US20140171481A1 - Solid compositions - Google Patents

Solid compositions Download PDF

Info

Publication number
US20140171481A1
US20140171481A1 US14/180,531 US201414180531A US2014171481A1 US 20140171481 A1 US20140171481 A1 US 20140171481A1 US 201414180531 A US201414180531 A US 201414180531A US 2014171481 A1 US2014171481 A1 US 2014171481A1
Authority
US
United States
Prior art keywords
pharmaceutically acceptable
solid
surfactant
solid dispersion
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/180,531
Inventor
Bernd Liepold
Tina Jung
Peter H?lig
Rudolf Schroeder
Nancy E. Sever
Justin S. Lafountaine
Brent D. Sinclair
Yi Gao
Jianwei Wu
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.)
Abbott GmbH and Co KG
AbbVie Inc
Original Assignee
Abbott GmbH and Co KG
AbbVie Inc
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=44317041&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20140171481(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Abbott GmbH and Co KG, AbbVie Inc filed Critical Abbott GmbH and Co KG
Priority to US14/180,531 priority Critical patent/US20140171481A1/en
Assigned to ABBOTT GMBH & CO. KG reassignment ABBOTT GMBH & CO. KG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOLIG, PETER, JUNG, TINA, LIEPOLD, BERND, SCHROEDER, RUDOLF
Assigned to ABBOTT LABORATORIES reassignment ABBOTT LABORATORIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABBOTT GMBH & CO.KG
Assigned to ABBOTT LABORATORIES reassignment ABBOTT LABORATORIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SEVER, NANCY E., GAO, YI, LAFOUNTAINE, JUSTIN S., SINCLAIR, BRENT D., WU, JIANWEI
Assigned to ABBVIE INC. reassignment ABBVIE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABBOTT LABORATORIES
Publication of US20140171481A1 publication Critical patent/US20140171481A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/146Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic macromolecular compounds
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4025Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil not condensed and containing further heterocyclic rings, e.g. cromakalim
    • 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
    • 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
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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

