPRESSIBLE GRANULES
US20040013736A1 (en) * 2000-09-25 2004-01-22 Tomio Nakano Process for producing medicinal solid dispersion
WO2002026372A2 (en) 2000-09-27 2002-04-04 Verion Inc. Instant water dissolvable encapsulate and process
WO2002035991A2 (en) 2000-10-30 2002-05-10 The Board Of Regents, The University Of Texas System Spherical particles produced by a hot-melt extrusion/spheronization process
GB0026593D0 (en) * 2000-10-31 2000-12-13 Quadrant Holdings Cambridge Derivatised carbohydrates and their use in solid delivery systems
WO2002038126A2 (en) 2000-11-08 2002-05-16 Aeromatic-Fielder Ag A process for production of particles for pharmaceutical compositions having increased bioavailability
WO2002038184A1 (en) 2000-11-09 2002-05-16 Astrazeneca Ab Oral pharmaceutical composition containing a block copolymer
GB0027357D0 (en) 2000-11-09 2000-12-27 Bradford Particle Design Plc Particle formation methods and their products
GB0029843D0 (en) 2000-12-07 2001-01-24 Univ Belfast Drug delivery system
US7883721B2 (en) 2001-01-30 2011-02-08 Smithkline Beecham Limited Pharmaceutical formulation
US20050175687A1 (en) 2001-01-30 2005-08-11 Mcallister Stephen M. Pharmaceutical formulations
US7842308B2 (en) 2001-01-30 2010-11-30 Smithkline Beecham Limited Pharmaceutical formulation
DE60211130T2 (en) 2001-02-13 2006-11-30 Astrazeneca Ab NEW FORMULATION WITH MODIFIED RELEASE
GB0104752D0 (en) 2001-02-27 2001-04-18 Astrazeneca Ab Pharmaceutical compositions
US6713081B2 (en) * 2001-03-15 2004-03-30 The United States Of America As Represented By The Department Of Health And Human Services Ocular therapeutic agent delivery devices and methods for making and using such devices
TWI231211B (en) * 2001-05-01 2005-04-21 Abbott Lab Compositions and methods for enhancing the bioavailability of pharmaceutical agents
AR033711A1 (en) 2001-05-09 2004-01-07 Novartis Ag PHARMACEUTICAL COMPOSITIONS
DE10127134A1 (en) 2001-06-05 2002-12-12 Roehm Gmbh Production of injection molded shaped articles, especially for retarded drug release, by blending (meth)acrylate copolymer with plasticizer and other additives, degassing and molding
US20030212102A1 (en) 2001-06-12 2003-11-13 Koretke Todd W Novel solid dispersion compositions
NZ529490A (en) 2001-06-22 2005-08-26 Pfizer Prod Inc Pharmaceutical compositions of adsorbates of amorphous drug
CA2450748A1 (en) 2001-06-22 2003-01-03 Pfizer Products Inc. Pharmaceutical compositions containing polymer and drug assemblies
ITMI20011337A1 (en) 2001-06-26 2002-12-26 Farmatron Ltd ORAL PHARMACEUTICAL COMPOSITIONS WITH MODIFIED RELEASE OF THE ACTIVE SUBSTANCE
ITMI20011338A1 (en) * 2001-06-26 2002-12-26 Farmatron Ltd ORAL PHARMACEUTICAL COMPOSITIONS WITH IMMEDIATE RELEASE OF THE ACTIVE INGREDIENT
US20030044474A1 (en) 2001-08-03 2003-03-06 Shaklee Corporation High molecular weight, lipophilic, orally ingestible bioactive agents in formulations having improved bioavailability
FR2828380B1 (en) 2001-08-10 2005-07-29 Fontarome COMPOSITE COMPOSITE COMPOSITION CONTAINING AROMATIC SUBSTANCES AND / OR VOLATILE NON-AROMATIC SUBSTANCES AND / OR SENSITIVE TO EXTERNAL AGENTS, AND PROCESS FOR PRODUCING THE SAME
JP4644397B2 (en) * 2001-09-05 2011-03-02 信越化学工業株式会社 Method for producing pharmaceutical solid preparation containing poorly soluble drug
WO2003024426A1 (en) 2001-09-21 2003-03-27 Egalet A/S Controlled release solid dispersions
ATE404179T1 (en) 2001-09-28 2008-08-15 Mcneil Ppc Inc DOSAGE FORMS WITH CORE AND OUTER SHELL
US6982094B2 (en) 2001-09-28 2006-01-03 Mcneil-Ppc, Inc. Systems, methods and apparatuses for manufacturing dosage forms
US7122143B2 (en) 2001-09-28 2006-10-17 Mcneil-Ppc, Inc. Methods for manufacturing dosage forms
DE10151290A1 (en) 2001-10-22 2003-04-30 Roehm Gmbh Process for the preparation of active ingredient-containing pellets
JP2005509001A (en) 2001-10-29 2005-04-07 セリクス, インコーポレイテッド Three-dimensional suspension printing of dosage forms
US7491407B2 (en) * 2001-10-31 2009-02-17 North Carolina State University Fiber-based nano drug delivery systems (NDDS)
JP2003146869A (en) 2001-11-14 2003-05-21 Scg:Kk Intraoral disintegration type solid preparation and its production
US6524606B1 (en) 2001-11-16 2003-02-25 Ap Pharma, Inc. Bioerodible polyorthoesters containing amine groups
WO2003043603A1 (en) 2001-11-20 2003-05-30 Advanced Inhalation Research, Inc. Particulate compositions for improving solubility of poorly soluble agents
US7179488B2 (en) * 2001-11-29 2007-02-20 Bob Sherwood Process for co-spray drying liquid herbal extracts with dry silicified MCC
AU2002219964A1 (en) 2001-11-29 2003-06-17 Penwest Pharmaceutical Company Agglomerated particles including an active agent coprocessed with silicified microcrystalline cellulose