Definitions

  • the present invention relates to solid compositions comprising anti-HCV compounds and methods of using the same to treat HCV infection.
  • the hepatitis C virus is an RNA virus belonging to the Hepacivirus genus in the Flaviviridae family.
  • the enveloped HCV virion contains a positive stranded RNA genome encoding all known virus-specific proteins in a single, uninterrupted, open reading frame.
  • the open reading frame comprises approximately 9500 nucleotides and encodes a single large polyprotein of about 3000 amino acids.
  • the polyprotein comprises a core protein, envelope proteins E1 and E2, a membrane bound protein p7, and the non-structural proteins NS2, NS3, NS4A, NS4B, NS5A and NS5B.
  • HCV infection is associated with progressive liver pathology, including cirrhosis and hepatocellular carcinoma.
  • Chronic hepatitis C may be treated with peginterferon-alpha in combination with ribavirin.
  • Substantial limitations to efficacy and tolerability remain as many users suffer from side effects, and viral elimination from the body is often inadequate. Therefore, there is a need for new drugs to treat HCV infection.
  • the present invention features solid compositions comprising dimethyl(2S,2′S)-1,1′-((2S,2′S)-2,2′-(4,4′-((2S,5S)-1-(4-tert-butylphenyl)pyrrolidine-2,5-diyl)bis(4,1-phenylene))bis(azanediyl)bis(oxomethylene)bis(pyrrolidine-2,1-diyl))bis(3-methyl-1-oxobutane-2,1-diyl)dicarbamate (
  • Compound I is a potent HCV inhibitor.
  • the solid compositions of the invention comprise (1) Compound I (or a pharmaceutically acceptable salt thereof) in an amorphous form, (2) a pharmaceutically acceptable hydrophilic polymer, and optionally (3) a pharmaceutically acceptable surfactant.
  • the present invention features a solid composition
  • a solid composition comprising a solid dispersion, wherein the solid dispersion comprises Compound I (or a pharmaceutically acceptable salt thereof) in an amorphous form and a pharmaceutically acceptable hydrophilic polymer, and the solid composition further comprises a pharmaceutically acceptable surfactant.
  • the surfactant can be, without limitation, either formulated in the solid dispersion or separately combined or mixed with the solid dispersion.
  • the hydrophilic polymer has a T g of at least 50° C. More preferably, the hydrophilic polymer has a T g of at least 80° C. Highly preferably, the hydrophilic polymer has a T g of at least 100° C. Also preferably, the surfactant has a HLB value of at least 10. Hydrophilic polymers with T g of at least 25° C. can also be used.
  • the hydrophilic polymer is selected from homopolymer of N-vinyl lactam, copolymer of N-vinyl lactam, cellulose ester, cellulose ether, polyalkylene oxide, polyacrylate, polymethacrylate, polyacrylamide, polyvinyl alcohol, vinyl acetate polymer, oligosaccharide, or polysaccharide.
  • Non-limiting examples of suitable hydrophilic polymers include homopolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone and vinyl acetate, copolymer of N-vinyl pyrrolidone and vinyl propionate, polyvinylpyrrolidone, methylcellulose, ethylcellulose, hydroxyalkylcelluloses, hydroxypropylcellulose, hydroxyalkylalkylcellulose, hydroxypropylmethylcellulose, cellulose phthalate, cellulose succinate, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, hydroxypropylmethylcellulose acetate succinate, polyethylene oxide, polypropylene oxide, copolymer of ethylene oxide and propylene oxide, methacrylic acid/ethyl acrylate copolymer, methacrylic acid/methyl methacrylate copoly
  • the surfactant is selected from polyoxyethylene castor oil derivates, mono fatty acid ester of polyoxyethylene sorbitan, polyoxyethylene alkyl ether, polyoxyethylene alkylaryl ether, polyethylene glycol fatty acid ester, alkylene glycol fatty acid mono ester, sucrose fatty acid ester, or sorbitan fatty acid mono ester.
  • Non-limiting examples of suitable surfactants include polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate such as polyethylenglycol 40 hydrogenated castor oil (Cremophor® RH 40, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate) or polyethylenglycol 60 hydrogenated castor oil (Cremophor® RH 60), mono fatty acid ester of polyoxyethylene sorbitan, such as mono fatty acid ester of polyoxyethylene (20) sorbitan, e.g.
  • the solid dispersion is an amorphous solid dispersion.
  • the solid dispersion is an amorphous solid dispersion which comprises Compound I (or a pharmaceutically acceptable salt thereof), the hydrophilic polymer, and the surfactant.
  • the solid dispersion is a solid solution comprising Compound I (or a pharmaceutically acceptable salt thereof) and the hydrophilic polymer.
  • the solid dispersion is a solid solution comprising Compound I (or a pharmaceutically acceptable salt thereof), the hydrophilic polymer and the surfactant.
  • the hydrophilic polymer is a homopolymer or copolymer of N-vinyl pyrrolidone.
  • the hydrophilic polymer is copovidone.
  • the surfactant is D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS).
  • the surfactant is a sorbitan fatty acid ester, such as sorbitan mono laurate (Span® 20).
  • a solid composition of the invention comprises an amorphous solid dispersion or a solid solution which comprises Compound I (or a pharmaceutically acceptable salt thereof), copovidone, and a surfactant selected from vitamin E TPGS, Span 20, or a combination thereof.
  • the present invention features processes of making a solid composition of the present invention.
  • the process comprises drying a solvent in a liquid solution, wherein said solution comprises: (1) Compound I or a pharmaceutically acceptable salt thereof; (2) a pharmaceutically acceptable hydrophilic polymer; and optionally (3) a pharmaceutically acceptable surfactant.
  • the drying process can be carried out using any suitable solvent evaporation techniques including but not limited to spray-drying techniques.
  • the process comprises solidifying a melt which comprises: (1) Compound I or a pharmaceutically acceptable salt thereof; (2) a pharmaceutically acceptable hydrophilic polymer; and optionally (3) a pharmaceutically acceptable surfactant.
  • a solid composition of the invention may also contain other additives or ingredients, such as coloring agents, flavoring agents, lubricants or preservatives.
  • a solid composition of the invention can be prepared into any suitable dosage forms, such as capsule, dragee, granule, powder, or tablet.
  • a solid composition of the invention may further comprise another anti-HCV agent, for example, an agent selected from HCV helicase inhibitors, HCV polymerase inhibitors.
  • HCV protease inhibitors HCV NS5A inhibitors, CD81 inhibitors, cyclophilin inhibitors, or internal ribosome entry site (IRES) inhibitors.
  • the present invention further features methods of using a solid composition of the present invention to treat HCV infection.
  • the methods comprise administering a solid composition of the present invention to a patient in need thereof, thereby reducing the blood or tissue level of HCV virus in the patient.
  • the present invention features solid compositions comprising amorphous Compound I (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable hydrophilic polymer, and optionally a pharmaceutically acceptable surfactant.
  • amorphous Compound I or a pharmaceutically acceptable salt thereof
  • a pharmaceutically acceptable hydrophilic polymer or a pharmaceutically acceptable hydrophilic polymer
  • optionally a pharmaceutically acceptable surfactant for example, a pharmaceutically acceptable surfactant.
  • a non-limiting way to form an amorphous form of Compound I is through the formation of solid dispersions with a polymeric carrier.
  • the presence of hydrophilic polymer(s) and optional surfactant(s), as well as the dispersion of Compound I in an amorphous form in a matrix containing the polymer(s), can significantly enhance the dissolution rate of Compound I.
  • a solid dispersion formulation can also effectively maintain Compound I in its supersaturation state to allow for better absorption.
  • solid dispersion defines a system in a solid state (as opposed to a liquid or gaseous state) comprising at least two components, wherein one component is dispersed throughout the other component or components.
  • an active ingredient or a combination of active ingredients can be dispersed in a matrix comprised of a pharmaceutically acceptable hydrophilic polymer(s) and a pharmaceutically acceptable surfactant(s).
  • solid dispersion encompasses systems having small particles of one phase dispersed in another phase. These particles are often of less than 400 ⁇ m in size, such as less than 100, 10, or 1 ⁇ m in size.
  • a solid dispersion of the components is such that the system is chemically and physically uniform or homogenous throughout or consists of one phase (as defined in thermodynamics)
  • a solid dispersion is called a “solid solution.”
  • a glassy solution is a solid solution in which a solute is dissolved in a glassy solvent.
  • AUC ⁇ or AUC 0-inf refers to the area under the plasma concentration time curve (AUC) extrapolated to infinity.
  • weight percent or “percent by weight” or “% by weight” or “wt %” denote the weight of an individual component in a composition or mixture as a percentage of the weight of the composition or mixture.
  • the present invention features a solid composition
  • a solid composition comprising Compound I (or a pharmaceutically acceptable salt thereof) in an amorphous form, a pharmaceutically acceptable hydrophilic polymer, and a pharmaceutically acceptable surfactant.
  • the Compound I (or the salt thereof) and the polymer are formulated in a solid dispersion.
  • the surfactant may also be formulated in the same solid dispersion; or the surfactant can be separately combined or mixed with the solid dispersion.
  • a solid composition of the invention comprises an amorphous solid dispersion which comprises Compound I (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable hydrophilic polymer and a pharmaceutically acceptable surfactant.
  • a solid composition of the invention comprises a solid solution which comprises Compound I (or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable hydrophilic polymer.
  • a solid composition of the invention comprises a solid solution which comprises Compound I (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable hydrophilic polymer and a pharmaceutically acceptable surfactant.
  • a solid composition of the invention comprises a glassy solution which includes Compound I (or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable hydrophilic polymer.
  • a solid composition of the invention comprises a glassy solution which includes Compound I (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable hydrophilic polymer and a pharmaceutically acceptable surfactant.
  • a solid composition (or a solid dispersion) of the invention can contain, for example, at least 1% by weight of Compound I, preferably at least 5%, including, e.g., at least 10%.
  • a solid composition (or a solid dispersion) of the invention can contain from 1 to 50% by weight of Compound I.
  • a solid composition (or a solid dispersion) of the invention can contain from 5 to 30% by weight of Compound I.
  • a solid composition (or a solid dispersion) of the invention contains from 5 to 15% by weight of Compound I.
  • a solid dispersion of the invention may contain at least 30% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such hydrophilic polymers.
  • the solid dispersion contains at least 40% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such hydrophilic polymers. More preferably, the solid dispersion contains at least 50% (including, e.g., at least 60%, 70%, 80% or 90%) by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers.
  • a solid dispersion of the invention may also contain at least 1% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants.
  • the solid dispersion contains at least 2% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. More preferably, the solid dispersion contains from 4% to 20% by weight of the surfactant(s), such as from 5% to 10% by weight of the surfactant(s).
  • a solid dispersion of the invention comprises at least 30% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and at least 1% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. In another embodiment, a solid dispersion of the invention comprises at least 50% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 2% to 20% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants.
  • a solid dispersion of the invention comprises from 50% to 90% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 3% to 15% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. In yet another embodiment, a solid dispersion of the invention comprises from 70% to 90% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 5% to 10% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants.
  • a hydrophilic polymer employed in the present invention has a T g of at least 50° C., more preferably at least 60° C., and highly preferably at least 80° C. including, but not limited to from, 80° C. to 180° C., or from 100° C. to 150° C.
  • T g values of organic polymers are described in I NTRODUCTION TO P HYSICAL P OLYMER S CIENCE (2nd Edition by L. H. Sperling, published by John Wiley & Sons, Inc., 1992).
  • T g values for the homopolymers may be taken from P OLYMER H ANDBOOK (2nd Edition by J. Brandrup and E. H. Immergut, Editors, published by John Wiley & Sons, Inc., 1975).
  • Hydrophilic polymers with a T g as described above may allow for the preparation of solid dispersions that are mechanically stable and, within ordinary temperature ranges, sufficiently temperature stable so that the solid dispersions may be used as dosage forms without further processing or be compacted to tablets with only a small amount of tabletting aids. Hydrophilic polymers having a T g of below 50° C. may also be used.
  • a hydrophilic polymer employed in the present invention is water-soluble.
  • a solid composition of the present invention can also comprise poorly water-soluble or water-insoluble polymer or polymers, such as cross-linked polymers.
  • a hydrophilic polymer comprised in a solid composition of the present invention preferably has an apparent viscosity, when dissolved at 20° C. in an aqueous solution at 2% (w/v), of 1 to 5000 mPa ⁇ s., and more preferably of 1 to 700 mPa ⁇ s, and most preferably of 5 to 100 mPa ⁇ s.
  • Hydrophilic polymers suitable for use in a solid composition of the invention include, but are not limited to, homopolymers or copolymers of N-vinyl lactams, such as homopolymers or copolymers of N-vinyl pyrrolidone (e.g., polyvinylpyrrolidone (PVP), or copolymers of N-vinyl pyrrolidone and vinyl acetate or vinyl propionate); cellulose esters or cellulose ethers, such as alkylcelluloses (e.g., methylcellulose or ethylcellulose), hydroxyalkylcelluloses (e.g., hydroxypropylcellulose), hydroxyalkylalkylcelluloses (e.g., hydroxypropylmethylcellulose), and cellulose phthalates or succinates (e.g., cellulose acetate phthalate and hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, or hydroxypropylmethylcellulose acetate succinate); high
  • Non-limiting examples of preferred hydrophilic polymers for the invention include polyvinylpyrrolidone (PVP) K17, PVP K25, PVP K30, PVP K90, hydroxypropyl methylcellulose (HPMC) E3, HPMC E5, HPMC E6, HPMC E15, HPMC K3, HPMC A4, HPMC A15, HPMC acetate succinate (AS) LF, HPMC AS MF, HPMC AS HF, HPMC AS LG, HPMC AS MG, HPMC AS HG, HPMC phthalate (P) 50, HPMC P 55, Ethocel 4, Ethocel 7, Ethocel 10, Ethocel 14, Ethocel 20, copovidone (vinylpyrrolidone-vinyl acetate copolymer 60/40), polyvinyl acetate, methacrylate/methacrylic acid copolymer (Eudragit) L100-55, Eudragit L100, Eudragit S100, polyethylene glycol (PEG) 400, PEG 600
  • homopolymers or copolymers of N-vinyl pyrrolidone such as copolymers of N-vinyl pyrrolidone and vinyl acetate
  • a non-limiting example of a preferred polymer is a copolymer of 60% by weight of N-vinyl pyrrolidone and 40% by weight of vinyl acetate.
  • Other preferred polymers include, without limitation, hydroxypropyl methylcellulose (HPMC, also known as hypromellose in USP), such as hydroxypropyl methylcellulose grade E5 (HPMC-E5); and hydroxypropyl methylcellulose acetate succinate (HPMC-AS).
  • a pharmaceutically acceptable surfactant employed in the present invention is preferably a non-ionic surfactant. More preferably, a solid composition of the present invention comprises a pharmaceutically acceptable surfactant having an HLB value of from 2-20.
  • a solid composition of the present invention can also include a mixture of pharmaceutically acceptable surfactants, with at least one surfactant having an HLB value of no less than 10 and at least another surfactant having an HLB value of below 10.
  • each surfactant comprised in a solid composition of the invention has an HLB value of at least 10.
  • each surfactant comprised in a solid composition of the invention has an HLB value of below 10.
  • a solid composition of the present invention includes at least two pharmaceutically acceptable surfactants, one having an HLB value of at least 10 and the other having an HLB value of below 10.
  • the HLB system (Fiedler, H. B., E NCYLOPEDIA OF E XCIPIENTS, 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.
  • Non-limiting examples of pharmaceutically acceptable surfactants that are suitable for the present invention include 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, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate) or polyethylenglycol 60 hydrogenated castor oil (Cremophor® RH 60); or a mono fatty acid ester of polyoxyethylene sorbitan, such as a mono fatty acid ester of polyoxyethylene (20) sorbitan, e.g.
  • polyoxyethylene castor oil derivates e.g. polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL; BASF
  • polyoxyethylene (20) sorbitan monooleate Tween® 80
  • polyoxyethylene (20) sorbitan monostearate Tween® 60
  • polyoxyethylene (20) sorbitan monopalmitate Tween® 40
  • polyoxyethylene (20) sorbitan monolaurate Tween® 20
  • suitable surfactants include 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.
  • sucrose monostearate sucrose distearate, sucrose monolaurate, sucrose dilaurate
  • sorbitan fatty acid mono esters such as sorbitan mono laurate (Span® 20), sorbitan monooleate, sorbitan monopalnitate (Span® 40), or sorbitan stearate.
  • surfactants include, but are not limited to, 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, or Poloxamer® 407 (BASE Wyandotte Corp.).
  • 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, or Poloxamer® 407 (BASE Wyandotte Corp.).
  • Poloxamer® 124, Poloxamer® 188, Poloxamer® 237, Poloxamer® 388, or Poloxamer® 407 BASE Wyandotte Corp.
  • Non-limiting examples of preferred surfactants for the invention include to polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, Cremophor RH 40, Cremophor EL, Gelucire 44/14, Gelucire 50/13, D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS), propylene glycol laurate, sodium lauryl sulfate, and sorbitan monolaurate.
  • a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes Compound I (or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable hydrophilic polymer.
  • the solid composition also includes a pharmaceutically acceptable surfactant which preferably is formulated in the amorphous solid dispersion or solid solution.
  • the hydrophilic polymer can be selected, for example, from the group consisting of homopolymer of N-vinyl lactam, copolymer of N-vinyl lactam, cellulose ester, cellulose ether, polyalkylene oxide, polyacrylate, polymethacrylate, polyacrylamide, polyvinyl alcohol, vinyl acetate polymer, oligosaccharide, and polysaccharide.
  • the hydrophilic polymer is selected from the group consisting of homopolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone and vinyl acetate, copolymer of N-vinyl pyrrolidone and vinyl propionate, polyvinylpyrrolidone, methylcellulose, ethylcellulose, hydroxyalkylcelluloses, hydroxypropylcellulose, hydroxyalkylalkylcellulose, hydroxypropylmethylcellulose, cellulose phthalate, cellulose succinate, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, hydroxypropylmethylcellulose acetate succinate, polyethylene oxide, polypropylene oxide, copolymer of ethylene oxide and propylene oxide, methacrylic acid/ethyl acrylate copolymer, methacrylic acid
  • the hydrophilic polymer is selected from polyvinylpyrrolidone (PVP) K17, PVP K25, PVP K30, PVP K90, hydroxypropyl methylcellulose (HPMC) E3, HPMC E5, HPMC E6, HPMC E15, HPMC K3, HPMC A4, HPMC A15, HPMC acetate succinate (AS) LF, HPMC AS MF, HPMC AS HF, HPMC AS LG, HPMC AS MG, HPMC AS HG, HPMC phthalate (P) 50, HPMC P 55, Ethocel 4, Ethocel 7, Ethocel 10, Ethocel 14, Ethocel 20, copovidone (vinylpyrrolidone-vinyl acetate copolymer 60/40), polyvinyl acetate, methacrylate/methacrylic acid copolymer (Eudragit) L100-55, Eudragit L100, Eudragit S100, polyethylene glycol (PEG) 400, PEG 600, PEG
  • the hydrophilic polymer is selected from homopolymers of vinylpyrrolidone (e.g., PVP with Fikentscher K values of from 12 to 100, or PVP with Fikentscher K values of from 17 to 30), or copolymers of 30 to 70% by weight of N-vinylpyrrolidone (VP) and 70 to 30% by weight of vinyl acetate (VA) (e.g., a copolymer of 60% by weight VP and 40% by weight VA).
  • homopolymers of vinylpyrrolidone e.g., PVP with Fikentscher K values of from 12 to 100, or PVP with Fikentscher K values of from 17 to 30
  • copolymers of 30 to 70% by weight of N-vinylpyrrolidone (VP) and 70 to 30% by weight of vinyl acetate (VA) e.g., a copolymer of 60% by weight VP and 40% by weight VA.
  • the surfactant can be selected, for example, from the group consisting of polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate, mono fatty acid ester of polyoxyethylene sorbitan, polyoxyethylene alkyl ether, polyoxyethylene alkylaryl ether, polyethylene glycol fatty acid ester, alkylene glycol fatty acid mono ester, sucrose fatty acid ester, and sorbitan fatty acid mono ester.
  • polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate, mono fatty acid ester of polyoxyethylene sorbitan, polyoxyethylene alkyl ether, polyoxyethylene alkylaryl ether, polyethylene glycol fatty acid ester, alkylene glycol fatty acid mono ester, sucrose
  • the surfactant is selected from the group consisting of polyethylenglycol 40 hydrogenated castor oil (Cremophor® RE 40, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate), polyethylenglycol 60 hydrogenated castor oil (Cremophor® RH 60), 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), or polyoxyethylene (20) sorbitan monolaurate (Tween® 20)), polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether, polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether, polyoxyethylene (3) octylphenyl ether, PEG-200 monolaurate, PEG-200 dilaurate, PEG-300 dilaurate, PEG-400 dilaurate, PEG-300 distearate, PEG-300 dioleate, propylene glycol monolaurate, sucrose monostea
  • the surfactant is selected from polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, Cremophor RH 40, Cremophor EL, Gelucire 44/14, Gelucire 50/13, D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS), propylene glycol laurate, sodium lauryl sulfate, or sorbitan monolaurate. More preferably, the surfactant is selected from sorbitan monolaurate or D-alpha-tocopheryl polyethylene glycol 1000 succinate.
  • a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes Compound I (or a pharmaceutically acceptable salt thereof) and a homopolymer or copolymer of N-vinyl pyrrolidone (e.g., copovidone).
  • the solid composition also comprises a pharmaceutically acceptable surfactant (e.g., vitamin E TPGS, or sorbitan monolaurate), wherein the surfactant preferably is formulated in the amorphous solid dispersion or solid solution.