US20030206978A1 (en) 2001-11-29 2003-11-06 Bob Sherwood Agglomerated particles including an active agent coprocessed with silicified microcrystalline cellulose
US20030147965A1 (en) 2001-12-10 2003-08-07 Spherics, Inc. Methods and products useful in the formation and isolation of microparticles
NZ533784A (en) 2002-01-03 2006-08-31 Smithkline Beecham Corp Novel pharmaceutical dosage forms and method for producing same
CN1688291A (en) 2002-02-01 2005-10-26 辉瑞产品公司 Immediate release dosage forms containing solid drug dispersions
KR100758045B1 (en) 2002-02-01 2007-09-11 화이자 프로덕츠 인크. Method for Making Homogeneous Spray-dried Solid Amorphous Drug Dispersions Using Pressure Nozzles
ATE395044T1 (en) 2002-02-01 2008-05-15 Pfizer Prod Inc PHARMACEUTICAL COMPOSITIONS OF AMORPHIC DISPERSIONS OF ACTIVE INGREDIENTS AND LIPOPHILIC MICROPHASE-FORMING MATERIALS
AR038681A1 (en) * 2002-02-14 2005-01-26 Solvay Pharm Bv ORAL FORMULATION OF SOLID SOLUTION OF A POVERLY SOLUBLE ACTIVE SUBSTANCE IN WATER
DE10208344A1 (en) 2002-02-27 2003-09-04 Roehm Gmbh Melt extrusion of active ingredient salts
GB0205253D0 (en) * 2002-03-06 2002-04-17 Univ Gent Immediate release pharmaceutical granule compositions and a continuous process for making them
CA2478519C (en) 2002-03-07 2011-08-02 Eurand Pharmaceuticals Limited Process for loading and thermodynamically activating drugs on polymers by means of supercritical fluids
DE60222734T2 (en) 2002-03-15 2008-07-17 Alrise Biosystems Gmbh Microparticles and process for their preparation
DE10213242A1 (en) 2002-03-25 2003-10-16 Abbott Gmbh & Co Kg Test system for evaluating the compatibility of biologically active substances with copolymers
DE10213977A1 (en) 2002-03-28 2003-10-16 Krauss Maffei Kunststofftech Process for the production of molded articles containing active ingredients
EP3241548A1 (en) 2002-04-05 2017-11-08 Euro-Celtique S.A. Matrix for sustained, invariant and independent release of active compounds
NZ535810A (en) 2002-04-19 2007-11-30 Novartis Ag Biomaterial precipitated from cyclodextrin, an anionic polymer component and an amphiphilic ammonium type compound
US20030203027A1 (en) 2002-04-26 2003-10-30 Ethicon, Inc. Coating technique for deposition of drug substance on a substrate
MXPA04011004A (en) * 2002-05-07 2005-01-25 Control Delivery Sys Inc Processes for forming a drug delivery device.
GB0214013D0 (en) 2002-06-18 2002-07-31 Euro Celtique Sa Pharmaceutical product
US7407670B2 (en) 2002-07-04 2008-08-05 Janssen Pharmaceutica, N.V. Solid dispersions comprising two different polymer matrixes
TW200410714A (en) * 2002-08-07 2004-07-01 Smithkline Beecham Corp Electrospun amorphous pharmaceutical compositions
JP2006500349A (en) 2002-08-12 2006-01-05 ファイザー・プロダクツ・インク Semi-order pharmaceutical and polymeric pharmaceutical compositions
AU2003258209A1 (en) 2002-08-13 2004-02-25 Medtronic, Inc. Active agent delivery systems, medical devices, and methods
CA2494188A1 (en) 2002-08-13 2004-02-19 Medtronic, Inc. Active agent delivery system including a hydrophobic cellulose derivative
US20040062778A1 (en) 2002-09-26 2004-04-01 Adi Shefer Surface dissolution and/or bulk erosion controlled release compositions and devices
DE10247037A1 (en) 2002-10-09 2004-04-22 Abbott Gmbh & Co. Kg Solid, rapid release dosage form, especially for sparingly soluble drugs, obtained by forming and cooling softened, shapable mixture of crosslinked non-thermoplastic carrier, adjuvant and active agent
FR2846557B1 (en) * 2002-10-30 2007-06-08 Statice Sante IMPLANTABLE STRUCTURE FOR PROLONGED AND CONTROLLED RELEASE OF AN ACTIVE INGREDIENT
AU2003304108B2 (en) 2002-10-30 2007-03-22 Spherics, Inc. Nanoparticulate bioactive agents
DE10250711A1 (en) 2002-10-31 2004-05-19 Degussa Ag Pharmaceutical and cosmetic preparations
AU2002350719A1 (en) 2002-11-29 2004-06-23 Janssen Pharmaceutica N.V. Pharmaceutical compositions comprising a basic respectively acidic drug compound, a surfactant and a physiologically tolerable water-soluble acid respectively base
US7670627B2 (en) 2002-12-09 2010-03-02 Salvona Ip Llc pH triggered targeted controlled release systems for the delivery of pharmaceutical active ingredients
CA2510261A1 (en) 2002-12-17 2004-07-01 Abbott Gmbh & Co. Kg Formulation comprising fenofibric acid, a physiologically acceptable salt or derivative thereof
US6872799B2 (en) 2002-12-18 2005-03-29 Ethicon, Inc. Functionalized polymers for medical applications
EP1575563B1 (en) * 2002-12-19 2007-02-14 Pharmacia Corporation Non-hygroscopic formulation comprising a hydroscopic drug
ITMI20022748A1 (en) 2002-12-23 2004-06-24 Eurand Int STABILIZED SOLID DISPERSIONS OF DRUG IN AN ORGANIC CAREER AND PROCEDURE FOR THEIR PREPARATION.
WO2004062643A1 (en) 2003-01-14 2004-07-29 Lifecycle Pharma A/S Dry dispersions
DE10304403A1 (en) * 2003-01-28 2004-08-05 Röhm GmbH & Co. KG Process for the preparation of an oral dosage form with immediate disintegration and drug release
US20040258752A1 (en) 2003-01-31 2004-12-23 Paruthi Manoj Kumar Taste masking pharmaceutical composition and process for its preparation
CN100594023C (en) 2003-02-03 2010-03-17 诺瓦提斯公司 Pharmaceutical formulation
US20040156894A1 (en) 2003-02-07 2004-08-12 Grother Leon Paul Use of edible acids in fast-dispersing pharmaceutical solid dosage forms
US6931888B2 (en) 2003-02-07 2005-08-23 Ferro Corporation Lyophilization method and apparatus for producing particles
WO2004071494A2 (en) 2003-02-13 2004-08-26 Phares Pharmaceutical Research N.V. Lipophilic compositions
GB0304726D0 (en) * 2003-03-01 2003-04-02 Ardana Bioscience Ltd New Process
GB0305941D0 (en) 2003-03-14 2003-04-23 Camurus Ab Composition
US20040185170A1 (en) 2003-03-21 2004-09-23 Shubha Chungi Method for coating drug-containing particles and formulations and dosage units formed therefrom
JP4989217B2 (en) * 2003-03-26 2012-08-01 エガレット エイ/エス Matrix composition for controlled delivery of drug substance
US20040208928A1 (en) 2003-04-15 2004-10-21 Animal Technology Institute Taiwan Method for preparing an orally administrable formulation for controlled release
CA2523218A1 (en) * 2003-04-22 2004-11-04 Dr. Reddy's Laboratories Limited Oral pharmaceutical formulations of acid-labile active ingredients and process for making same
JP2007516220A (en) 2003-05-06 2007-06-21 ビーピーエスアイ ホールディングス,インコーポレーテッド Method for producing thermoforming composition containing acrylic polymer adhesive, pharmaceutical preparation and method for producing the preparation
GB0310300D0 (en) 2003-05-06 2003-06-11 Univ Belfast Nanocomposite drug delivery composition
JP2004339162A (en) 2003-05-16 2004-12-02 Shin Etsu Chem Co Ltd Pharmaceutical solid preparation containing sparingly soluble medicine and method for producing the same
EP1479381A1 (en) 2003-05-19 2004-11-24 Euro-Celtique S.A. Pharmaceutical dosage form comprising a solid solution
MXPA05012821A (en) * 2003-05-28 2006-02-13 Nektar Therapeutics Pharmaceutical formulation comprising a water-insoluble active agent.
US20040247624A1 (en) 2003-06-05 2004-12-09 Unger Evan Charles Methods of making pharmaceutical formulations for the delivery of drugs having low aqueous solubility
EP1638533A1 (en) 2003-06-18 2006-03-29 John Michael Newton Controlled release devices with lumens
WO2005000237A2 (en) 2003-06-25 2005-01-06 University Of Tennessee Research Foundation Granules containing biologically active substances
CA2531486C (en) 2003-07-11 2012-10-02 Antonio A. Albano Saquinavir mesylate oral dosage form
DE10332160A1 (en) 2003-07-15 2005-02-03 Röhm GmbH & Co. KG Multiparticulate dosage form containing mucoadhesively formulated peptide or protein active substances, and a method for producing the dosage form
TW200526274A (en) 2003-07-21 2005-08-16 Smithkline Beecham Plc Pharmaceutical formulations
BR0302523A (en) 2003-07-23 2005-04-05 Cristalia Prod Quimicos Farm Stable pharmaceutical composition for administration of hiv protease inhibitors and process for obtaining concentrated pharmaceutical composition for administration of hiv protease inhibitors
US7785512B1 (en) 2003-07-31 2010-08-31 Advanced Cardiovascular Systems, Inc. Method and system of controlled temperature mixing and molding of polymers with active agents for implantable medical devices
US7645474B1 (en) * 2003-07-31 2010-01-12 Advanced Cardiovascular Systems, Inc. Method and system of purifying polymers for use with implantable medical devices
DE602004017198D1 (en) 2003-08-04 2008-11-27 Camurus Ab METHOD FOR LOADING AMPHIPHILIC PARTICLES WITH ACTIVE SUBSTANCES
CL2004001884A1 (en) * 2003-08-04 2005-06-03 Pfizer Prod Inc DRYING PROCEDURE FOR SPRAYING FOR THE FORMATION OF SOLID DISPERSIONS AMORPHES OF A PHARMACO AND POLYMERS.