  • a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes Compound I (or a pharmaceutically acceptable salt thereof), copovidone, and a pharmaceutically acceptable surfactant selected from vitamin E TPGS or sorbitan monolaurate.
  • the amorphous solid dispersion or solid solution may also include another pharmaceutically acceptable surfactant.
  • a solid dispersion employed in the present invention preferably comprises or consists of a single-phase (defined in thermodynamics) which the therapeutic agent(s) (e.g., Compound I, or a combination of Compound I and another anti-HCV agent) is molecularly dispersed in a matrix containing the pharmaceutically acceptable hydrophilic polymer(s).
  • the therapeutic agent(s) e.g., Compound I, or a combination of Compound I and another anti-HCV agent
  • thermal analysis of the solid dispersion using differential scanning calorimetry (DSC) typically shows only one single T g , and the solid dispersion does not contain any detectable crystalline Compound I as measured by X-ray powder diffraction spectroscopy.
  • Compound I can be prepared according to the procedures described in U.S. Provisional Patent Application No. 61/243,596, filed Sep. 18, 2009, To a mixture of (S)-2-(methoxycarbonylamino)-3-methylbutanoic acid (19.66 g, 112 mmol) and N-hydroxysuccinimide (13.29 g, 116 mmol) was added ethyl acetate (250 ml), and the mixture was cooled to 0-5° C. Diisopropylcarbodiimide (13.88 g, 110 mmol) was added and the reaction mixture was stirred at 0-5° C. for about 1 hour. The reaction mixture was warmed to room temperature.
  • (S)-1-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)pyrrolidine-2-carboxylic acid can also be prepared according to the following procedure. To a flask was charged L-valine (35 g, 299 mmol), 1N sodium hydroxide solution (526 ml, 526 mmol) and sodium carbonate (17.42 g, 164 mmol). The mixture was stirred for 15 min to dissolve solids and then cooled to 15° C. Methyl chloroformate (29.6 g, 314 mmol) was added slowly to the reaction mixture. The mixture was then stirred at rt for 30 min. The mixture was cooled to 15° C.
  • the mixture was stirred at rt for 30 min, filtered and washed with water/DMF mixture (1:2, 250 mL) and then with water (1500 mL).
  • the wetcake was dried at 55° C. for 24 h to give 282 g of product as a white solid (90%).
  • the resulting solids (40 g) and 5% Pd/Alumina were charged to a Parr reactor followed by THF (160 mL).
  • the reactor was sealed and purged with nitrogen (6 ⁇ 2.0 psig) followed by a hydrogen purge (6 ⁇ 30 psig).
  • the reactor was pressurized to 30 psig with hydrogen and agitated at room temperature for approximately 15 hours.
  • the resulting slurry was filtered through a GET filter and concentrated to approximately 135 g solution.
  • the organic phase was treated with 1 M HCl (140 mL), 2-methyltetrahydrofuran (50 mL), and 25 wt % aq. NaCl (100 mL), and the phases were partitioned.
  • the organic phase was washed with 25 wt % aq. NaCl (50 mL), dried over MgSO 4 , filtered, and concentrated in vacuo to approximately 20 mL.
  • Heptane (30 mL) and additional 2-methyltetrahydrofuran were added in order to induce crystallization.
  • the slurry was filtered by vacuum filtration using a GF/F Whatman glass fiber filter. Evaporation of the filtrate to afford a slurry followed by the addition heptane and filtration gave the crude 4,4′-((2S,5S)-1-(4-tert-butylphenyl)pyrrolidine-2,5-diyl)dianiline, which was dried and used directly in the next step.
  • T3P® 1-propanephosphonic acid cyclic anhydride
  • the seeds described above were a EtOH—H 2 O solvate of Compound I and was originally made according to the following procedure.
  • a solution was first prepared by slurrying the amorphous, solid Compound I in 37 weight % EtOH in heptane at room temperature. The saturated solution (with respect to the amorphous solid) was then filtered over to a new vial to give a clear solution and seeded with partially crystalline solid obtained from the same solvent system. Crystallization took place within a few hours to produce the “Anhydrate A” form of Compound I. Then a solution of 60 w % EtOH in H 2 O at 5° C. was prepared using the amorphous Compound I. To this clear solution were added seeds of mainly Anhydrate A and desolvated EtOH solvate. Conversion to the EtOH—H2O solvate took place within 2 days to produce the original seeds, which can be used to make additional seeds.
  • a solid composition of the present invention can further include one or more other anti-HCV agents.
  • These other anti-HCV agents can be, for example, HCV polymerase inhibitors (including nucleoside or non-nucleoside type of polymerase inhibitors), HCV protease inhibitors, HCV helicase inhibitors, CD81 inhibitors, cyclophilin inhibitors, internal ribosome entry site inhibitors, or HCV NS5A inhibitors.
  • anti-HCV agents include, but are not limited to, ribavirin, ⁇ -interferon, ⁇ -interferon, pegylated interferon- ⁇ , pegylated interferon-lambda, PSI-7851 (Pharmasset) (nucleoside polymerase inhibitor), PSI-938 (Pharmasset) (nucleoside polymerase inhibitor), PF-00868554, ANA-598, IDX184 (nucleoside polymerase inhibitor), IDX102, IDX375 (non-nucleoside polymerase inhibitor), GS-9190 (non-nucleoside polymerase inhibitor), VCH-759, VCH-916, MK-3281, BCX-4678, MK-3281, VBY-708, ANA598, GL59728, GL60667, BMS-790052 (NS5A inhibitor), BMS-791325 (protease Inhibitor), BMS-650032, BMS-8
  • a solid composition of the invention comprises Compound I (a pharmaceutically acceptable salt thereof), and a HCV protease inhibitor.
  • a solid composition of the invention comprises Compound I (a pharmaceutically acceptable salt thereof), and a HCV polymerase inhibitor (e.g., a non-nucleoside polymerase inhibitor, or preferably a nucleoside polymerase inhibitor).
  • a solid composition of the invention comprises (1) Compound I (a pharmaceutically acceptable salt thereof), (2) a HCV protease inhibitor, and (3) a HCV polymerase inhibitor (e.g., a non-nucleoside polymerase inhibitor, or preferably a nucleoside polymerase inhibitor).
  • protease and polymerase inhibitors are described above.
  • a solid composition of the present invention preferably is a solid oral dosage form.
  • Common solid oral dosage forms suitable for the present invention include, but are not limited to, capsules, dragees, granules, pills, powders and tablets, with capsules and tablets being preferred.
  • a solid oral dosage form of the present invention can also include other excipients or inset diluents, such as sucrose, lactose or starch.
  • Lubricants, coloring agents, releasing agents, coating agents, sweetening or flavoring agents, buffering agents, preservatives, or antioxidants can also be included in a solid oral dosage form of the present invention.
  • a solid composition of the present invention can be prepared by a variety of techniques such as, without limitation, melt-extrusion, spray-drying, co-precipitation, freeze drying, or other solvent evaporation techniques, with melt-extrusion and spray-drying being preferred.
  • the melt-extrusion process typically comprises the steps of preparing a melt which includes the active ingredient(s), the hydrophilic polymer(s) and preferably the surfactant(s), and then 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, preferably homogeneously embedded, in the other component or components.
  • the polymer component(s) will melt and the other components including the active ingredient(s) and surfactant(s) will dissolve in the melt thereby forming a solution.
  • Melting usually involves heating above the softening point of the polymer(s).
  • 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 can also be homogenized in order to disperse the active ingredient(s) efficiently.
  • all materials except surfactant(s) are blended and fed into an extruder, while the surfactant(s) is molten externally and pumped in during extrusion.
  • the melt comprises Compound I and one or more hydrophilic polymers described above, and the melt temperature is in the range of from 100 to 170° C., preferably from 120 to 150° C., and highly preferably from 135 to 140° C.
  • the melt can also include a pharmaceutically acceptable surfactant described above.
  • the melt comprises Compound I, at least another anti-HCV agent described above, and one or more hydrophilic polymers described above.
  • the melt can also include a pharmaceutically acceptable surfactant described above.
  • the active ingredient(s) e.g., Compound I, or a combination of Compound I and at least another anti-HCV agent
  • the active ingredient(s) can be employed in their solid forms, such as their respective crystalline forms.
  • the active ingredient(s) can also be employed as a solution or dispersion in a suitable liquid solvent such as alcohols, aliphatic hydrocarbons, esters or, in some cases, liquid carbon dioxide.
  • a suitable liquid solvent such as alcohols, aliphatic hydrocarbons, esters or, in some cases, liquid carbon dioxide.
  • the solvent can be removed, e.g. evaporated, upon preparation of the melt.
  • additives can also be included in the melt, for example, flow regulators (e.g., colloidal silica), binders, lubricants, fillers, disintegrants, plasticizers, colorants, or stabilizers (e.g., antioxidants, light stabilizers, radical scavengers, and stabilizers against microbial attack).
  • flow regulators e.g., colloidal silica
  • binders e.g., colloidal silica
  • lubricants e.g., fillers, disintegrants, plasticizers, colorants
  • stabilizers e.g., antioxidants, light stabilizers, radical scavengers, and stabilizers against microbial attack.
  • extruders or kneaders are extruders or kneaders.
  • Suitable extruders include single screw extruders, intermeshing screw extruders or multiscrew extruders, preferably twin screw extruders, which can be corotating or counterrotating and, optionally, be equipped with kneading disks.
  • the working temperatures will 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.
  • 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 can range from thin to pasty to viscous. Shaping of the extrudate can be conveniently carried out by a calendar with two counter-rotating rollers with mutually matching depressions on their surface.
  • the extrudate can be cooled and allow to solidify.
  • the extrudate can also be cut into pieces, either before (hot-cut) or after solidification (cold-cut).
  • the solidified extrusion product can be further milled, ground or otherwise reduced to granules.
  • the solidified extrudate, as well as each granule produced comprises a solid dispersion, preferably a solid solution, of the active ingredient(s) in a matrix comprised of the hydrophilic polymer(s) and optionally the pharmaceutically acceptable surfactant(s). Where the granules do not contain any surfactant, a pharmaceutically acceptable surfactant described above can be added to and blended with the granules.
  • the extrusion product can also be blended with other active ingredient(s) and/or additive(s) before being milled or ground to granules.
  • the granules can be further processed into suitable solid oral dosage forms.
  • copovidone and one or more surfactants are mixed and granulated, followed by the addition of aerosil and Compound I.
  • the mixture which may contain for example 5% by weight of Compound I, is then milled.
  • the mixture is then subject to extrusion, and the extrudate thus produced can be milled and sieved for further processing to make capsules or tablets.
  • Surfactant(s) employed in this example can also be added through liquid dosing during extrusion.
  • the approach of solvent evaporation, via spray-drying provides the advantage of allowing for proccessability at lower temperatures, if needed, and allows for other modifications to the process in order to further improve powder properties.
  • the spray-dried powder can then be formulated further, if needed, and final drug product is flexible with regards to whether capsule, tablet or any other solid dosage form is desired.
  • Spray-drying processes and spray-drying equipment are described in K. Masters, S PRAY D RYING H ANDBOOK (Halstead Press, New York, 4 th ed., 1985).
  • Non-limiting examples of spray-drying devices that are suitable for the present invention include spray dryers manufactured by Niro Inc. or GEA Process Engineering Inc., Buchi Labortechnik AG, and Spray Drying Systems, Inc.
  • a spray-drying process generally involves breaking up a liquid mixture into small droplets and rapidly removing solvent from the droplets in a container (spray drying apparatus) where there is a strong driving force for evaporation of solvent from the droplets.
  • Atomization techniques include, for example, two-fluid or pressure nozzles, or rotary atomizers.
  • the strong driving force for solvent evaporation can be provided, for example, by maintaining the partial pressure of solvent in the spray drying apparatus well below the vapor pressure of the solvent at the temperatures of the drying droplets. This may be accomplished by either (1) maintaining the pressure in the spray drying apparatus at a partial vacuum; (2) mixing the liquid droplets with a warm drying gas (e.g., heated nitrogen); or (3) both.
  • a warm drying gas e.g., heated nitrogen
  • the temperature and flow rate of the drying gas, as well as the spray dryer design, can be selected so that the droplets are dry enough by the time they reach the wall of the apparatus. This help to ensure that the dried droplets are essentially solid and can form a fine powder and do not stick to the apparatus wall.
  • the spray-dried product can be collected by removing the material manually, pneumatically, mechanically or by other suitable means. The actual length of time to achieve the preferred level of dryness depends on the size of the droplets, the formulation, and spray dryer operation. Following the solidification, the solid powder may stay in the spray drying chamber for additional time (e.g., 5-60 seconds) to further evaporate solvent from the solid powder.
  • the final solvent content in the solid dispersion as it exits the dryer is preferably at a sufficiently low level so as to improve the stability of the final product.
  • the residual solvent content of the spray-dried powder can be less than 2% by weight.
  • the residual solvent content is within the limits set forth in the international Conference on Harmonization (ICH) Guidelines.
  • Methods to further lower solvent levels include, but are not limited to, fluid bed drying, infra-red drying, tumble drying, vacuum drying, and combinations of these and other processes.
  • the spray dried product contains a solid dispersion, preferably a solid solution, of the active ingredient(s) in a matrix comprised of the hydrophilic polymer(s) and optionally the pharmaceutically acceptable surfactant(s).
  • a pharmaceutically acceptable surfactant described above can be added to and blended with the spray-dried product before further processing.
  • the active ingredient(s) e.g., Compound I, or a combination of Compound I and at least another anti-HCV agent
  • the hydrophilic polymer(s) as well as other optional active ingredients or excipients such as the pharmaceutically acceptable surfactant(s)
  • Suitable solvents include, but are not limited to, alkanols (e.g., methanol, ethanol, 1-propanol, 2-propanol or mixtures thereof), acetone, acetone/water, alkanol/water mixtures (e.g., ethanol/water mixtures), or combinations thereof.
  • the solution can also be preheated before being fed into the spray dryer.
  • the solid dispersion produced by melt-extrusion, spray-drying or other techniques can be prepared into any suitable solid oral dosage forms.
  • the solid dispersion prepared by melt-extrusion, spray-drying or other techniques e.g., the extrudate or the spray-dried powder
  • the solid dispersion can be either directly compressed, or milled or ground to granules or powders before compression. Compression can be done in a tablet press, such as in a steel die between two moving punches.
  • a solid composition of the present invention comprises Compound I and another anti-HCV agent, it is possible to separately prepare solid dispersions of each individual active ingredient and then blend the optionally milled or ground solid dispersions before compacting.
  • Compound I and other active ingredient(s) can also be prepared in the same solid dispersion, optionally milled and/or blended with other additives, and then compressed into tablets.
  • At least one additive selected from flow regulators, binders, lubricants, fillers, disintegrants, or plasticizers may be used in compressing the solid dispersion. These additives can be mixed with ground or milled solid dispersion before compacting. Disintegrants promote a rapid disintegration of the compact in the stomach and keeps the liberated granules separate from one another.
  • suitable disintegrants are cross-linked polymers such as cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethylcellulose or sodium croscarmellose.
  • Non-limiting examples of suitable fillers are lactose monohydrate, calcium hydrogenphosphate, microcrystalline cellulose (e.g., Avicell), silicates, in particular silicium dioxide, magnesium oxide, talc, potato or corn starch, isomalt, or polyvinyl alcohol.
  • suitable flow regulators include highly dispersed silica (e.g., colloidal silica such as Aerosil), and animal or vegetable fats or waxes.
  • suitable lubricants include polyethylene glycol (e.g., having a molecular weight of from 1000 to 6000), magnesium and calcium stearates, sodium stearyl fumarate, and the like.
  • additives may also be used in preparing a solid composition of the present invention, 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.
  • Solid compositions according to certain embodiments of the present invention may contain 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.
  • the dosage form In order to facilitate the intake of a solid dosage form, 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 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. polysorbates, and optionally a pigment, e.g. titanium dioxide or iron oxides.
  • the film-coating may also comprise talc as anti-adhesive.
  • the film coat accounts for less than 5% by weight of a pharmaceutical composition of the present invention.
  • the present invention feature methods of using solid compositions of the present invention to treat HIV infection.
  • the methods comprise administering a solid composition of the present invention to a patient in need thereof.
  • a solid composition of the present invention can be administered either alone, or in combination with one or more other anti-HCV agents, such as those described hereinabove.
  • the specific inhibitory dose for any particular patient will depend upon a variety of factors including the severity of the HCV infection; the activity of Compound I in the particular patient; the specific solid composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration and rate of excretion; the duration of the treatment; drugs used in combination or coincidental with Compound I; and like factors well known in the medical arts.
  • a method of the present invention comprises administering to a patient in need thereof a solid composition of the present invention and at least another anti-HCV agent, wherein said another anti-HCV agent is selected from HCV polymerase inhibitors (e.g., nucleoside or non-nucleoside HCV polymerase inhibitors), HCV protease inhibitors, HCV helicase inhibitors, CD81 inhibitors, cyclophilin inhibitors, internal ribosome entry site inhibitors, or HCV NS5A inhibitors.
  • said another anti-HCV agent is an HCV polymerase inhibitor (e.g., nucleoside or non-nucleoside HCV polymerase inhibitor) or an HCV NS5A inhibitor.
  • said another anti-HCV agent is interferon or ribavirin, or preferably a combination thereof.
  • the interferon preferably is ⁇ -interferon, and more preferably, pegylated interferon- ⁇ such as PEGASYS (peginterferon alfa-2a).
  • PEGASYS peginterferon alfa-2a
  • the present invention also features use of a solid composition of the present invention for the manufacture of medicaments for the treatment of HCV infection.
  • Compound I was extruded using melt-extrusion. Two extrudates were prepared, and then milled and filled into capsules.
  • the 1st extrudate contained Compound I, copovidone, and Vitamin E-TPGS in a weight ratio of 5:88:7 (hereinafter Formulation 1).
  • the 2nd extrudate contained Compound I, copovidone and Sorbitan monolaurate in a weight ratio of 5:90:5 (hereinafter Formulation 2).
  • the extrusion mixtures were prepared by use of mortar and pestle. Both formulations were extruded at 140° C. The obtained extruded strands were milled and the fractions of over 0.2 mm were combined with 100 mg mannite/collidal silica (99:1) and then filled into capsules. Each of these extrudate capsules contained 5 mg Compound I.
  • Example 1 The pharmacokinetic profile of each formulation described in Example 1 was evaluated in dogs after single oral (PO) administration of the formulation.
  • dogs Four dogs (two male and two female dogs) were used in this study. The animals were fasted overnight and received food 30 min prior to dosing and throughout the duration of the study.
  • Plasma samples were collected at 0.25, 0.5, 1, 2, 4, 6, 8, 12 and 24 hours post-dose administration. Plasma samples were analyzed for Compound I by LC-MS/MS. AUC 0-inf and Cmax were normalized to a dose of 0.5 mg/kg Compound I.
  • Mean dose-normalized AUC 0-inf values of Compound I were 512.2 and 432.0 ng ⁇ h/ml, at a 0.5 mg/kg dose in Formulations 1 and 2, respectively.
  • Mean dose-normalized C max values of Compound I were 36.1 and 15.2 ng/ml at a 0.5 mg/kg dose in Formulations 1 and 2, respectively.
  • Compound I was mixed with hydrophilic polymers and pharmaceutically acceptable surfactants at various ratios, and dissolved in an organic solvent (acetone or acetone/water mixtures). The solvent was then removed from the system under heat ( ⁇ 75° C.) and vacuum, using a Genevac rotary evaporator or Buchi Rotavap. Solid dispersions of Compound I at various drug loading levels and using different surfactants or polymers were sieved through a 30 mesh screen to reduce particle size. The resultant solid dispersion samples were used for amorphous characterization by X-ray powder diffraction (RXRD), chemical stability, in-vitro dissolution test and dog bioavailability studies.
  • RXRD X-ray powder diffraction
  • the solid dispersion powder was mixed with other excipients and compressed into tablets to achieve strengths of 0.5 mg, 5.0 mg, and 25.0 mg.
  • the release of Compound I was evaluated.
  • hydrophilic polymers employed were copovidone, soluplus, hydroxypropyl methylcellulose phthalate (HPMCP), and hydroxypropyl methylcellulose grade E5 (HPMC-E5).
  • the surfactants employed were Vitamin E TPGS and Cremaphor RH40.
  • the amount of the surfactant(s) in each solid dispersion was no more than 10% by weight, and the amount of Compound I in each solid dispersion ranged from 5 to 20% by weight.
  • One solid dispersion formulation was prepared using spray-drying to produce a solid dispersion powder of amorphous Compound I within a polymer matrix.
  • the spray dried powder contained 10% by weight of Compound I, 85% by weight of copovidone, and 5% by weight of Vitamin E TPGS. Acetone and water in a 9:1 ratio was used as a solvent for spray-drying.
  • the spray dried powder was further dried under vacuum to remove residual solvent.
  • the vacuum dried powder was blended with microcrystalline cellulose, lactose monohydrate, colloidal silicon dioxide, sodium stearyl fumarate, and optionally croscarmellose sodium. This blend was then compressed into the final tablet dosage form.

Abstract

The present invention features solid compositions comprising amorphous Compound I. For instance, Compound I may be formulated in an amorphous solid dispersion which comprises a pharmaceutically acceptable hydrophilic polymer and preferably a pharmaceutically acceptable surfactant.

Description

  • This application is a divisional of U.S. patent application Ser. No. 13/156,783, filed Jun. 9, 2011, which claims the benefit of U.S. Provisional Application Ser. No. 61/353,553, filed Jun. 10, 2010, all of which are incorporated herein by reference in their entireties.
  • FIELD OF THE INVENTION
  • The present invention relates to solid compositions comprising anti-HCV compounds and methods of using the same to treat HCV infection.
  • BACKGROUND
  • The hepatitis C virus (HCV) is an RNA virus belonging to the Hepacivirus genus in the Flaviviridae family. The enveloped HCV virion contains a positive stranded RNA genome encoding all known virus-specific proteins in a single, uninterrupted, open reading frame. The open reading frame comprises approximately 9500 nucleotides and encodes a single large polyprotein of about 3000 amino acids. The polyprotein comprises a core protein, envelope proteins E1 and E2, a membrane bound protein p7, and the non-structural proteins NS2, NS3, NS4A, NS4B, NS5A and NS5B.
  • HCV infection is associated with progressive liver pathology, including cirrhosis and hepatocellular carcinoma. Chronic hepatitis C may be treated with peginterferon-alpha in combination with ribavirin. Substantial limitations to efficacy and tolerability remain as many users suffer from side effects, and viral elimination from the body is often inadequate. Therefore, there is a need for new drugs to treat HCV infection.
  • SUMMARY OF THE INVENTION
  • The present invention features solid compositions comprising dimethyl(2S,2′S)-1,1′-((2S,2′S)-2,2′-(4,4′-((2S,5S)-1-(4-tert-butylphenyl)pyrrolidine-2,5-diyl)bis(4,1-phenylene))bis(azanediyl)bis(oxomethylene)bis(pyrrolidine-2,1-diyl))bis(3-methyl-1-oxobutane-2,1-diyl)dicarbamate (
  • Figure US20140171481A1-20140619-C00001
  • or a pharmaceutically acceptable salt thereof. Compound I is a potent HCV inhibitor. The solid compositions of the invention comprise (1) Compound I (or a pharmaceutically acceptable salt thereof) in an amorphous form, (2) a pharmaceutically acceptable hydrophilic polymer, and optionally (3) a pharmaceutically acceptable surfactant.
  • In one aspect, the present invention features a solid composition comprising a solid dispersion, wherein the solid dispersion comprises Compound I (or a pharmaceutically acceptable salt thereof) in an amorphous form and a pharmaceutically acceptable hydrophilic polymer, and the solid composition further comprises a pharmaceutically acceptable surfactant. The surfactant can be, without limitation, either formulated in the solid dispersion or separately combined or mixed with the solid dispersion. Preferably, the hydrophilic polymer has a Tg of at least 50° C. More preferably, the hydrophilic polymer has a Tg of at least 80° C. Highly preferably, the hydrophilic polymer has a Tg of at least 100° C. Also preferably, the surfactant has a HLB value of at least 10. Hydrophilic polymers with Tg of at least 25° C. can also be used.
  • In one embodiment of this aspect of the invention, the hydrophilic polymer is selected from homopolymer of N-vinyl lactam, copolymer of N-vinyl lactam, cellulose ester, cellulose ether, polyalkylene oxide, polyacrylate, polymethacrylate, polyacrylamide, polyvinyl alcohol, vinyl acetate polymer, oligosaccharide, or polysaccharide. Non-limiting examples of suitable hydrophilic polymers include homopolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone and vinyl acetate, copolymer of N-vinyl pyrrolidone and vinyl propionate, polyvinylpyrrolidone, methylcellulose, ethylcellulose, hydroxyalkylcelluloses, hydroxypropylcellulose, hydroxyalkylalkylcellulose, hydroxypropylmethylcellulose, cellulose phthalate, cellulose succinate, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, hydroxypropylmethylcellulose acetate succinate, polyethylene oxide, polypropylene oxide, copolymer of ethylene oxide and propylene oxide, methacrylic acid/ethyl acrylate copolymer, methacrylic acid/methyl methacrylate copolymer, butyl methacrylate/2-dimethylaminoethyl methacrylate copolymer, poly(hydroxyalkyl acrylate), poly(hydroxyalkyl methacrylate), copolymer of vinyl acetate and crotonic acid, partially hydrolyzed polyvinyl acetate, carrageenan, galactomannan, or xanthan gum.
  • In another embodiment of this aspect of the invention, the surfactant is selected from polyoxyethylene castor oil derivates, mono fatty acid ester of polyoxyethylene sorbitan, polyoxyethylene alkyl ether, polyoxyethylene alkylaryl ether, polyethylene glycol fatty acid ester, alkylene glycol fatty acid mono ester, sucrose fatty acid ester, or sorbitan fatty acid mono ester. Non-limiting examples of suitable surfactants include polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate such as polyethylenglycol 40 hydrogenated castor oil (Cremophor® RH 40, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate) or polyethylenglycol 60 hydrogenated castor oil (Cremophor® RH 60), mono fatty acid ester of polyoxyethylene sorbitan, such as 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) or polyoxyethylene (20) sorbitan monolaurate (Tween® 20), polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether, polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether, polyoxyethylene (3) octylphenyl ether, PEG-200 monolaurate, PEG-200 dilaurate, PEG-300 dilaurate, PEG-400 dilaurate, PEG-300 distearate, PEG-300 dioleate, propylene glycol monolaurate, sucrose monostearate, sucrose distearate, sucrose monolaurate, sucrose dilaurate, sorbitan mono laurate, sorbitan monooleate, sorbitan monopalnitate, or sorbitan stearate.
  • In yet another embodiment, the solid dispersion is an amorphous solid dispersion. In still another embodiment, the solid dispersion is an amorphous solid dispersion which comprises Compound I (or a pharmaceutically acceptable salt thereof), the hydrophilic polymer, and the surfactant. In a further embodiment, the solid dispersion is a solid solution comprising Compound I (or a pharmaceutically acceptable salt thereof) and the hydrophilic polymer. In yet another embodiment, the solid dispersion is a solid solution comprising Compound I (or a pharmaceutically acceptable salt thereof), the hydrophilic polymer and the surfactant.
  • In yet another embodiment of this aspect of the invention, the hydrophilic polymer is a homopolymer or copolymer of N-vinyl pyrrolidone. Preferably, the hydrophilic polymer is copovidone.
  • In still another embodiment, the surfactant is D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS).
  • In still yet another embodiment, the surfactant is a sorbitan fatty acid ester, such as sorbitan mono laurate (Span® 20).
  • In yet another embodiment, a solid composition of the invention comprises an amorphous solid dispersion or a solid solution which comprises Compound I (or a pharmaceutically acceptable salt thereof), copovidone, and a surfactant selected from vitamin E TPGS, Span 20, or a combination thereof.
  • In another aspect, the present invention features processes of making a solid composition of the present invention. In one embodiment, the process comprises drying a solvent in a liquid solution, wherein said solution comprises: (1) Compound I or a pharmaceutically acceptable salt thereof; (2) a pharmaceutically acceptable hydrophilic polymer; and optionally (3) a pharmaceutically acceptable surfactant. The drying process can be carried out using any suitable solvent evaporation techniques including but not limited to spray-drying techniques.
  • In another embodiment, the process comprises solidifying a melt which comprises: (1) Compound I or a pharmaceutically acceptable salt thereof; (2) a pharmaceutically acceptable hydrophilic polymer; and optionally (3) a pharmaceutically acceptable surfactant.
  • A solid composition of the invention may also contain other additives or ingredients, such as coloring agents, flavoring agents, lubricants or preservatives. A solid composition of the invention can be prepared into any suitable dosage forms, such as capsule, dragee, granule, powder, or tablet.
  • A solid composition of the invention may further comprise another anti-HCV agent, for example, an agent selected from HCV helicase inhibitors, HCV polymerase inhibitors. HCV protease inhibitors, HCV NS5A inhibitors, CD81 inhibitors, cyclophilin inhibitors, or internal ribosome entry site (IRES) inhibitors.
  • The present invention further features methods of using a solid composition of the present invention to treat HCV infection. The methods comprise administering a solid composition of the present invention to a patient in need thereof, thereby reducing the blood or tissue level of HCV virus in the patient.
  • Other features, objects, and advantages of the present invention are apparent in the detailed description that follows. It should be understood, however, that the detailed description, while indicating preferred embodiments of the invention, are given by way of illustration only, not limitation. Various changes and modifications within the scope of the invention will become apparent to those skilled in the art from the detailed description.
  • DETAILED DESCRIPTION
  • The present invention features solid compositions comprising amorphous Compound I (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable hydrophilic polymer, and optionally a pharmaceutically acceptable surfactant. Formulating Compound I in an amorphous form can increase the inherent drug solubility and dissolution rate, thereby enhancing the bioavailability of the compound.
  • A non-limiting way to form an amorphous form of Compound I is through the formation of solid dispersions with a polymeric carrier. The presence of hydrophilic polymer(s) and optional surfactant(s), as well as the dispersion of Compound I in an amorphous form in a matrix containing the polymer(s), can significantly enhance the dissolution rate of Compound I. In some cases, a solid dispersion formulation can also effectively maintain Compound I in its supersaturation state to allow for better absorption.
  • As used herein, the term “solid dispersion” defines a system in a solid state (as opposed to a liquid or gaseous state) comprising at least two components, wherein one component is dispersed throughout the other component or components. For example, an active ingredient or a combination of active ingredients can be dispersed in a matrix comprised of a pharmaceutically acceptable hydrophilic polymer(s) and a pharmaceutically acceptable surfactant(s). The term “solid dispersion” encompasses systems having small particles of one phase dispersed in another phase. These particles are often of less than 400 μm in size, such as less than 100, 10, or 1 μm in size. When a solid dispersion of the components is such that the system is chemically and physically uniform or homogenous throughout or consists of one phase (as defined in thermodynamics), such a solid dispersion is called a “solid solution.” A glassy solution is a solid solution in which a solute is dissolved in a glassy solvent.
  • The term AUC or AUC0-inf refers to the area under the plasma concentration time curve (AUC) extrapolated to infinity.
  • The terms “weight percent” or “percent by weight” or “% by weight” or “wt %” denote the weight of an individual component in a composition or mixture as a percentage of the weight of the composition or mixture.
  • In one aspect, the present invention features a solid composition comprising Compound I (or a pharmaceutically acceptable salt thereof) in an amorphous form, a pharmaceutically acceptable hydrophilic polymer, and a pharmaceutically acceptable surfactant. The Compound I (or the salt thereof) and the polymer are formulated in a solid dispersion. The surfactant may also be formulated in the same solid dispersion; or the surfactant can be separately combined or mixed with the solid dispersion.
  • In one embodiment, a solid composition of the invention comprises an amorphous solid dispersion which comprises Compound I (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable hydrophilic polymer and a pharmaceutically acceptable surfactant. In another embodiment, a solid composition of the invention comprises a solid solution which comprises Compound I (or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable hydrophilic polymer. In still another embodiment, a solid composition of the invention comprises a solid solution which comprises Compound I (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable hydrophilic polymer and a pharmaceutically acceptable surfactant. In yet another embodiment, a solid composition of the invention comprises a glassy solution which includes Compound I (or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable hydrophilic polymer. In a further embodiment, a solid composition of the invention comprises a glassy solution which includes Compound I (or a pharmaceutically acceptable salt thereof), a pharmaceutically acceptable hydrophilic polymer and a pharmaceutically acceptable surfactant.
  • A solid composition (or a solid dispersion) of the invention can contain, for example, at least 1% by weight of Compound I, preferably at least 5%, including, e.g., at least 10%. For instance, a solid composition (or a solid dispersion) of the invention can contain from 1 to 50% by weight of Compound I. For another instance, a solid composition (or a solid dispersion) of the invention can contain from 5 to 30% by weight of Compound I. Preferably, a solid composition (or a solid dispersion) of the invention contains from 5 to 15% by weight of Compound I.
  • A solid dispersion of the invention may contain at least 30% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such hydrophilic polymers. Preferably, the solid dispersion contains at least 40% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such hydrophilic polymers. More preferably, the solid dispersion contains at least 50% (including, e.g., at least 60%, 70%, 80% or 90%) by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers. A solid dispersion of the invention may also contain at least 1% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. Preferably, the solid dispersion contains at least 2% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. More preferably, the solid dispersion contains from 4% to 20% by weight of the surfactant(s), such as from 5% to 10% by weight of the surfactant(s).
  • In one embodiment, a solid dispersion of the invention comprises at least 30% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and at least 1% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. In another embodiment, a solid dispersion of the invention comprises at least 50% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 2% to 20% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. In yet another embodiment, a solid dispersion of the invention comprises from 50% to 90% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 3% to 15% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. In yet another embodiment, a solid dispersion of the invention comprises from 70% to 90% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 5% to 10% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants.
  • Preferably, a hydrophilic polymer employed in the present invention has a Tg of at least 50° C., more preferably at least 60° C., and highly preferably at least 80° C. including, but not limited to from, 80° C. to 180° C., or from 100° C. to 150° C. Methods for determining Tg values of 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., the polymer Tg=Σ Wi·Xi where Wi is the weight percent of monomer i in the organic polymer, and Xi 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). Hydrophilic polymers with a Tg as described above may allow for the preparation of solid dispersions that are mechanically stable and, within ordinary temperature ranges, sufficiently temperature stable so that the solid dispersions may be used as dosage forms without further processing or be compacted to tablets with only a small amount of tabletting aids. Hydrophilic polymers having a Tg of below 50° C. may also be used.
  • Preferably, a hydrophilic polymer employed in the present invention is water-soluble. A solid composition of the present invention can also comprise poorly water-soluble or water-insoluble polymer or polymers, such as cross-linked polymers. A hydrophilic polymer comprised in a solid composition of the present invention preferably has an apparent viscosity, when dissolved at 20° C. in an aqueous solution at 2% (w/v), of 1 to 5000 mPa·s., and more preferably of 1 to 700 mPa·s, and most preferably of 5 to 100 mPa·s.
  • Hydrophilic polymers suitable for use in a solid composition of the invention include, but are not limited to, homopolymers or copolymers of N-vinyl lactams, such as homopolymers or copolymers of N-vinyl pyrrolidone (e.g., polyvinylpyrrolidone (PVP), or copolymers of N-vinyl pyrrolidone and vinyl acetate or vinyl propionate); cellulose esters or cellulose ethers, such as alkylcelluloses (e.g., methylcellulose or ethylcellulose), hydroxyalkylcelluloses (e.g., hydroxypropylcellulose), hydroxyalkylalkylcelluloses (e.g., hydroxypropylmethylcellulose), and cellulose phthalates or succinates (e.g., cellulose acetate phthalate and hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, or hydroxypropylmethylcellulose acetate succinate); high molecular polyalkylene oxides, such as polyethylene oxide, polypropylene oxide, and copolymers of ethylene oxide and propylene oxide; polyacrylates or polymethacrylates, such as methacrylic acid/ethyl acrylate copolymers, methacrylic acid/methyl methacrylate copolymers, butyl methacrylate/2-dimethylaminoethyl methacrylate copolymers, poly(hydroxyalkyl acrylates), and poly(hydroxyalkyl methacrylates); polyacrylamides; vinyl acetate polymers, such as copolymers of vinyl acetate and crotonic acid, and partially hydrolyzed polyvinyl acetate (also referred to as partially saponified “polyvinyl alcohol”); polyvinyl alcohol; oligo- or polysaccharides, such as carrageenans, galactomannans, and xanthan gum; polyhydroxyalkylacrylates; polyhydroxyalkyl-methacrylates; copolymers of methyl methacrylate and acrylic acid; polyethylene glycols (PEGs); or any mixture thereof.
  • Non-limiting examples of preferred hydrophilic polymers for the invention include polyvinylpyrrolidone (PVP) K17, PVP K25, PVP K30, PVP K90, hydroxypropyl methylcellulose (HPMC) E3, HPMC E5, HPMC E6, HPMC E15, HPMC K3, HPMC A4, HPMC A15, HPMC acetate succinate (AS) LF, HPMC AS MF, HPMC AS HF, HPMC AS LG, HPMC AS MG, HPMC AS HG, HPMC phthalate (P) 50, HPMC P 55, Ethocel 4, Ethocel 7, Ethocel 10, Ethocel 14, Ethocel 20, copovidone (vinylpyrrolidone-vinyl acetate copolymer 60/40), polyvinyl acetate, methacrylate/methacrylic acid copolymer (Eudragit) L100-55, Eudragit L100, Eudragit S100, polyethylene glycol (PEG) 400, PEG 600, PEG 1450, PEG 3350, PEG 4000, PEG 6000, PEG 8000, poloxamer 124, poloxamer 188, poloxamer 237, poloxamer 338, and poloxamer 407.
  • Of these, homopolymers or copolymers of N-vinyl pyrrolidone, such as copolymers of N-vinyl pyrrolidone and vinyl acetate, are preferred. A non-limiting example of a preferred polymer is a copolymer of 60% by weight of N-vinyl pyrrolidone and 40% by weight of vinyl acetate. Other preferred polymers include, without limitation, hydroxypropyl methylcellulose (HPMC, also known as hypromellose in USP), such as hydroxypropyl methylcellulose grade E5 (HPMC-E5); and hydroxypropyl methylcellulose acetate succinate (HPMC-AS).
  • A pharmaceutically acceptable surfactant employed in the present invention is preferably a non-ionic surfactant. More preferably, a solid composition of the present invention comprises a pharmaceutically acceptable surfactant having an HLB value of from 2-20. A solid composition of the present invention can also include a mixture of pharmaceutically acceptable surfactants, with at least one surfactant having an HLB value of no less than 10 and at least another surfactant having an HLB value of below 10. In one example, each surfactant comprised in a solid composition of the invention has an HLB value of at least 10. In another example, each surfactant comprised in a solid composition of the invention has an HLB value of below 10. In yet another example, a solid composition of the present invention includes at least two pharmaceutically acceptable surfactants, one having an HLB value of at least 10 and the other having an HLB value of below 10. 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.