WO2005011635A2 (en) * 2003-08-04 2005-02-10 Pfizer Products Inc. Pharmaceutical compositions of adsorbates of amorphous drugs and lipophilic microphase-forming materials
JP2007501224A (en) * 2003-08-05 2007-01-25 フジ フォト フィルム ビー.ブイ. Use of recombinant or synthetic gelatin-like proteins as stabilizers in lyophilized pharmaceutical compositions
DE10339197A1 (en) 2003-08-22 2005-03-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Spray-dried amorphous powder with low residual moisture and good storage stability
EP1675576A1 (en) 2003-10-03 2006-07-05 LifeCycle Pharma A/S A method for preparing modified release pharmaceutical compositions
US7598412B2 (en) 2003-10-08 2009-10-06 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
US8349361B2 (en) 2003-10-15 2013-01-08 Fuji Chemical Industry Co., Ltd. Composition for rapid disintegrating tablet in oral cavity
KR101159617B1 (en) 2003-10-15 2012-06-27 후지카가쿠고교가부시키가이샤 Tablet quickly disintegrating in oral cavity
US7413690B1 (en) 2003-10-29 2008-08-19 The University Of Mississippi Process and apparatus for producing spherical pellets using molten solid matrices
CA2549539C (en) 2003-12-15 2013-02-12 Council Of Scientific & Industrial Research Taste masked pharmaceutical composition comprising ph sensitive polymer
CA2568378C (en) 2004-05-28 2013-03-19 Abbott Gmbh & Co. Kg Formulation obtained from a powder mixture comprising an inorganic pigment
US20050281876A1 (en) 2004-06-18 2005-12-22 Shun-Por Li Solid dosage form for acid-labile active ingredient
DE102004040104A1 (en) 2004-08-18 2006-02-23 Basf Ag Use of amphiphilic copolymers as solubilizers
GB0501835D0 (en) 2005-01-28 2005-03-09 Unilever Plc Improvements relating to spray dried compositions
US20090104269A1 (en) 2005-02-11 2009-04-23 Brian Graham Nanoformulations
AU2006218193A1 (en) 2005-02-25 2006-08-31 F. Hoffmann-La Roche Ag Tablets with improved drug substance dispersibility
US20080187612A1 (en) 2005-06-03 2008-08-07 David Kannar Delivery Systems
WO2007002041A2 (en) 2005-06-21 2007-01-04 Foster Corporation Drug-filled polymer films
EP1906940A2 (en) 2005-07-26 2008-04-09 Glaxo Group Limited Encapsulation of lipid-based formulations in enteric polymers
CA2618264C (en) 2005-08-08 2015-11-24 Abbott Gmbh & Co. Kg Dosage forms with improved bioavailability
CN101283008A (en) 2005-08-11 2008-10-08 巴斯夫欧洲公司 N-vinylcaprolactam-based copolymers and the use thereof as solubilizers
UA91376C2 (en) 2005-08-24 2010-07-26 Рубикон Рисеч Пвт Лтд. Controlled release formulation
US20070077305A1 (en) 2005-10-03 2007-04-05 Le Tien C Biocompatible polymeric matrix and preparation thereof
WO2007050631A2 (en) 2005-10-25 2007-05-03 Cima Labs Inc. Dosage form with coated active
ITMI20052461A1 (en) * 2005-12-22 2007-06-23 Univ Degli Studi Milano MICROPARTELAR SYSTEMS FOR ORAL ADMINISTRATION OF BIOLOGICALLY ACTIVE SUBSTANCES
EP1986591A1 (en) 2006-02-10 2008-11-05 LG Household & Health Care Ltd. In-situ melting and gelling tablet composition for oral care
US20070287664A1 (en) 2006-03-23 2007-12-13 Schering Corporation Combinations of HCV protease inhibitor(s) and CYP3A4 inhibitor(s), and methods of treatment related thereto
US7771632B2 (en) 2006-05-15 2010-08-10 American Leistritz Extruder Corp. Continuous melt spheronization apparatus and process for the production of pharmaceutical pellets
EP2029107A2 (en) 2006-06-07 2009-03-04 Basf Se Utilization of vinyl acetate sulfonate copolymers as solubilizers for compounds with low solubility in water
US8343548B2 (en) * 2006-08-08 2013-01-01 Shin-Etsu Chemical Co., Ltd. Solid dosage form comprising solid dispersion
US7923026B2 (en) 2006-10-20 2011-04-12 Solvay Pharmaceuticals B.V. Embedded micellar nanoparticles
WO2008067164A2 (en) 2006-11-15 2008-06-05 Abbott Laboratories Solid pharmaceutical dosage formulations
WO2008065502A1 (en) * 2006-11-29 2008-06-05 Pfizer Products Inc. Pharmaceutical compositions based on a) nanoparticles comprising enteric polymers and b) casein
DE102007009242A1 (en) 2007-02-22 2008-09-18 Evonik Röhm Gmbh Pellets with enteric-coated matix
WO2009013202A1 (en) 2007-07-26 2009-01-29 Basf Se Process for preparing copolymers obtained by graft polymerization in solution and based on polyethers in solid form
EP2022805A3 (en) * 2007-08-03 2009-02-25 Basf Se Copolymers based on N-vinyllactams and olefins as their use as solubilizers for slightly water-soluble compounds
MY185300A (en) 2007-10-11 2021-04-30 Philip Morris Products Sa Smokeless tobacco product