  • Non-limiting examples of pharmaceutically acceptable surfactants that are suitable for the present invention include 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, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate) or polyethylenglycol 60 hydrogenated castor oil (Cremophor® RH 60); or a mono fatty acid ester of polyoxyethylene sorbitan, such as 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), or polyoxyethylene (20) sorbitan monolaurate (Tween® 20). Other non-limiting examples of suitable surfactants include 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, PEG-400 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; sorbitan fatty acid mono esters such as sorbitan mono laurate (Span® 20), sorbitan monooleate, sorbitan monopalnitate (Span® 40), or sorbitan stearate. Other suitable surfactants include, but are not limited to, 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, or Poloxamer® 407 (BASE Wyandotte Corp.). As described above, a mixture of surfactants can be used in a solid composition of the present invention.
  • Non-limiting examples of preferred surfactants for the invention include to polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, Cremophor RH 40, Cremophor EL, Gelucire 44/14, Gelucire 50/13, D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS), propylene glycol laurate, sodium lauryl sulfate, and sorbitan monolaurate.
  • In one embodiment, a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes Compound I (or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable hydrophilic polymer. The solid composition also includes a pharmaceutically acceptable surfactant which preferably is formulated in the amorphous solid dispersion or solid solution. The hydrophilic polymer can be selected, for example, from the group consisting of homopolymer of N-vinyl lactam, copolymer of N-vinyl lactam, cellulose ester, cellulose ether, polyalkylene oxide, polyacrylate, polymethacrylate, polyacrylamide, polyvinyl alcohol, vinyl acetate polymer, oligosaccharide, and polysaccharide. As a non-limiting example, the hydrophilic polymer is selected from the group consisting of homopolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone and vinyl acetate, copolymer of N-vinyl pyrrolidone and vinyl propionate, polyvinylpyrrolidone, methylcellulose, ethylcellulose, hydroxyalkylcelluloses, hydroxypropylcellulose, hydroxyalkylalkylcellulose, hydroxypropylmethylcellulose, cellulose phthalate, cellulose succinate, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, hydroxypropylmethylcellulose acetate succinate, polyethylene oxide, polypropylene oxide, copolymer of ethylene oxide and propylene oxide, methacrylic acid/ethyl acrylate copolymer, methacrylic acid/methyl methacrylate copolymer, butyl methacrylate/2-dimethylaminoethyl methacrylate copolymer, poly(hydroxyalkyl acrylate), poly(hydroxyalkyl methacrylate), copolymer of vinyl acetate and crotonic acid, partially hydrolyzed polyvinyl acetate, carrageenan, galactomannan, and xanthan gum. Preferably, the hydrophilic polymer is selected from polyvinylpyrrolidone (PVP) K17, PVP K25, PVP K30, PVP K90, hydroxypropyl methylcellulose (HPMC) E3, HPMC E5, HPMC E6, HPMC E15, HPMC K3, HPMC A4, HPMC A15, HPMC acetate succinate (AS) LF, HPMC AS MF, HPMC AS HF, HPMC AS LG, HPMC AS MG, HPMC AS HG, HPMC phthalate (P) 50, HPMC P 55, Ethocel 4, Ethocel 7, Ethocel 10, Ethocel 14, Ethocel 20, copovidone (vinylpyrrolidone-vinyl acetate copolymer 60/40), polyvinyl acetate, methacrylate/methacrylic acid copolymer (Eudragit) L100-55, Eudragit L100, Eudragit S100, polyethylene glycol (PEG) 400, PEG 600, PEG 1450, PEG 3350, PEG 4000, PEG 6000, PEG 8000, poloxamer 124, poloxamer 188, poloxamer 237, poloxamer 338, or poloxamer 407. More preferably, the hydrophilic polymer is selected from homopolymers of vinylpyrrolidone (e.g., PVP with Fikentscher K values of from 12 to 100, or PVP with Fikentscher K values of from 17 to 30), or copolymers of 30 to 70% by weight of N-vinylpyrrolidone (VP) and 70 to 30% by weight of vinyl acetate (VA) (e.g., a copolymer of 60% by weight VP and 40% by weight VA). The surfactant can be selected, for example, from the group consisting of polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate, mono fatty acid ester of polyoxyethylene sorbitan, polyoxyethylene alkyl ether, polyoxyethylene alkylaryl ether, polyethylene glycol fatty acid ester, alkylene glycol fatty acid mono ester, sucrose fatty acid ester, and sorbitan fatty acid mono ester. As a non-limited example, the surfactant is selected from the group consisting of polyethylenglycol 40 hydrogenated castor oil (Cremophor® RE 40, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate), polyethylenglycol 60 hydrogenated castor oil (Cremophor® RH 60), 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), or polyoxyethylene (20) sorbitan monolaurate (Tween® 20)), polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether, polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether, polyoxyethylene (3) octylphenyl ether, PEG-200 monolaurate, PEG-200 dilaurate, PEG-300 dilaurate, PEG-400 dilaurate, PEG-300 distearate, PEG-300 dioleate, propylene glycol monolaurate, sucrose monostearate, sucrose distearate, sucrose monolaurate, sucrose dilaurate, sorbitan monolaurate, sorbitan monooleate, sorbitan monopalnitate, and sorbitan stearate. Preferably, the surfactant is selected from polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, Cremophor RH 40, Cremophor EL, Gelucire 44/14, Gelucire 50/13, D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS), propylene glycol laurate, sodium lauryl sulfate, or sorbitan monolaurate. More preferably, the surfactant is selected from sorbitan monolaurate or D-alpha-tocopheryl polyethylene glycol 1000 succinate.
  • In another embodiment, a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes Compound I (or a pharmaceutically acceptable salt thereof) and a homopolymer or copolymer of N-vinyl pyrrolidone (e.g., copovidone). The solid composition also comprises a pharmaceutically acceptable surfactant (e.g., vitamin E TPGS, or sorbitan monolaurate), wherein the surfactant preferably is formulated in the amorphous solid dispersion or solid solution.
  • In yet another embodiment, a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes Compound I (or a pharmaceutically acceptable salt thereof), copovidone, and a pharmaceutically acceptable surfactant selected from vitamin E TPGS or sorbitan monolaurate. The amorphous solid dispersion or solid solution may also include another pharmaceutically acceptable surfactant.
  • A solid dispersion employed in the present invention preferably comprises or consists of a single-phase (defined in thermodynamics) which the therapeutic agent(s) (e.g., Compound I, or a combination of Compound I and another anti-HCV agent) is molecularly dispersed in a matrix containing the pharmaceutically acceptable hydrophilic polymer(s). In such cases, thermal analysis of the solid dispersion using differential scanning calorimetry (DSC) typically shows only one single Tg, and the solid dispersion does not contain any detectable crystalline Compound I as measured by X-ray powder diffraction spectroscopy.
  • Compound I can be prepared according to the procedures described in U.S. Provisional Patent Application No. 61/243,596, filed Sep. 18, 2009, To a mixture of (S)-2-(methoxycarbonylamino)-3-methylbutanoic acid (19.66 g, 112 mmol) and N-hydroxysuccinimide (13.29 g, 116 mmol) was added ethyl acetate (250 ml), and the mixture was cooled to 0-5° C. Diisopropylcarbodiimide (13.88 g, 110 mmol) was added and the reaction mixture was stirred at 0-5° C. for about 1 hour. The reaction mixture was warmed to room temperature. The solids (diisopropylurea by-product) were filtered and rinsed with ethyl acetate. The filtrate was concentrated in vacuo to an oil. Isopropyl alcohol (200 ml) was added to the oil and the mixture was heated to about 50° C. to obtain a homogeneous solution. Upon cooling, crystalline solids formed. The solids were filtered and washed with isopropyl alcohol (3×20 ml) and dried to give (S)-2,5-dioxopyrrolidin-1-yl 2-(methoxycarbonylamino)-3-methylbutanoate as a white solid (23.2 g, 77% yield).
  • To a mixture of L-proline (4.44 g, 38.6 mmol), water (20 ml), acetonitrile (20 ml) and DIEA (9.5 g, 73.5 mmol) was added a solution of (S)-2,5-dioxopyrrolidin-1-yl 2-(methoxycarbonylamino)-3-methylbutanoate (10 g, 36.7 mmol) in acetonitrile (20 mL) over 10 minutes. The reaction mixture was stirred overnight at room temperature. The solution was concentrated under vacuum to remove the acetonitrile. To the resulting clear water solution was added 6N HCl (9 ml) until pH 2.The solution was transferred to a separatory funnel and 25% NaCl (10 ml) was added and the mixture was extracted with ethyl acetate (75 ml), and then again with ethyl acetate (6×20 ml), and the combined extracts were washed with 25% NaCl (2×10 ml). The solvent was evaporated to give a thick oil. Heptane was added and the solvent was evaporated to give a foam, which was dried under high vacuum. Diethyl ether was added and the solvent was evaporated to give a foam, which was dried under high vacuum to give (S)-1-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)pyrrolidine-2-carboxylic acid (10.67 g) as a white solid.
  • (S)-1-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)pyrrolidine-2-carboxylic acid can also be prepared according to the following procedure. To a flask was charged L-valine (35 g, 299 mmol), 1N sodium hydroxide solution (526 ml, 526 mmol) and sodium carbonate (17.42 g, 164 mmol). The mixture was stirred for 15 min to dissolve solids and then cooled to 15° C. Methyl chloroformate (29.6 g, 314 mmol) was added slowly to the reaction mixture. The mixture was then stirred at rt for 30 min. The mixture was cooled to 15° C. and pH adjusted to ˜5.0 with concentrated HCl solution. 100 mL of 2-methytetrahydrofuran (2-MeTHF) was added and the adjustment of pH continued until the pH reached ˜2.0. 150 mL of 2-MeTHF was added and the mixture was stirred for 15 min. Layers were separated and the aqueous layer extracted with 100 mL of 2-MeTHF. The combined organic layer was dried over anhyd Na2SO4 and filtered, and Na2SO4 cake was washed with 50 mL of 2-MeTHF. The product solution was concentrated to ˜100 mL, chased with 120 mL of IPAc twice. 250 mL of heptanes was charged slowly and then the volume of the mixture was concentrated to 300 mL. The mixture was heated to 45° C. and 160 mL of heptanes charged. The mixture was cooled to rt in 2 h, stirred for 30 min, filtered and washed with 2-MeTHF/heptanes mixture (1:7, 80 mL). The wetcake was dried at 55° C. for 24 h to give 47.1 g of Moc-L-Val-OH product as a white solid (90%). Moc-L-Val-OH (150 g, 856 mmol), HOBt hydrate (138 g, 899 mmol) and DMF (1500 ml) were charged to a flask. The mixture was stirred for 15 min to give a clear solution. EDC hydrochloride (172 g, 899 mmol) was charged and mixed for 20 min. The mixture was cooled to 13° C. and (L)-proline benzyl ester hydrochloride (207 g, 856 mmol) charged. Triethylamine (109 g, 1079 mmol) was then charged in 30 min. The resulting suspension was mixed at rt for 1.5 h. The reaction mixture was cooled to 15° C. and 1500 mL of 6.7% NaHCO3 charged in 1.5 h, followed by the addition of 1200 mL of water over 60 min. The mixture was stirred at rt for 30 min, filtered and washed with water/DMF mixture (1:2, 250 mL) and then with water (1500 mL). The wetcake was dried at 55° C. for 24 h to give 282 g of product as a white solid (90%). The resulting solids (40 g) and 5% Pd/Alumina were charged to a Parr reactor followed by THF (160 mL). The reactor was sealed and purged with nitrogen (6×2.0 psig) followed by a hydrogen purge (6×30 psig). The reactor was pressurized to 30 psig with hydrogen and agitated at room temperature for approximately 15 hours. The resulting slurry was filtered through a GET filter and concentrated to approximately 135 g solution. Heptane was added (120 mL), and the solution was stirred until solids formed. After an addition 2-3 hours additional heptane was added drop-wise (240 mL), the slurry was stirred for approximately 1 hour, then filtered. The solids were dried to afford (S)-1-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)pyrrolidine-2-carboxylic acid.
  • Anhydrous zinc(II) chloride (2.73 g, 20.00 mmol) was stirred in dry benzene (15 ml) while diethylamine (1.558 ml, 15.00 mmol) and t-butanol (1.435 ml, 15.00 mmol) were added, and the resulting mixture was stirred at room temperature for 90 min to give a cloudy solution. To this mixture was added 2-bromo-1-(4-nitrophenyl)ethanone (2.44 g, 10.00 mmol) and 1-(4-nitrophenyl)ethanone (2.477 g, 15.00 mmol), and the resulting mixture was stirred at room temperature overnight. The mixture was poured into water (50 mL) and extracted with ethyl acetate (3×50 ml). The combined organic layers were dried over Na2SO4, filtered and concentrated. The resulting residue was triturated with dichloromethane to give an orange solid that was collected by filtration and dried to give 1,4-Bis(4-nitrophenyl)butane-1,4-dione (2.0 gm, 61% yield).
  • To (S)-(−)-α,α-diphenyl-2-pyrrolidinemethanol (2.71 g, 10.70 mmol) was added THF (80 mL) at 23° C. The very thin suspension was treated with trimethyl borate (1.44 g, 13.86 mmol) over 30 seconds, and the resulting solution was mixed at 23° C. for 1 h. The solution was cooled to 16-19° C., and N,N-diethylaniline borane (21.45 g, 132 mmol) was added dropwise via syringe over 3-5 min (caution: vigorous H2 evolution), while the internal temperature was maintained at 16-19° C. After 15 min, the H2 evolution had ceased. To a separate vessel was added 1,4-Bis(4-nitrophenyl)butane-1,4-dione (22.04 g, 95 wt %, 63.8 mmol), followed by THF (80 mL), to form an orange slurry. After cooling the slurry to 11° C., the borane solution was transferred via cannula into the dime slurry over 3-5 min. During this period, the internal temperature of the slurry rose to 16° C. After the addition was complete, the reaction was maintained at 20-27° C. for an additional 2.5 h. After reaction completion, the mixture was cooled to 5° C. and methanol (16.7 g, 521 mmol) was added dropwise over 5-10 min, maintaining an internal temperature <20° C. (note: vigorous H2 evolution). After the exotherm had ceased (ca. 10 min), the temperature was adjusted to 23° C., and the reaction was mixed until complete dissolution of the solids had occurred. Ethyl acetate (300 mL) and 1 M HCl (120 mL) were added, and the phases were partitioned. The organic phase was then washed successively with 1 M HCl (2×120 mL), H2O (65 mL), and 10% aq. NaCl (65 mL). The organics were dried over MgSO4, filtered, and concentrated in vacuo. Crystallization of the product occurred during the concentration. The slurry was warmed to 50° C., and heptane (250 mL) was added over 15 min. The slurry was then allowed to mix at 23° C. for 30 min and filtered. The wet cake was washed with 3:1 heptane:ethyl acetate (75 mL), and the orange, crystalline solids were dried at 45° C. for 24 h to provide (1R,4R)-1,4-bis(4-nitrophenyl)butane-1,4-diol (15.35 g, 99.3% ee, 61% yield), which was contaminated with 11% of the meso isomer (vs. dl isomer).
  • (1R,4R)-1,4-bis(4-nitrophenyflbutane-1,4-diol (5.01 g, 13.39 mmol) was combined with 2-methyltetrahydrofuran (70 mL) and cooled to −5° C., and N,N-diisopropylethylamine (6.81 g, 52.7 mmol) was added over 30 seconds. Separately, a solution of methanesulfonic anhydride (6.01 g, 34.5 mmol) in 2-methyltetrahydrofuran (30 mL) was prepared and added to the diol slurry over 3 min., maintaining the internal temperature between −15° C. and −25° C. After mixing for 5 min at −15° C., the cooling bath was removed and the reaction was allowed to warm slowly to 23° C. and mixed for 30 minutes. After reaction completion, the crude slurry (1R,4R)-1,4-bis(4-nitrophenyl)butane-1,4-diyl dimethanesulfonate) was carried immediately into the next step.
  • To the crude product solution (1R,4R)-1,4-bis(4-nitrophenyl)butane-1,4-diyl dimethanesulfonate (7.35 g, 13.39 mmol) was added 4-tert-butylaniline (13.4 g, 90 mmol) at 23° C. over 1 minute. The reaction was heated to 65° C. for 2 h. After completion, the reaction mixture was cooled to 23° C. and diluted with 2-methyltetrahydrofuran (100 mL) and 1 M HCl (150 mL). After partitioning the phases, the organic phase was treated with 1 M HCl (140 mL), 2-methyltetrahydrofuran (50 mL), and 25 wt % aq. NaCl (100 mL), and the phases were partitioned. The organic phase was washed with 25 wt % aq. NaCl (50 mL), dried over MgSO4, filtered, and concentrated in vacuo to approximately 20 mL. Heptane (30 mL) and additional 2-methyltetrahydrofuran were added in order to induce crystallization. The slurry was concentrated further, and additional heptane (40 mL) was slowly added and the slurry was filtered, washing with 2-methyltetrahydrofuran:heptane (1:4, 20 mL). The solids were suspended in MeOH (46 mL) for 3 h, filtered, and the wet solid was washed with additional MeOH (18 mL). The solid was dried at 45° C. in a vacuum oven for 16 h to provide (2S,5S)-1-(4-tert-butylphenyl)-2,5-bis(4-nitrophenyl)pyrrolidine (3.08 g, 51% 2-step yield).
  • To a 160 ml Parr stirrer hydrogenation vessel was added (2S,5S)-1-(4-tert-butylphenyl)-2,5-bis(4-nitrophenyl)pyrrolidine (2 g, 4.49 mmol), followed by 60 ml of THF, and Raney Nickel Grace 2800 (1 g, 50 wt % (dry basis)) under a stream of nitrogen. The reactor was assembled and purged with nitrogen (8×20 psig) followed by purging with hydrogen (8×30 psig). The reactor was then pressurized to 30 psig with hydrogen and agitation (700 rpm) began and continued for a total of 16 h at room temperature. The slurry was filtered by vacuum filtration using a GF/F Whatman glass fiber filter. Evaporation of the filtrate to afford a slurry followed by the addition heptane and filtration gave the crude 4,4′-((2S,5S)-1-(4-tert-butylphenyl)pyrrolidine-2,5-diyl)dianiline, which was dried and used directly in the next step.
  • To a solution of 4,4′-((2S,5S)-1-(4-tert-butylphenyl)pyrrolidine-2,5-diyl)dianiline (1.64 g, 4.25 mmol) in DMF (20 ml), (S)-1-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)pyrrolidine-2-carboxylic acid (2.89 g, 10.63 mmol), and HATU (4.04 g, 10.63 mmol) in DMF (150 mL) was added triethylamine (1.07 g, 10.63 mmol), and the solution was stirred at room temperature for 90 min. To the reaction mixture was poured 20 mL of water, and the white precipitate obtained was filtered, and the solid was washed with water (3×5 mL). The solid was blow dried for 1 h. The crude material was loaded on a silica gel column and eluted with a gradient starting with ethyl acetate/heptane (3/7), and ending with pure ethyl acetate. The desired fractions were combined and solvent distilled off to give a very light yellow solid, which was dried at 45° C. in a vacuum oven with nitrogen purge for 15 h to give Compound I (2.3 g, 61% yield).
  • Alternately, 4,4′-((2S,5S)-1-(4-tert-butylphenyl)pyrrolidine-2,5-diyl)dianiline (11.7 g, 85 wt %, 25.8 mmol) and (S)-1-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)pyrrolidine-2-carboxylic acid (15.45 g, 56.7 mmol) are suspended in EtOAc (117 mL), diisopropylethylamine (18.67 g, 144 mmol) is added and the solution is cooled to 0° C. In a separate flask, 1-propanephosphonic acid cyclic anhydride (T3P®) (46.0 g, 50 wt % in EtOAc, 72.2 mmol) was dissolved in EtOAc (58.5 mL), and charged to an addition funnel. The T3P solution is added to the reaction mixture drop-wise over 3-4 h and stirred until the reaction is complete. The reaction is warmed to room temperature, and washed with 1M HCl/7.5 wt % NaCl (100 mL), then washed with 5% NaHCO3 (100 mL), then washed with 5% NaCl solution (100 mL). The solution was concentrated to approximately 60 mL, EtOH (300 mL) was added, and the solution was concentrated to 84 g solution.
  • A portion of the EtOH solution of product (29 g) was heated to 40° C., and added 134 g 40 w % EtOH in H2O. A slurry of seeds in 58 wt/wt % EtOH/H2O was added, allowed to stir at 35-40° C. (e.g., at 35 or 40° C.) for several hours, then cooled to 0° C. The slurry was then filtered, and washed with 58 wt/wt % EtOH/H2O. The product was dried at 40-60° C. (e.g., 40° C.) under vacuum, and then rehydrated by placing a tray of water in the vacuum oven (or in a tray dryer using 40° C. and a humidified atmosphere) to give Compound I in a hydrate crystalline form (Hydrate B).
  • The seeds described above were a EtOH—H2O solvate of Compound I and was originally made according to the following procedure. A solution was first prepared by slurrying the amorphous, solid Compound I in 37 weight % EtOH in heptane at room temperature. The saturated solution (with respect to the amorphous solid) was then filtered over to a new vial to give a clear solution and seeded with partially crystalline solid obtained from the same solvent system. Crystallization took place within a few hours to produce the “Anhydrate A” form of Compound I. Then a solution of 60 w % EtOH in H2O at 5° C. was prepared using the amorphous Compound I. To this clear solution were added seeds of mainly Anhydrate A and desolvated EtOH solvate. Conversion to the EtOH—H2O solvate took place within 2 days to produce the original seeds, which can be used to make additional seeds.
  • A solid composition of the present invention can further include one or more other anti-HCV agents. These other anti-HCV agents can be, for example, HCV polymerase inhibitors (including nucleoside or non-nucleoside type of polymerase inhibitors), HCV protease inhibitors, HCV helicase inhibitors, CD81 inhibitors, cyclophilin inhibitors, internal ribosome entry site inhibitors, or HCV NS5A inhibitors. Specific examples of these other anti-HCV agents include, but are not limited to, ribavirin, α-interferon, β-interferon, pegylated interferon-α, pegylated interferon-lambda, PSI-7851 (Pharmasset) (nucleoside polymerase inhibitor), PSI-938 (Pharmasset) (nucleoside polymerase inhibitor), PF-00868554, ANA-598, IDX184 (nucleoside polymerase inhibitor), IDX102, IDX375 (non-nucleoside polymerase inhibitor), GS-9190 (non-nucleoside polymerase inhibitor), VCH-759, VCH-916, MK-3281, BCX-4678, MK-3281, VBY-708, ANA598, GL59728, GL60667, BMS-790052 (NS5A inhibitor), BMS-791325 (protease Inhibitor), BMS-650032, BMS-824393, GS-9132, ACH-1095 (protease inhibitor), AP-H005, A-831 (Arrow Therapeutics) (NS5A inhibitor), A-689 (Arrow Therapeutics) (NS5A inhibitor), INX08189 (Inhibitex) (polymerase inhibitor), AZD2836, telaprevir (protease Inhibitor), boceprevir (protease Inhibitor), ITMN-191 (Intermune/Roche), BI-201335 (protease Inhibitor), VBY-376, VX-500 (Vertex) (protease Inhibitor), PHX-B, ACH-1625, IDX136, IDX316, VX-813 (Vertex) (protease Inhibitor), SCH 900518 (Schering-Plough), TMC-435 (Tibotec) (protease Inhibitor), ITMN-191 (Intermune, Roche) (protease inhibitor), MK-7009 (Merck) (protease Inhibitor), (Novartis), BI-201335 (Boethringer Ingelheim), R7128 (Roche) (nucleoside polymerase inhibitor), MK-3281 (Merck), MK-0608 (Merck) (nucleoside polymerase inhibitor), PF-868554 (Pfizer) (non-nucleoside polymerase inhibitor), PF-4878691 (Pfizer), IDX-184 (Novartis), IDX-375 (Pharmasset), PPI-461 (Presidio) (NS5A inhibitor), BILB-1941 (Boehringer Ingelheim), GS-9190 (Gilead), BMS-790052 (BMS), Albuferon (Novartis), ABT-450 (Abbott/Enanta) (protease Inhibitor), ABT-333 (Abbott) (non-nucleoside polymerase inhibitor), and ABT-072 (Abbott) (non-nucleoside polymerase inhibitor).
  • In one embodiment, a solid composition of the invention comprises Compound I (a pharmaceutically acceptable salt thereof), and a HCV protease inhibitor. In another embodiment, a solid composition of the invention comprises Compound I (a pharmaceutically acceptable salt thereof), and a HCV polymerase inhibitor (e.g., a non-nucleoside polymerase inhibitor, or preferably a nucleoside polymerase inhibitor). In yet another embodiment, a solid composition of the invention comprises (1) Compound I (a pharmaceutically acceptable salt thereof), (2) a HCV protease inhibitor, and (3) a HCV polymerase inhibitor (e.g., a non-nucleoside polymerase inhibitor, or preferably a nucleoside polymerase inhibitor). Non-limiting examples of protease and polymerase inhibitors are described above.
  • A solid composition of the present invention preferably is a solid oral dosage form. Common solid oral dosage forms suitable for the present invention include, but are not limited to, capsules, dragees, granules, pills, powders and tablets, with capsules and tablets being preferred. A solid oral dosage form of the present invention can also include other excipients or inset diluents, such as sucrose, lactose or starch. Lubricants, coloring agents, releasing agents, coating agents, sweetening or flavoring agents, buffering agents, preservatives, or antioxidants can also be included in a solid oral dosage form of the present invention.
  • A solid composition of the present invention can be prepared by a variety of techniques such as, without limitation, melt-extrusion, spray-drying, co-precipitation, freeze drying, or other solvent evaporation techniques, with melt-extrusion and spray-drying being preferred. The melt-extrusion process typically comprises the steps of preparing a melt which includes the active ingredient(s), the hydrophilic polymer(s) and preferably the surfactant(s), and then 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, preferably homogeneously embedded, in the other component or components. In many cases, the polymer component(s) will melt and the other components including the active ingredient(s) and surfactant(s) will dissolve in the melt thereby forming a solution. Melting usually involves heating above the softening point of the polymer(s). 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. The melt can also be homogenized in order to disperse the active ingredient(s) efficiently. In addition, it may be convenient first to melt the polymer(s) and then to mix in and homogenize the active ingredient(s). In one example, all materials except surfactant(s) are blended and fed into an extruder, while the surfactant(s) is molten externally and pumped in during extrusion.
  • In another example, the melt comprises Compound I and one or more hydrophilic polymers described above, and the melt temperature is in the range of from 100 to 170° C., preferably from 120 to 150° C., and highly preferably from 135 to 140° C. The melt can also include a pharmaceutically acceptable surfactant described above.
  • In still another example, the melt comprises Compound I, at least another anti-HCV agent described above, and one or more hydrophilic polymers described above. The melt can also include a pharmaceutically acceptable surfactant described above.
  • To start a melt-extrusion process, the active ingredient(s) (e.g., Compound I, or a combination of Compound I and at least another anti-HCV agent) can be employed in their solid forms, such as their respective crystalline forms. The active ingredient(s) can also be employed as a solution or dispersion in a suitable liquid solvent such as alcohols, aliphatic hydrocarbons, esters or, in some cases, liquid carbon dioxide. The solvent can be removed, e.g. evaporated, upon preparation of the melt.
  • Various additives can also be included in the melt, for example, flow regulators (e.g., colloidal silica), binders, lubricants, fillers, disintegrants, plasticizers, colorants, or stabilizers (e.g., antioxidants, light stabilizers, radical scavengers, and stabilizers against microbial attack).
  • The melting and/or mixing can take 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 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 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 can range from thin to pasty to viscous. Shaping of the extrudate can be conveniently carried out by a calendar with two counter-rotating rollers with mutually matching depressions on their surface. The extrudate can be cooled and allow to solidify. The extrudate can also be cut into pieces, either before (hot-cut) or after solidification (cold-cut).
  • The solidified extrusion product can be further milled, ground or otherwise reduced to granules. The solidified extrudate, as well as each granule produced, comprises a solid dispersion, preferably a solid solution, of the active ingredient(s) in a matrix comprised of the hydrophilic polymer(s) and optionally the pharmaceutically acceptable surfactant(s). Where the granules do not contain any surfactant, a pharmaceutically acceptable surfactant described above can be added to and blended with the granules. The extrusion product can also be blended with other active ingredient(s) and/or additive(s) before being milled or ground to granules. The granules can be further processed into suitable solid oral dosage forms.
  • In one example, copovidone and one or more surfactants are mixed and granulated, followed by the addition of aerosil and Compound I. The mixture, which may contain for example 5% by weight of Compound I, is then milled. The mixture is then subject to extrusion, and the extrudate thus produced can be milled and sieved for further processing to make capsules or tablets. Surfactant(s) employed in this example can also be added through liquid dosing during extrusion.
  • The approach of solvent evaporation, via spray-drying, provides the advantage of allowing for proccessability at lower temperatures, if needed, and allows for other modifications to the process in order to further improve powder properties. The spray-dried powder can then be formulated further, if needed, and final drug product is flexible with regards to whether capsule, tablet or any other solid dosage form is desired.
  • Exemplary spray-drying processes and spray-drying equipment are described in K. Masters, SPRAY DRYING HANDBOOK (Halstead Press, New York, 4th ed., 1985). Non-limiting examples of spray-drying devices that are suitable for the present invention include spray dryers manufactured by Niro Inc. or GEA Process Engineering Inc., Buchi Labortechnik AG, and Spray Drying Systems, Inc. A spray-drying process generally involves breaking up a liquid mixture into small droplets and rapidly removing solvent from the droplets in a container (spray drying apparatus) where there is a strong driving force for evaporation of solvent from the droplets. Atomization techniques include, for example, two-fluid or pressure nozzles, or rotary atomizers. The strong driving force for solvent evaporation can be provided, for example, by maintaining the partial pressure of solvent in the spray drying apparatus well below the vapor pressure of the solvent at the temperatures of the drying droplets. This may be accomplished by either (1) maintaining the pressure in the spray drying apparatus at a partial vacuum; (2) mixing the liquid droplets with a warm drying gas (e.g., heated nitrogen); or (3) both.
  • The temperature and flow rate of the drying gas, as well as the spray dryer design, can be selected so that the droplets are dry enough by the time they reach the wall of the apparatus. This help to ensure that the dried droplets are essentially solid and can form a fine powder and do not stick to the apparatus wall. The spray-dried product can be collected by removing the material manually, pneumatically, mechanically or by other suitable means. The actual length of time to achieve the preferred level of dryness depends on the size of the droplets, the formulation, and spray dryer operation. Following the solidification, the solid powder may stay in the spray drying chamber for additional time (e.g., 5-60 seconds) to further evaporate solvent from the solid powder. The final solvent content in the solid dispersion as it exits the dryer is preferably at a sufficiently low level so as to improve the stability of the final product. For instance, the residual solvent content of the spray-dried powder can be less than 2% by weight. Highly preferably, the residual solvent content is within the limits set forth in the international Conference on Harmonization (ICH) Guidelines. In addition, it may be useful to subject the spray-dried composition to further drying to lower the residual solvent to even lower levels. Methods to further lower solvent levels include, but are not limited to, fluid bed drying, infra-red drying, tumble drying, vacuum drying, and combinations of these and other processes.
  • Like the solid extrudate described above, the spray dried product contains a solid dispersion, preferably a solid solution, of the active ingredient(s) in a matrix comprised of the hydrophilic polymer(s) and optionally the pharmaceutically acceptable surfactant(s). Where the spray dried product does not contain any surfactant, a pharmaceutically acceptable surfactant described above can be added to and blended with the spray-dried product before further processing.
  • Before feeding into a spray dryer, the active ingredient(s) (e.g., Compound I, or a combination of Compound I and at least another anti-HCV agent), the hydrophilic polymer(s), as well as other optional active ingredients or excipients such as the pharmaceutically acceptable surfactant(s), can be dissolved in a solvent. Suitable solvents include, but are not limited to, alkanols (e.g., methanol, ethanol, 1-propanol, 2-propanol or mixtures thereof), acetone, acetone/water, alkanol/water mixtures (e.g., ethanol/water mixtures), or combinations thereof. The solution can also be preheated before being fed into the spray dryer.
  • The solid dispersion produced by melt-extrusion, spray-drying or other techniques can be prepared into any suitable solid oral dosage forms. In one embodiment, the solid dispersion prepared by melt-extrusion, spray-drying or other techniques (e.g., the extrudate or the spray-dried powder) can be compressed into tablets. The solid dispersion can be either directly compressed, or milled or ground to granules or powders before compression. Compression can be done in a tablet press, such as in a steel die between two moving punches. When a solid composition of the present invention comprises Compound I and another anti-HCV agent, it is possible to separately prepare solid dispersions of each individual active ingredient and then blend the optionally milled or ground solid dispersions before compacting. Compound I and other active ingredient(s) can also be prepared in the same solid dispersion, optionally milled and/or blended with other additives, and then compressed into tablets.
  • At least one additive selected from flow regulators, binders, lubricants, fillers, disintegrants, or plasticizers may be used in compressing the solid dispersion. These additives can be mixed with ground or milled solid dispersion before compacting. Disintegrants promote a rapid disintegration of the compact in the stomach and keeps the liberated granules separate from one another. Non-limiting examples of suitable disintegrants are cross-linked polymers such as cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethylcellulose or sodium croscarmellose. Non-limiting examples of suitable fillers (also referred to as bulking agents) are lactose monohydrate, calcium hydrogenphosphate, microcrystalline cellulose (e.g., Avicell), silicates, in particular silicium dioxide, magnesium oxide, talc, potato or corn starch, isomalt, or polyvinyl alcohol. Non-limiting examples of suitable flow regulators include highly dispersed silica (e.g., colloidal silica such as Aerosil), and animal or vegetable fats or waxes. Non-limiting examples of suitable lubricants include polyethylene glycol (e.g., having a molecular weight of from 1000 to 6000), magnesium and calcium stearates, sodium stearyl fumarate, and the like.
  • Various other additives may also be used in preparing a solid composition of the present invention, 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.
  • Solid compositions according to certain embodiments of the present invention may contain 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.
  • In order to facilitate the intake of a solid dosage form, 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 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. polysorbates, and optionally a pigment, e.g. titanium dioxide or iron oxides. The film-coating may also comprise talc as anti-adhesive. Preferably, the film coat accounts for less than 5% by weight of a pharmaceutical composition of the present invention.
  • In another aspect, the present invention feature methods of using solid compositions of the present invention to treat HIV infection. The methods comprise administering a solid composition of the present invention to a patient in need thereof. A solid composition of the present invention can be administered either alone, or in combination with one or more other anti-HCV agents, such as those described hereinabove. The specific inhibitory dose for any particular patient will depend upon a variety of factors including the severity of the HCV infection; the activity of Compound I in the particular patient; the specific solid composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration and rate of excretion; the duration of the treatment; drugs used in combination or coincidental with Compound I; and like factors well known in the medical arts.
  • In one embodiment, a method of the present invention comprises administering to a patient in need thereof a solid composition of the present invention and at least another anti-HCV agent, wherein said another anti-HCV agent is selected from HCV polymerase inhibitors (e.g., nucleoside or non-nucleoside HCV polymerase inhibitors), HCV protease inhibitors, HCV helicase inhibitors, CD81 inhibitors, cyclophilin inhibitors, internal ribosome entry site inhibitors, or HCV NS5A inhibitors. Preferably, said another anti-HCV agent is an HCV polymerase inhibitor (e.g., nucleoside or non-nucleoside HCV polymerase inhibitor) or an HCV NS5A inhibitor. Also preferably, said another anti-HCV agent is interferon or ribavirin, or preferably a combination thereof. The interferon preferably is α-interferon, and more preferably, pegylated interferon-α such as PEGASYS (peginterferon alfa-2a). The administration of a solid composition of the present invention and another anti-HCV agent(s) can be concurrent or sequential.
  • The present invention also features use of a solid composition of the present invention for the manufacture of medicaments for the treatment of HCV infection.
  • It should be understood that the above-described embodiments and the following examples are given by way of illustration, not limitation. Various changes and modifications within the scope of the present invention will become apparent to those skilled in the art from the present description.
  • EXAMPLE 1
  • Compound I was extruded using melt-extrusion. Two extrudates were prepared, and then milled and filled into capsules. The 1st extrudate contained Compound I, copovidone, and Vitamin E-TPGS in a weight ratio of 5:88:7 (hereinafter Formulation 1). The 2nd extrudate contained Compound I, copovidone and Sorbitan monolaurate in a weight ratio of 5:90:5 (hereinafter Formulation 2). The extrusion mixtures were prepared by use of mortar and pestle. Both formulations were extruded at 140° C. The obtained extruded strands were milled and the fractions of over 0.2 mm were combined with 100 mg mannite/collidal silica (99:1) and then filled into capsules. Each of these extrudate capsules contained 5 mg Compound I.
  • EXAMPLE 2
  • The pharmacokinetic profile of each formulation described in Example 1 was evaluated in dogs after single oral (PO) administration of the formulation. Four dogs (two male and two female dogs) were used in this study. The animals were fasted overnight and received food 30 min prior to dosing and throughout the duration of the study. Plasma samples were collected at 0.25, 0.5, 1, 2, 4, 6, 8, 12 and 24 hours post-dose administration. Plasma samples were analyzed for Compound I by LC-MS/MS. AUC0-inf and Cmax were normalized to a dose of 0.5 mg/kg Compound I.
  • Mean dose-normalized AUC0-inf values of Compound I were 512.2 and 432.0 ng·h/ml, at a 0.5 mg/kg dose in Formulations 1 and 2, respectively. Mean dose-normalized Cmax values of Compound I were 36.1 and 15.2 ng/ml at a 0.5 mg/kg dose in Formulations 1 and 2, respectively.
  • EXAMPLE 3
  • Compound I was mixed with hydrophilic polymers and pharmaceutically acceptable surfactants at various ratios, and dissolved in an organic solvent (acetone or acetone/water mixtures). The solvent was then removed from the system under heat (˜75° C.) and vacuum, using a Genevac rotary evaporator or Buchi Rotavap. Solid dispersions of Compound I at various drug loading levels and using different surfactants or polymers were sieved through a 30 mesh screen to reduce particle size. The resultant solid dispersion samples were used for amorphous characterization by X-ray powder diffraction (RXRD), chemical stability, in-vitro dissolution test and dog bioavailability studies.
  • For dog bioavailability studies, the solid dispersion powder was mixed with other excipients and compressed into tablets to achieve strengths of 0.5 mg, 5.0 mg, and 25.0 mg. For in-vitro dissolution studies, the release of Compound I was evaluated.
  • The hydrophilic polymers employed were copovidone, soluplus, hydroxypropyl methylcellulose phthalate (HPMCP), and hydroxypropyl methylcellulose grade E5 (HPMC-E5). The surfactants employed were Vitamin E TPGS and Cremaphor RH40. The amount of the surfactant(s) in each solid dispersion was no more than 10% by weight, and the amount of Compound I in each solid dispersion ranged from 5 to 20% by weight.
  • All solid dispersions showed that Compound I was in an amorphous form, as indicated by their PXRD patterns. Solid dispersions containing copovidone were evaluated for stability and showed chemical stability after 4 weeks at 40° C. and 75% relative humidity in closed dish studies. These solid dispersions also exhibited rapid dissolution rate.
  • EXAMPLE 4
  • One solid dispersion formulation was prepared using spray-drying to produce a solid dispersion powder of amorphous Compound I within a polymer matrix. The spray dried powder contained 10% by weight of Compound I, 85% by weight of copovidone, and 5% by weight of Vitamin E TPGS. Acetone and water in a 9:1 ratio was used as a solvent for spray-drying.
  • The spray dried powder was further dried under vacuum to remove residual solvent. The vacuum dried powder was blended with microcrystalline cellulose, lactose monohydrate, colloidal silicon dioxide, sodium stearyl fumarate, and optionally croscarmellose sodium. This blend was then compressed into the final tablet dosage form.
  • The foregoing description of the present invention provides illustration and description, but is not intended to be exhaustive or to limit the invention to the precise one disclosed. Modifications and variations are possible in light of the above teachings or may be acquired from practice of the invention. Thus, it is noted that the scope of the invention is defined by the claims and their equivalents.