CA2718255C (en) * 2008-03-11 2016-08-23 Aska Pharmaceutical Co., Ltd. Solid dispersion and pharmaceutical composition of the same, and production processes thereof
AR071375A1 (en) 2008-04-22 2010-06-16 Solvay Pharm Gmbh FORMULATIONS FOR ACTIVE PHARMACEUTICAL INGREDIENTS OF DEFICIENT PERMEABILITY, PREPARATION AND PRODUCT PROCESS
WO2010017053A1 (en) 2008-08-06 2010-02-11 Isp Investments, Inc. Solid excipient compositions
GB0815852D0 (en) 2008-09-01 2008-10-08 Unilever Plc Improvements relating to pharmaceutical compositions
US20100068268A1 (en) * 2008-09-15 2010-03-18 Miloud Rahmouni Starch-based microparticles for the release of agents disposed therein
US8778401B2 (en) 2008-10-28 2014-07-15 Agency For Science, Technology And Research Mesoporous material excipients for poorly aqueous soluble ingredients
US8715715B2 (en) 2008-11-03 2014-05-06 Nal Pharmaceuticals Ltd. Dosage form for insertion into the mouth
WO2010078429A1 (en) 2008-12-30 2010-07-08 Impax Laboratories, Inc. Pharmaceutical dosage forms and methods of manufacturing same
CN101444494B (en) 2008-12-31 2011-03-30 江苏大学 Efficient long-acting sustained-release preparation of slightly soluble medicine and preparation method thereof
CA2751667C (en) 2009-02-06 2016-12-13 Egalet Ltd. Immediate release composition resistant to abuse by intake of alcohol
EP2311435A1 (en) 2009-10-07 2011-04-20 LEK Pharmaceuticals d.d. Pharmaceutical composition comprising poorly soluble active ingredient and hyperbranched polymer
WO2011090724A2 (en) 2009-12-29 2011-07-28 Impax Laboratories, Inc. Gastroretentive solid oral dosage forms with lipid-based low-density excipient
EP2552402B1 (en) 2010-03-26 2016-11-23 Dow Global Technologies LLC Multilayer melt-extruded film
US20110288181A1 (en) 2010-05-21 2011-11-24 Basf Se Preparations of biologically active substances with enlarged surface based on amphiphilic copolymers
CN103153343B (en) 2010-06-14 2015-02-11 陶氏环球技术有限责任公司 Hydroxypropyl methyl cellulose acetate succinate with enhanced acetate and succinate substitution

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4758427A (en) * 1985-08-08 1988-07-19 Ciba-Geigy Corporation Enhanced absorption of psychoactive 2-aryl-pyrazolo quinolines as a solid molecular dispersion in polyvinylpyrrolidone
US4851438A (en) * 1985-08-19 1989-07-25 S. C. Johnson & Son, Inc. Aqueous pyrethroid insecticidal formulations in polyvinyl chloride containers, and methods of producing such formulations that are stable
US4801460A (en) * 1986-04-11 1989-01-31 Basf Aktiengesellschaft Preparation of solid pharmaceutical forms
US4904699A (en) * 1986-12-18 1990-02-27 Bauer Kurt H Nifedipine concentrate stabilized against the influence of light and a process for its preparation
US4804699A (en) * 1987-05-15 1989-02-14 Ici Americas Inc. Monomeric and oligomeric glutarate-based light stabilizers for plastics
US4996058A (en) * 1987-09-18 1991-02-26 Ciba-Geigy Corporation Covered retard forms
US5648497A (en) * 1989-05-23 1997-07-15 Abbott Laboraotries Retroviral protease inhibiting compounds
US5541206A (en) * 1989-05-23 1996-07-30 Abbott Laboratories Retroviral protease inhibiting compounds
US5405616A (en) * 1992-01-17 1995-04-11 Alfatec Pharma Gmbh Means for containing active substances, having a shell of hydrophilic macromolecules, active substances and process for preparation thereof
US5525628A (en) * 1992-06-12 1996-06-11 Edmond Pharma S.R.L. Salts of a glutathione alkylester and anaminoacids
US5490990A (en) * 1992-06-25 1996-02-13 Basf Aktiengesellschaft Production of solid pharmaceutical depot forms
US5641516A (en) * 1992-08-13 1997-06-24 Basf Aktiengesellschaft Compositions which contain active substances and are in the form of solid particles
US5773025A (en) * 1993-09-09 1998-06-30 Edward Mendell Co., Inc. Sustained release heterodisperse hydrogel systems--amorphous drugs
US5725878A (en) * 1993-09-13 1998-03-10 Abbott Laboratories Pharmaceutical composition comprising HIV protease inhibiting compounds
US5610193A (en) * 1993-10-01 1997-03-11 Abbott Laboratories Pharmaceutical composition
US5707648A (en) * 1993-11-17 1998-01-13 Lds Technologies, Inc. Transparent liquid for encapsulated drug delivery
US5776495A (en) * 1994-07-26 1998-07-07 Laboratoires Effik Process for the production of dry pharmaceutical forms and the thus obtained pharmaceutical compositions
US5897910A (en) * 1994-12-23 1999-04-27 Basf Aktiengesellschaft Production of covered tablets
US6009690A (en) * 1994-12-23 2000-01-04 Basf Aktiengesellschaft Process and apparatus for the production of divisible tablets