Claims (20)

What is claimed is:
1. A process of making a solid pharmaceutical dosage form, comprising drying a liquid solution to form a solid composition, wherein said liquid solution comprises:
(1) dimethyl(2S,2′S)-1,1′-((2S,2′S)-2,2′-(4,4′-((2S,5S)-1-(4-tert-butylphenyl)pyrrolidine-2,5-diyl)bis(4,1-phenylene))bis(azanediyl)bis(oxomethylene)bis(pyrrolidine-2,1-diyl))bis(3-methyl-1-oxobutane-2,1-diyl)dicarbamate (
Figure US20140171481A1-20140619-C00002
or a pharmaceutically acceptable salt thereof;
(2) a pharmaceutically acceptable hydrophilic polymer; and
(3) a pharmaceutically acceptable surfactant,
and wherein said Compound I or salt is in an amorphous form in said solid composition.
2. The process of claim 1, wherein said solid composition is a solid dispersion which includes:
(1) said Compound I or salt,
(2) said polymer, and
(3) said surfactant.
3. The process of claim 2, wherein said polymer has a Tg of at least 50° C.
4. The process of claim 3, wherein said surfactant has a HLB value of at least 10.
5. The process of claim 2, wherein said solid dispersion is an amorphous solid dispersion.
6. The process of claim 2, wherein said polymer is copovidone, and said surfactant is D-alpha-tocopheryl polyethylene glycol 1000 succinate.
7. The process of claim 6, wherein said solid dispersion is an amorphous solid dispersion.
8. The process of claim 1, wherein said dosage form further comprises another anti-HCV agent.
9. The process of claim 1, wherein said dosage form further comprises an HCV protease inhibitor.
10. The process of claim 1, wherein said dosage form further comprises an MY polymerase inhibitor.
11. A process of making a solid pharmaceutical dosage form, comprising solidifying a melt, wherein said melt comprises:
(1) dimethyl(2S,2′S)-1,1′-((2S,2′S)-2,2′-(4,4′-((2S,5S)-1-(4-tert-butylphenyl)pyrrolidine-2,5-diyl)bis(4,1-phenylene))bis(azanediyl)bis(oxomethylene)bis(pyrrolidine-2,1-diyl))bis(3-methyl-1-oxobutane-2,1-diyl)dicarbamate (
Figure US20140171481A1-20140619-C00003
or a pharmaceutically acceptable salt thereof;
(2) a pharmaceutically acceptable hydrophilic polymer; and
(3) a pharmaceutically acceptable surfactant.
and wherein said Compound I or salt is in an amorphous form in the solidified melt.
12. The process of claim 11, wherein the solidified melt is a solid dispersion which includes:
(1) said Compound I or salt,
(2) said polymer, and
(3) said surfactant.
13. The process of claim 12, wherein said polymer has a Tg of at least 50° C.
14. The process of claim 13, wherein said surfactant has a HLB value of at least 10.
15. The process of claim 12, wherein said solid dispersion is an amorphous solid dispersion.
16. The process of claim 12, wherein said polymer is copovidone, and said surfactant is D-alpha-tocopheryl polyethylene glycol 1000 succinate.
17. The process of claim 16, wherein said solid dispersion is an amorphous solid dispersion.
18. The process of claim 11, wherein said dosage form further comprises another anti-HCV agent.
19. The process of claim 11, wherein said dosage form further comprises an HCV protease inhibitor.
20. The process of claim 11, wherein said dosage form further comprises an HCV polymerase inhibitor.
US14/180,531 2010-06-10 2014-02-14 Solid compositions Abandoned US20140171481A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/180,531 US20140171481A1 (en) 2010-06-10 2014-02-14 Solid compositions

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US35355310P 2010-06-10 2010-06-10
US13/156,783 US8686026B2 (en) 2010-06-10 2011-06-09 Solid compositions
US14/180,531 US20140171481A1 (en) 2010-06-10 2014-02-14 Solid compositions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/156,783 Division US8686026B2 (en) 2010-06-10 2011-06-09 Solid compositions

Publications (1)

Publication Number Publication Date
US20140171481A1 true US20140171481A1 (en) 2014-06-19

Family

ID=44317041

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/156,783 Active US8686026B2 (en) 2010-06-10 2011-06-09 Solid compositions
US14/180,531 Abandoned US20140171481A1 (en) 2010-06-10 2014-02-14 Solid compositions

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/156,783 Active US8686026B2 (en) 2010-06-10 2011-06-09 Solid compositions

Country Status (41)

Country Link
US (2) US8686026B2 (en)
EP (3) EP3456318A1 (en)
JP (1) JP5814356B2 (en)
KR (3) KR20180023014A (en)
CN (1) CN103209686B (en)
AR (1) AR083240A1 (en)
AU (1) AU2011264823B2 (en)
BR (2) BR212012031500U2 (en)
CA (1) CA2802180C (en)
CL (1) CL2012003470A1 (en)
CO (1) CO6660490A2 (en)
CR (1) CR20120650A (en)
CY (2) CY1116748T1 (en)
DK (2) DK2579854T4 (en)
DO (1) DOP2012000307A (en)
EA (1) EA024538B1 (en)
EC (1) ECSP13012382A (en)
ES (2) ES2691625T3 (en)
GT (1) GT201200332A (en)
HK (1) HK1184068A1 (en)
HR (2) HRP20150926T4 (en)
HU (2) HUE039719T2 (en)
IL (1) IL223535A (en)
LT (1) LT2954892T (en)
ME (1) ME02201B (en)
MX (1) MX2012014384A (en)
MY (1) MY164607A (en)
NZ (1) NZ605440A (en)
PE (1) PE20131036A1 (en)
PL (2) PL2954892T3 (en)
PT (2) PT2579854E (en)
RS (2) RS54282B2 (en)
SG (1) SG186251A1 (en)
SI (2) SI2954892T1 (en)
SM (1) SMT201500263B (en)
TR (1) TR201815161T4 (en)
TW (1) TWI487700B (en)
UA (1) UA105434C2 (en)
UY (1) UY33446A (en)
WO (1) WO2011156578A1 (en)
ZA (1) ZA201300112B (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016010771A1 (en) * 2014-07-17 2016-01-21 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9598407B2 (en) 2011-08-10 2017-03-21 Janssen Sciences Ireland Uc Antibacterial piperidinyl substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US20180185399A1 (en) * 2015-07-03 2018-07-05 Zhejiang Hisun Pharmaceutical Co., Ltd. Ginsenoside c-k oral solid preparation and preparation method thereof
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form

Families Citing this family (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2740193A1 (en) 2008-12-23 2010-07-01 Abbott Laboratories Anti-viral compounds
CA2740195A1 (en) 2008-12-23 2010-07-01 Abbott Laboratories Anti-viral compounds
JP5734956B2 (en) 2009-04-15 2015-06-17 アッヴィ・インコーポレイテッド Antiviral compounds
RS53856B1 (en) 2009-06-11 2015-08-31 Abbvie Bahamas Ltd. Heterocyclic compounds as inhibitors of hepatitis c virus (hcv)
US8937150B2 (en) 2009-06-11 2015-01-20 Abbvie Inc. Anti-viral compounds
NZ605440A (en) 2010-06-10 2014-05-30 Abbvie Bahamas Ltd Solid compositions comprising an hcv inhibitor
US8552047B2 (en) 2011-02-07 2013-10-08 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9546160B2 (en) 2011-05-12 2017-01-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US9303061B2 (en) 2011-07-09 2016-04-05 Sunshine Luke Pharma Co., Ltd. Spiro compounds as Hepatitis C virus inhibitors
AU2013201758B2 (en) * 2011-10-21 2014-11-27 Abbvie Ireland Unlimited Company Methods for treating HCV
EA201490837A1 (en) * 2011-10-21 2014-11-28 Эббви Инк. METHODS OF TREATING HCV, INCLUDING, AT THE LITERATURE, TWO ANTI-VIRAL AGENTS OF DIRECT ACTION, RIBAVIRIN, BUT NOT INTERFERON
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV
AU2015100285B4 (en) * 2011-10-21 2015-06-11 Abbvie Ireland Unlimited Company Methods for treating HCV
US8492386B2 (en) 2011-10-21 2013-07-23 Abbvie Inc. Methods for treating HCV
DE112012003457T5 (en) 2011-10-21 2015-03-12 Abbvie Inc. Combination treatment (e.g., with ABT-072 or ABT-333 from DAAs for use in the treatment of HCV)
WO2013101552A1 (en) * 2011-12-28 2013-07-04 Abbvie Inc. Methods for treating hcv
US20130172239A1 (en) * 2011-12-29 2013-07-04 Abbvie Inc. Solid compositions
US9034832B2 (en) 2011-12-29 2015-05-19 Abbvie Inc. Solid compositions
US9326973B2 (en) 2012-01-13 2016-05-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US20150119317A1 (en) * 2012-05-07 2015-04-30 Bristol-Myers Squibb Company Oral solid dosage formulation of 1,1-dimethylethyl [(1s)-1-carbamoyl)pyrrolidin-1-yl]carbonyl}-2,2-dimethylpropyl]carbamate
TWI610916B (en) 2012-08-03 2018-01-11 廣東東陽光藥業有限公司 Bridged ring compounds as hepatitis c virus (hcv) inhibitors and pharmaceuticals applications thereof
CN103848821B (en) 2012-11-29 2016-10-12 广东东阳光药业有限公司 Spiro-compound, pharmaceutical composition and their purposes as hepatitis c inhibitor
CN103848818B (en) 2012-11-29 2017-03-15 广东东阳光药业有限公司 Simultaneously cycle compound, pharmaceutical composition and their applications in medicine as hepatitis c inhibitor
MY172166A (en) * 2013-01-31 2019-11-15 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
US11484534B2 (en) 2013-03-14 2022-11-01 Abbvie Inc. Methods for treating HCV
JP6130585B2 (en) * 2013-03-15 2017-05-17 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Solid oral dosage form of amorphous HCV inhibitor
US20150023913A1 (en) 2013-07-02 2015-01-22 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
EP3016651A1 (en) 2013-07-02 2016-05-11 AbbVie Inc. Methods for treating hcv
US9717712B2 (en) 2013-07-02 2017-08-01 Bristol-Myers Squibb Company Combinations comprising tricyclohexadecahexaene derivatives for use in the treatment of hepatitis C virus
WO2015009744A1 (en) 2013-07-17 2015-01-22 Bristol-Myers Squibb Company Combinations comprising biphenyl derivatives for use in the treatment of hcv
EP4005560A1 (en) 2013-08-27 2022-06-01 Gilead Pharmasset LLC Combination formulation of two antiviral compounds
JP2016535794A (en) * 2013-11-11 2016-11-17 インパックス ラボラトリーズ,インコーポレーテッド Rapidly disintegrating preparations and methods of use
JP6232262B2 (en) * 2013-11-13 2017-11-15 アクアス株式会社 Solid water treatment agent having sustained release and production method thereof
KR101561964B1 (en) 2013-11-15 2015-10-20 한국과학기술연구원 Oxazolidinone derivatives and composition for preventing or treating Hepatitis C containing the same
US20150141351A1 (en) * 2013-11-18 2015-05-21 AbbVie Deutschland GmbH & Co. KG Solid Pharmaceutical Compositions
WO2015103490A1 (en) 2014-01-03 2015-07-09 Abbvie, Inc. Solid antiviral dosage forms
US9738629B2 (en) 2014-01-23 2017-08-22 Sunshine Lake Pharma Co., Ltd. Bridged ring compounds as Hepatitis C virus inhibitors, pharmaceutical compositions and uses thereof
MX2016010169A (en) * 2014-02-05 2016-10-07 Merck Sharp & Dohme Tablet formulation for cgrp-active compounds.
WO2015171162A1 (en) * 2014-05-09 2015-11-12 Abbvie Inc. Anti-viral compound
TWI721947B (en) 2014-06-11 2021-03-21 美商基利法瑪席特有限責任公司 Solid forms of an antiviral compound
US20160193151A1 (en) * 2015-01-06 2016-07-07 Maria Del Pilar Noriega Escobar Dosage form incorporating an amorphous drug solid solution
WO2016134058A1 (en) 2015-02-18 2016-08-25 Abbvie Inc. Combinations useful to treat hepatitis c virus
US20160375017A1 (en) 2015-06-26 2016-12-29 Abbvie Inc. Solid Pharmaceutical Compositions for Treating HCV
RU2021102950A (en) * 2015-06-26 2021-03-01 Эббви Инк. SOLID PHARMACEUTICAL COMPOSITIONS FOR TREATMENT OF HEPATITIS C VIRUS
RU2018105849A (en) * 2015-07-17 2019-08-19 Эббви Инк. SOLID PHARMACEUTICAL COMPOSITIONS FOR TREATMENT OF HEPATITIS C VIRUS
US10617675B2 (en) 2015-08-06 2020-04-14 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2017038925A1 (en) * 2015-09-03 2017-03-09 日本化薬株式会社 Pharmaceutical composition containing rapamycin or derivative thereof
TW201718608A (en) * 2015-11-23 2017-06-01 Chia Tai Tianqing Pharmaceutical Group Co Ltd Silicone-containing compound for resisting hepatitis c virus infection
EA201892448A1 (en) 2016-04-28 2019-06-28 Эмори Юниверсити ALKYN-CONTAINING NUCLEOTIDE AND NUCLEOSIDE THERAPEUTIC COMPOSITIONS AND RELATED APPLICATION METHODS
US20200323815A1 (en) * 2017-07-20 2020-10-15 Amri Ssci, Llc Amorphous dispersions of epigallocatechin gallate
KR102286706B1 (en) 2020-09-29 2021-08-06 솔브레인 주식회사 Method for producing vanadium electrolyte
KR102286707B1 (en) 2020-10-30 2021-08-06 솔브레인 주식회사 Method and apparatus for producing vanadium electrolyte
KR102286709B1 (en) 2020-11-03 2021-08-06 솔브레인 주식회사 Apparatus for producing vanadium electrolyte
CA3206184A1 (en) 2020-12-22 2022-06-30 Allergan Pharmaceuticals International Limited Treatment of migraine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050084529A1 (en) * 2003-08-28 2005-04-21 Joerg Rosenberg Solid pharmaceutical dosage form
US8691938B2 (en) * 2009-06-11 2014-04-08 Abbvie Inc. Anti-viral compounds