US5545628A (en) * 1995-01-10 1996-08-13 Galephar P.R. Inc. Pharmaceutical composition containing fenofibrate
US5939099A (en) * 1995-02-14 1999-08-17 Basf Aktiengesellschaft Solid active extrusion compound preparations containing low-substituted hydroxypropylcellulose
US5945127A (en) * 1995-03-21 1999-08-31 Basf Aktiengesellschaft Storage-stable drug form
US6063821A (en) * 1995-03-21 2000-05-16 Basf Aktiengesellschaft Transparent rapid release compositions of non-steroidal analgesics
US5741519A (en) * 1995-03-21 1998-04-21 Basf Aktiengesellschaft The production of active substance compositions in the form of a solid solution of the active substance in a polymer matrix, and active substance compositions produced by this process
US6042847A (en) * 1995-05-19 2000-03-28 Lek, Tovarna Farmacevtskih In Kemicnih Izdelkov, D.D. Three-phase pharmaceutical form with constant and controlled release of amorphous active ingredient for single daily application
US6083518A (en) * 1995-06-20 2000-07-04 Bioglan Ab Composition comprising an active agent dissolved in a glass-forming carrier and a process for the preparation thereof
US6703403B2 (en) * 1995-06-29 2004-03-09 Abbott Laboratories Method for improving pharmacokinetics
US6037157A (en) * 1995-06-29 2000-03-14 Abbott Laboratories Method for improving pharmacokinetics
US6387401B2 (en) * 1995-08-25 2002-05-14 Basf Aktiengesellschaft Use of lipids as adjuvents in the production of solid medicinal forms by the melt extrusion process
US6187342B1 (en) * 1995-09-29 2001-02-13 Basf Aktiengesellschaft Solid medicaments obtained by extrusion of an isomalt-containing polymer-active substance melt
US5914332A (en) * 1995-12-13 1999-06-22 Abbott Laboratories Retroviral protease inhibiting compounds
US6251434B1 (en) * 1996-01-23 2001-06-26 Basf Aktiengesellschaft Preparations of non-steroidal analgesics
US6281282B1 (en) * 1996-05-03 2001-08-28 Basf Aktiengesellschaft Polymer powders redispersible in aqueous solution
US6221368B1 (en) * 1996-09-13 2001-04-24 Basf Aktiengesellschaft Process for producing solid dosage forms by extrusion
US5727878A (en) * 1996-10-17 1998-03-17 Cdf Corporation Liner for a mixing container and an assembly and method for mixing fluid components
US6274727B1 (en) * 1996-11-15 2001-08-14 Merck Patent Gesellschaft Mit Beschrankter Haftung Method for producing shaped and unshaped polyol masses
US6232333B1 (en) * 1996-11-21 2001-05-15 Abbott Laboratories Pharmaceutical composition
US6045829A (en) * 1997-02-13 2000-04-04 Elan Pharma International Limited Nanocrystalline formulations of human immunodeficiency virus (HIV) protease inhibitors using cellulosic surface stabilizers
US6066334A (en) * 1997-03-10 2000-05-23 Basf Aktiengesellschaft Use of redispersible polymer powders or polymer granules as binders for producing solid pharmaceutical presentations
US5889051A (en) * 1997-07-15 1999-03-30 Development Center For Biotechnology Stabilization of prostaglandin drug
US20010006650A1 (en) * 1997-09-19 2001-07-05 Beth A. Burnside Solid solution beadlet
US6692767B2 (en) * 1997-09-19 2004-02-17 Shire Laboratories Inc. Solid solution beadlet
US6027747A (en) * 1997-11-11 2000-02-22 Terracol; Didier Process for the production of dry pharmaceutical forms and the thus obtained pharmaceutical compositions
US6547997B1 (en) * 1997-11-28 2003-04-15 Abbot Laboratories Method for producing solvent-free noncrystalline biologically active substances
US6190781B1 (en) * 1998-01-20 2001-02-20 Mitsubishi Paper Mills Limited Support for imaging material
US20040062802A1 (en) * 1998-04-02 2004-04-01 Hermelin Victor M. Maximizing effectiveness of substances used to improve health and well being
US5945123A (en) * 1998-04-02 1999-08-31 K-V Pharmaceutical Company Maximizing effectiveness of substances used to improve health and well being
US6436440B1 (en) * 1998-04-02 2002-08-20 Basf Aktiengesellschaft Use of N-vinyllactam-or-N-vinylamine-containing copolymers as matrix for producing solid pharmaceutical and cosmetic presentations
US6894171B1 (en) * 1998-07-20 2005-05-17 Abbott Laboratories Polymorph of a pharmaceutical
US6350398B1 (en) * 1998-09-03 2002-02-26 Basf Aktiengesellschaft Process for producing coated solid dosage forms
US6599931B1 (en) * 1998-09-18 2003-07-29 Abbott Gmbh & Co. Kg Test system for characterizing the compatibility of bioactive substances and polyvinylpyrrolidone
US20020015814A1 (en) * 1998-10-07 2002-02-07 Weinstein Larry J. Pre-cut fibrous insulation for custom fitting building cavities of different widths
US6423256B1 (en) * 1998-10-15 2002-07-23 Basf Aktiengesellschaft Process for producing solid dosage forms
US6737005B1 (en) * 1998-11-23 2004-05-18 Abbott Gmbh & Co. Kg Method of producing solid dosage forms
US20040013734A1 (en) * 1999-02-10 2004-01-22 Pfizer Inc. Pharmaceutical solid dispersions
US6440946B1 (en) * 1999-02-25 2002-08-27 Takeda Chemical Industries, Ltd. Multiple-agents-binding compound, production and use thereof
US6599528B1 (en) * 1999-03-25 2003-07-29 Abbott Gmbh & Co. Kg Mechanically stable pharmaceutical presentations form containing liquid or semisolid surface-active substances
US6383471B1 (en) * 1999-04-06 2002-05-07 Lipocine, Inc. Compositions and methods for improved delivery of ionizable hydrophobic therapeutic agents
US20050100586A1 (en) * 1999-05-19 2005-05-12 Michel Sournac Transdermal device for administering testosterone or one of the derivatives thereof
US7364752B1 (en) * 1999-11-12 2008-04-29 Abbott Laboratories Solid dispersion pharamaceutical formulations
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US7014810B2 (en) * 1999-12-15 2006-03-21 Abbott Gmbh & Co. Kg Method of producing solid shape forms containing an active ingredient
US20020006443A1 (en) * 1999-12-23 2002-01-17 Curatolo William J. Pharmaceutical compositions providing enhanced drug concentrations
US20050014304A1 (en) * 2000-01-25 2005-01-20 Kook-Chul Moon Low temperature polycrystalline silicon type thin film transistor and a method of the thin film transistor fabrication
US20030153608A1 (en) * 2000-03-17 2003-08-14 Markus Maegerlein Torasemide-containing pharmaceutical preparations
US6608198B2 (en) * 2000-03-30 2003-08-19 Abbott Laboratories Crystalline pharmaceutical
US20030161884A1 (en) * 2000-05-30 2003-08-28 Jorg Rosenberg Formulation based on heparin, glycosaminoglycan or heparinoid, use of the formulation and the formulation base
US20040013697A1 (en) * 2000-05-30 2004-01-22 Gunther Berndl Self-emulsifying active substance formulation and use of this formulation
US7229641B2 (en) * 2000-07-05 2007-06-12 Capricorn Pharma, Inc. Rapid-melt compositions methods of making same and methods of using same
US20040029892A1 (en) * 2000-08-03 2004-02-12 Joerg Rosenberg Compositions and dosage forms for application in the oral cavity in the treatment of mykoses
US6372905B1 (en) * 2000-08-31 2002-04-16 Abbott Laboratories Processes and intermediates for preparing retroviral protease inhibitors
US20040014817A1 (en) * 2000-09-19 2004-01-22 Joerg Rosenberg Stable dosage forms containing ubiquinones
US6579521B2 (en) * 2000-10-20 2003-06-17 Chiron Corporation Methods of therapy for HIV infection
US20020114833A1 (en) * 2000-12-06 2002-08-22 Abu-Izza Khawla A. Fast dissolving tablet
US6733781B2 (en) * 2000-12-06 2004-05-11 Wyeth Fast dissolving tablet
US20030021840A1 (en) * 2001-05-03 2003-01-30 Infeld Martin Howard High dose solid unit oral pharmaceutical dosage form of amorphous nelfinavir mesylate and process for making same
US20030096791A1 (en) * 2001-05-31 2003-05-22 Cima Labs Inc. Taste masking of highly water-soluble drugs
US20030039686A1 (en) * 2001-06-06 2003-02-27 Karsten Maeder Pharmaceutical compositions having depressed melting points
US6730319B2 (en) * 2001-06-06 2004-05-04 Hoffmann-La Roche Inc. Pharmaceutical compositions having depressed melting points
US20030072801A1 (en) * 2001-06-22 2003-04-17 Pfizer Inc. Pharmaceutical compositions comprising drug and concentration-enhancing polymers
US20030091643A1 (en) * 2001-06-22 2003-05-15 Friesen Dwayne T. Pharmaceutical compositions of dispersions of drugs and neutral polymers
US20030104063A1 (en) * 2001-06-22 2003-06-05 Babcock Walter C. Pharmaceutical compositions of dispersions of amorphous drugs mixed with polymers
US20030054038A1 (en) * 2001-06-22 2003-03-20 Crew Marshall D. Pharmaceutical compositions of drugs and neutralized acidic polymers
US20050008706A1 (en) * 2001-07-06 2005-01-13 Per Holm Controlled agglomeration
US20030091630A1 (en) * 2001-10-25 2003-05-15 Jenny Louie-Helm Formulation of an erodible, gastric retentive oral dosage form using in vitro disintegration test data
US6763607B2 (en) * 2002-02-01 2004-07-20 Pfizer Inc. Method for making homogeneous spray-dried solid amorphous drug dispersions utilizing modified spray-drying apparatus
US20040024031A1 (en) * 2002-05-03 2004-02-05 Transform Pharmaceuticals, Inc. Solvates and polymorphs of ritonavir and methods of making and using the same
US20050025791A1 (en) * 2002-06-21 2005-02-03 Julius Remenar Pharmaceutical compositions with improved dissolution
US20040091529A1 (en) * 2002-06-26 2004-05-13 David Edgren Methods and dosage forms for increasing solubility of drug compositions for controlled delivery
US20040001888A1 (en) * 2002-06-26 2004-01-01 Biopharm Solutions Inc. Solid dosage forms for rapid dissolution of poorly soluble drugs
US20040003004A1 (en) * 2002-06-28 2004-01-01 Microsoft Corporation Time-bound database tuning
US20040096499A1 (en) * 2002-08-05 2004-05-20 Navin Vaya Novel dosage form
US20050031691A1 (en) * 2002-09-11 2005-02-10 Elan Pharma International Ltd. Gel stabilized nanoparticulate active agent compositions
US20050079138A1 (en) * 2002-12-19 2005-04-14 Chickering Donald E. Methods for making pharmaceutical formulations comprising microparticles with improved dispersibility, suspendability or wettability
US20060115539A1 (en) * 2003-06-07 2006-06-01 Armin Prasch Micropellets method for the production thereof, and use thereof
US20050048112A1 (en) * 2003-08-28 2005-03-03 Jorg Breitenbach Solid pharmaceutical dosage form
US20050084529A1 (en) * 2003-08-28 2005-04-21 Joerg Rosenberg Solid pharmaceutical dosage form
US20050143404A1 (en) * 2003-08-28 2005-06-30 Joerg Rosenberg Solid pharmaceutical dosage formulation
US20060003942A1 (en) * 2003-10-27 2006-01-05 Roger Tung Combinations for HCV treatment

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8691878B2 (en) 2003-08-28 2014-04-08 Abbvie Inc. Solid pharmaceutical dosage form

Also Published As

Publication number Publication date
EP3482762A1 (en) 2019-05-15
RS57938B1 (en) 2019-01-31
PT2206500T (en) 2018-07-04
TR201816268T4 (en) 2018-11-21
EA014446B1 (en) 2010-12-30
CY1120377T1 (en) 2019-07-10
PE20100123A1 (en) 2010-03-03
JP2008531565A (en) 2008-08-14
JP5087409B2 (en) 2012-12-05
GEP20105083B (en) 2010-09-27
TW200716119A (en) 2007-05-01
CY1122777T1 (en) 2020-10-14
CA2598827A1 (en) 2006-08-31
CA2821046A1 (en) 2006-08-31
MY146247A (en) 2012-07-31
KR20070114294A (en) 2007-11-30
ES2675548T3 (en) 2018-07-11
NO20074807L (en) 2007-09-21
HRP20180999T1 (en) 2018-08-10
WO2006091529A2 (en) 2006-08-31
CA2598827C (en) 2013-09-24
SI2283844T1 (en) 2018-12-31
EP2206500A1 (en) 2010-07-14
PE20061016A1 (en) 2006-10-03
CL2017001847A1 (en) 2018-04-27
SI2206500T1 (en) 2018-08-31
CN101163479B (en) 2011-12-07
DOP2006000050A (en) 2006-11-15
BR122012031169A2 (en) 2015-07-07
MX2007010275A (en) 2007-11-08
DK2206500T3 (en) 2018-07-16
HUE038073T2 (en) 2018-09-28
AR055734A1 (en) 2007-09-05
CR9353A (en) 2008-03-18
CN101163479A (en) 2008-04-16
AR077411A2 (en) 2011-08-24
IL185390A (en) 2017-06-29
DK2283844T3 (en) 2018-11-26
CL2009001844A1 (en) 2009-12-11
WO2006091529A3 (en) 2007-02-08
EP1855683A2 (en) 2007-11-21
BRPI0609173A2 (en) 2010-02-23
ES2694073T3 (en) 2018-12-17
TR201809084T4 (en) 2018-07-23
PL2206500T3 (en) 2018-09-28
UY32116A (en) 2011-04-29
RS57378B1 (en) 2018-08-31
ZA200707022B (en) 2009-08-26
LT2283844T (en) 2018-11-12
US20050143404A1 (en) 2005-06-30
US20130004578A1 (en) 2013-01-03
AU2006216856A1 (en) 2006-08-31
SV2007002427A (en) 2007-01-08
NZ599361A (en) 2013-10-25
KR101429024B1 (en) 2014-08-12
EP2283844B1 (en) 2018-08-01
CL2013003554A1 (en) 2014-05-09
DOP2019000185A (en) 2019-08-15
HRP20181819T1 (en) 2019-01-11
GT200600295AA (en) 2012-03-12
US8377952B2 (en) 2013-02-19
PT2283844T (en) 2018-11-16
EP2206500B1 (en) 2018-03-28
LT2206500T (en) 2018-07-10
AU2006216856B2 (en) 2012-03-08
TWI381840B (en) 2013-01-11
HUE041591T2 (en) 2019-05-28
NZ560829A (en) 2010-12-24
UA89220C2 (en) 2010-01-11
CA2821046C (en) 2018-04-24
UY29391A1 (en) 2006-10-02
EP2283844A1 (en) 2011-02-16
EA200701790A1 (en) 2008-02-28
IL185390A0 (en) 2008-02-09
GT200600295A (en) 2007-09-13
PL2283844T3 (en) 2019-03-29

Similar Documents

Publication Publication Date Title
US8377952B2 (en) Solid pharmaceutical dosage formulation
US8691878B2 (en) Solid pharmaceutical dosage form
EP2258346B1 (en) Solid pharmaceutical dosage form comprising an hiv protease inhibitor solid dispersion
AU2012202831B2 (en) A solid pharmaceutical dosage formulation

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBOTT LABORATORIES, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ROSENBERG, JOERG;REINHOLD, ULRICH;LIEPOLD, BERND;AND OTHERS;REEL/FRAME:021729/0964;SIGNING DATES FROM 20050901 TO 20050908

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: ABBVIE INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABBOTT LABORATORIES;REEL/FRAME:030182/0547

Effective date: 20120801