Family Cites Families (188)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE75755C (en) 1894-06-14 DAHL & COMP, in Barmen Process for the preparation of aromatically substituted amidodinaphthyl methanes.
US353553A (en) 1886-11-30 Du bois f
US813301A (en) 1903-09-09 1906-02-20 Frank S Hyde Steam-engine.
WO1994027627A1 (en) 1993-05-24 1994-12-08 Smithkline Beecham Corporation Hemoregulatory peptides
SI9500173B (en) 1995-05-19 2002-02-28 Lek, Three-phase pharmaceutical form with constant and controlled release of amorphous active ingredient for single daily application
US6037157A (en) 1995-06-29 2000-03-14 Abbott Laboratories Method for improving pharmacokinetics
US5935982A (en) 1997-02-28 1999-08-10 The University Of North Carolina At Chapel Hill Methods of treating retroviral infection and compounds useful therefor
US6235493B1 (en) 1997-08-06 2001-05-22 The Regents Of The University Of California Amino acid substituted-cresyl violet, synthetic fluorogenic substrates for the analysis of agents in individual in vivo cells or tissue
US6919366B2 (en) 1998-05-22 2005-07-19 Avanir Pharmaceuticals Benzimidazole derivatives as modulators of IgE
HUP0102128A3 (en) 1998-05-22 2002-05-28 Avanir Pharmaceuticals San Die Mediciaments containing benzimidazole derivatives as modulators of ige
US6911462B2 (en) 1998-05-22 2005-06-28 Avanir Pharmaceuticals Benzimidazole compounds for regulating IgE
US6369091B1 (en) 1998-05-22 2002-04-09 Avanir Pharmaceuticals Benzimidazole analogs as down-regulators of IgE
KR100336090B1 (en) 1998-06-27 2002-05-27 윤승원 Solid dispersed preparation of poorly water-soluble drug containing oil, fatty acid or mixture thereof
US6387885B1 (en) 1998-08-26 2002-05-14 Abbott Laboratories 3′,3′-N-bis-desmethyl-3′-N-cycloalkyl erythromycin derivatives as LHRH antagonists
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
DE19913692A1 (en) 1999-03-25 2000-09-28 Basf Ag Mechanically stable pharmaceutical dosage forms containing liquid or semi-solid surface-active substances
AR029423A1 (en) 1999-12-21 2003-06-25 Sugen Inc COMPOSITE DERIVED FROM PIRROLO- [PIRIMIDIN OR PIRIDIN] -6-ONA, METHOD OF PREPARATION OF THESE COMPOUNDS, PHARMACEUTICAL COMPOSITIONS THAT INCLUDE THEM, A METHOD FOR REGULATING, MODULATING OR INHIBITING THE ACTIVITY OF THE PROTEIN QUINASA AND ONE METHOD MAMMALS DISEASE
KR20030036152A (en) 2000-04-05 2003-05-09 쉐링 코포레이션 Macrocyclic NS3-serine protease inhibitors of hepatitis C virus comprising N-cyclic P2 moieties
DE10026698A1 (en) 2000-05-30 2001-12-06 Basf Ag Self-emulsifying active ingredient formulation and use of this formulation
US7065453B1 (en) 2000-06-15 2006-06-20 Accelrys Software, Inc. Molecular docking technique for screening of combinatorial libraries
DK1309591T3 (en) 2000-08-14 2007-03-26 Ortho Mcneil Pharm Inc Substituted pyrazoles
US6388093B1 (en) 2001-01-05 2002-05-14 Sun Chemical Corporation Syntheses for preparing 1,4-diketopyrrolo [3,4-c]pyrroles
SK288015B6 (en) 2001-06-11 2012-11-05 Virochem Pharma Inc. Thiophene derivatives as antiviral agents for flavivirus infection
EP2335700A1 (en) 2001-07-25 2011-06-22 Boehringer Ingelheim (Canada) Ltd. Hepatitis C virus polymerase inhibitors with a heterobicylic structure
RU2286343C2 (en) 2001-08-10 2006-10-27 Орто-Макнейл Фармасьютикал, Инк. Substituted pyrazoles
JP3925265B2 (en) 2002-03-25 2007-06-06 コニカミノルタホールディングス株式会社 Organic electroluminescence element and display device using the same
TW200304820A (en) 2002-03-25 2003-10-16 Avanir Pharmaceuticals Use of benzimidazole analogs in the treatment of cell proliferation
MXPA04012642A (en) 2002-06-14 2005-03-23 Merck & Co Inc Mitotic kinesin inhibitors.
WO2004005283A1 (en) 2002-07-09 2004-01-15 Vertex Pharmaceuticals Incorporated Imidazoles, oxazoles and thiazoles with protein kinase inhibiting activities
US7183302B2 (en) 2002-08-12 2007-02-27 Bristol-Myers Squibb Company Iminothiazolidinones as inhibitors of HCV replication
GB0229518D0 (en) 2002-12-19 2003-01-22 Astrazeneca Ab Chemical compounds
TW200508224A (en) 2003-02-12 2005-03-01 Bristol Myers Squibb Co Cyclic derivatives as modulators of chemokine receptor activity
MXPA06004723A (en) 2003-10-27 2006-07-05 Vertex Pharma Combinations for hcv treatment.
BRPI0401908A (en) 2004-06-04 2006-01-17 Univ Rio De Janeiro Serine protease inhibitor compounds, process for obtaining and using for treatment of flaviviroses
US7772271B2 (en) 2004-07-14 2010-08-10 Ptc Therapeutics, Inc. Methods for treating hepatitis C
JP5014129B2 (en) 2004-07-27 2012-08-29 ギリアード サイエンシーズ, インコーポレイテッド Imidazo [4,5-d] pyrimidines, their use and methods of preparation
WO2006020951A1 (en) 2004-08-13 2006-02-23 Praecis Pharmaceuticals, Inc. Methods and compositions for modulating sphingosine-1-phosphate (s1p) receptor activity
TW200633718A (en) 2004-12-16 2006-10-01 Applied Research Systems Treatment of hepatitis c in the asian population
RU2007124935A (en) 2005-02-01 2009-03-10 Вайет (Us) AMINOPYRIDINES AS β-SECTRETASIS INHIBITORS
CN101146769A (en) 2005-02-14 2008-03-19 惠氏公司 Azolylacylguanidines as beta-secretase inhibitors
GB2438802A (en) 2005-02-28 2007-12-05 Univ Rockefeller Structure of the hepatitis C virus NS5A protein
US8143288B2 (en) 2005-06-06 2012-03-27 Bristol-Myers Squibb Company Inhibitors of HCV replication
US7541359B2 (en) 2005-06-30 2009-06-02 Janssen Pharmaceutica N.V. N-heteroarylpiperazinyl ureas as modulators of fatty acid amide hydrolase
WO2007011284A1 (en) 2005-07-15 2007-01-25 Astrazeneca Ab Therapeutic agents
CA2633541A1 (en) 2005-12-12 2007-06-21 Genelabs Technologies, Inc. N-(6-membered aromatic ring)-amido anti-viral compounds
CA2633569A1 (en) 2005-12-12 2007-06-21 Genelabs Technologies, Inc. N-(5-membered heteroaromatic ring)-amido anti-viral compounds
DE602006015861D1 (en) 2005-12-21 2010-09-09 Abbott Lab ANTIVIRAL CONNECTIONS
ATE541844T1 (en) 2005-12-21 2012-02-15 Abbott Lab ANTIVIRAL COMPOUNDS
WO2007073405A1 (en) 2005-12-21 2007-06-28 Decode Genetics Ehf N-linked aryl heteroaryl inhibitors of lta4h for treating inflammation
US7915411B2 (en) 2005-12-21 2011-03-29 Abbott Laboratories Anti-viral compounds
WO2007082554A1 (en) 2006-01-23 2007-07-26 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Modulators of hcv replication
JP5419685B2 (en) 2006-05-16 2014-02-19 ファーマサイエンス・インコーポレイテッド IAPBIR domain binding protein
US8263772B2 (en) 2006-06-08 2012-09-11 Eli Lilly And Company MCH receptor antagonists
ES2392104T3 (en) 2006-06-16 2012-12-04 Syngenta Participations Ag Ethyl carboxamide derivatives useful as microbiocides
US20100056495A1 (en) 2006-07-24 2010-03-04 Tetralogic Pharmaceuticals Corporation Dimeric iap inhibitors
WO2008014238A2 (en) 2006-07-24 2008-01-31 Tetralogic Pharmaceuticals Corporation Dimeric iap inhibitors
US7659270B2 (en) 2006-08-11 2010-02-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7759495B2 (en) * 2006-08-11 2010-07-20 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7745636B2 (en) 2006-08-11 2010-06-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US20100158862A1 (en) 2006-08-11 2010-06-24 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US8329159B2 (en) * 2006-08-11 2012-12-11 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8303944B2 (en) 2006-08-11 2012-11-06 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
JP2010510245A (en) 2006-11-21 2010-04-02 スミスクライン ビーチャム コーポレーション Antiviral compounds
US20080181866A1 (en) 2006-11-21 2008-07-31 Genelabs Technologies, Inc. Amido Anti-viral Compounds
WO2008074450A2 (en) 2006-12-20 2008-06-26 Nicox S.A. Non-peptidic renin inhibitors nitroderivatives
TWI399380B (en) 2006-12-20 2013-06-21 Abbott Lab Anti-viral compounds
EP2079309B1 (en) 2007-04-12 2015-11-11 Joyant Pharmaceuticals Inc Smac mimetic dimers and trimers useful as anti-cancer agents
US7741347B2 (en) * 2007-05-17 2010-06-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2009003009A1 (en) 2007-06-26 2008-12-31 Enanta Pharmaceuticals, Inc. Substituted pyrrolidine as anti-infectives
EP2185154A2 (en) 2007-08-03 2010-05-19 Schering Corporation Method of treating cxcr3 mediated diseases using heterocyclic substituted piperazines
US7728027B2 (en) 2007-08-08 2010-06-01 Bristol-Myers Squibb Company Process for synthesizing compounds useful for treating hepatitis C
US8629171B2 (en) 2007-08-08 2014-01-14 Bristol-Myers Squibb Company Crystalline form of methyl ((1S)-1-((25)-2-(5-(4'-(2-((25)-1((2S)-2-((methoxycarbonyl)amino)-3-methylbutanoyl)-2-pyrrolidinyl)-1H-imidazol-2-yl)-1-pyrrolidinyl)carbonyl)-2-methylpropyl)carbamate dihydrochloride salt
GB0801199D0 (en) 2008-01-23 2008-02-27 Acal Energy Ltd Fuel cells
EP2250160B1 (en) 2008-01-25 2015-11-11 Millennium Pharmaceuticals, Inc. Thiophenes and their use as phosphatidylinositol 3-kinase (pi3k) inhibitors
KR101468765B1 (en) 2008-02-12 2014-12-04 브리스톨-마이어스 스큅 컴퍼니 Hepatitis c virus inhibitors
AU2009214977A1 (en) 2008-02-12 2009-08-20 Bristol-Myers Squibb Company Heterocyclic derivatives as hepatitis C virus inhibitors
BRPI0822323A2 (en) 2008-02-13 2015-06-16 Bristol Myers Squibb Co Imidazolyl biphenyl imidazoles as hepatitis c virus inhibitors
US7704992B2 (en) 2008-02-13 2010-04-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8147818B2 (en) 2008-02-13 2012-04-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
CN101959506A (en) 2008-02-28 2011-01-26 雅培制药有限公司 Tablets and preparation thereof
EP2262539B1 (en) 2008-04-01 2015-07-15 Novo Nordisk A/S Insulin albumin conjugates
US20110117081A1 (en) 2008-05-05 2011-05-19 Aegera Therapeutics, Inc. Functionalized pyrrolidines and use thereof as iap inhibitors
WO2009143361A1 (en) 2008-05-22 2009-11-26 Smithkline Beecham Corporation Amido anti-viral compounds
CA2728933A1 (en) 2008-06-27 2009-12-30 Aegera Therapeutics Inc. Bridged secondary amines and use thereof as iap bir domain binding compounds
DK2540350T3 (en) 2008-07-22 2014-08-04 Merck Sharp & Dohme COMBINATIONS OF A MACROCYCLIC QUINOXAL COMPOUND, WHICH IS ANHCV-NS3 PROTEASE INHIBITOR WITH OTHER HCV AGENTS
CA2730448A1 (en) 2008-08-02 2010-02-11 Genentech, Inc. Inhibitors of iap
WO2010015090A1 (en) 2008-08-07 2010-02-11 Aegera Therapeutics Inc. Functionalized pyrrolidines and use thereof as iap inhibitors
US7906655B2 (en) 2008-08-07 2011-03-15 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8383094B2 (en) 2008-10-01 2013-02-26 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2010059858A1 (en) 2008-11-19 2010-05-27 Duke University Manassantin compounds and methods of making and using same
JP2010126571A (en) 2008-11-26 2010-06-10 Toyo Ink Mfg Co Ltd Organic electroluminescent element material and organic electroluminescent element
WO2010062821A1 (en) 2008-11-28 2010-06-03 Glaxosmithkline Llc Anti-viral compounds, compositions, and methods of use
BRPI0922364A2 (en) 2008-12-03 2017-08-29 Presidio Pharmaceuticals Inc COMPOUND, PHARMACEUTICAL COMPOSITION AND USE OF A COMPOUND
EP2682393A1 (en) 2008-12-03 2014-01-08 Presidio Pharmaceuticals, Inc. Inhibitors of HCV NS5A comprising a bicyclic core.
CA2740193A1 (en) 2008-12-23 2010-07-01 Abbott Laboratories Anti-viral compounds
CA2740195A1 (en) 2008-12-23 2010-07-01 Abbott Laboratories Anti-viral compounds
WO2010091413A1 (en) 2009-02-09 2010-08-12 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US8314135B2 (en) 2009-02-09 2012-11-20 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole antivirals
TWI438200B (en) 2009-02-17 2014-05-21 必治妥美雅史谷比公司 Hepatitis c virus inhibitors
US8188132B2 (en) 2009-02-17 2012-05-29 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US8637561B2 (en) 2009-02-17 2014-01-28 Enanta Pharmaceuticals, Inc. Linked diimidazole derivatives
US8242156B2 (en) 2009-02-17 2012-08-14 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US8394968B2 (en) 2009-02-17 2013-03-12 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2010096462A1 (en) 2009-02-17 2010-08-26 Enanta Pharmaceuticals, Inc Linked diimidazole derivatives
US8420686B2 (en) 2009-02-17 2013-04-16 Enanta Pharmaceuticals, Inc. Linked diimidazole antivirals
UY32462A (en) 2009-02-23 2010-09-30 Arrow Therapeutics Ltd NEW BIFENYLL DERIVATIVES FOR HEPATITIS C 644 VIRUS INFECTION TREATMENT
CA2753313A1 (en) 2009-02-23 2010-08-26 Presidio Pharmaceuticals, Inc. Inhibitors of hcv ns5a
US8101643B2 (en) 2009-02-27 2012-01-24 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US8426458B2 (en) 2009-02-27 2013-04-23 Enanta Pharmaceuticals, Inc. Hepatitis C Virus inhibitors
NZ595280A (en) 2009-02-27 2013-11-29 Enanta Pharm Inc Hepatitis c virus inhibitors
US8673954B2 (en) 2009-02-27 2014-03-18 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US8507522B2 (en) 2009-03-06 2013-08-13 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
CA2756255A1 (en) 2009-03-27 2010-09-30 Presidio Pharmaceuticals, Inc. Substituted bicyclic hcv inhibitors
JP5735482B2 (en) 2009-03-27 2015-06-17 プレシディオ ファーマシューティカルズ インコーポレイテッド Fused hepatitis C inhibitor
SI2410844T1 (en) 2009-03-27 2016-06-30 Merck Sharp & Dohme Corp. Inhibitors of hepatitis c virus replication
TWI476190B (en) 2009-03-30 2015-03-11 必治妥美雅史谷比公司 Hepatitis c virus inhibitors
US8796466B2 (en) 2009-03-30 2014-08-05 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US20110237636A1 (en) 2009-03-30 2011-09-29 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US9067942B2 (en) 2009-04-08 2015-06-30 Basf Se Pyrrolopyrrole derivatives, their manufacture and use as semiconductors
TW201038559A (en) 2009-04-09 2010-11-01 Bristol Myers Squibb Co Hepatitis C virus inhibitors
US8143414B2 (en) 2009-04-13 2012-03-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
JP5734956B2 (en) 2009-04-15 2015-06-17 アッヴィ・インコーポレイテッド Antiviral compounds
US8436036B2 (en) 2009-04-24 2013-05-07 Janssen R&D Ireland Diaryl ethers
EP2430015B1 (en) 2009-05-12 2015-06-17 Merck Sharp & Dohme Corp. Fused tricyclic compounds useful for the treatment of viral diseases
US8088368B2 (en) 2009-05-13 2012-01-03 Gilead Sciences, Inc. Antiviral compounds
EP2435424B1 (en) 2009-05-29 2015-01-21 Merck Sharp & Dohme Corp. Antiviral compounds composed of three linked aryl moieties to treat diseases such as hepatitis c
US8211928B2 (en) 2009-05-29 2012-07-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8138215B2 (en) 2009-05-29 2012-03-20 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2010138790A1 (en) 2009-05-29 2010-12-02 Schering Corporation Antiviral compounds composed of three aligned aryl moieties to treat diseases such as hepatitis c
US9394279B2 (en) 2009-06-11 2016-07-19 Abbvie Inc. Anti-viral compounds
US8937150B2 (en) 2009-06-11 2015-01-20 Abbvie Inc. Anti-viral compounds
US8221737B2 (en) 2009-06-16 2012-07-17 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
WO2010148006A1 (en) 2009-06-16 2010-12-23 Enanta Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US8609648B2 (en) 2009-07-02 2013-12-17 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
WO2011004276A1 (en) 2009-07-06 2011-01-13 Pfizer Limited Hepatitis c virus inhibitors
US8354419B2 (en) 2009-07-16 2013-01-15 Vertex Pharmaceuticals Incorporated Benzimidazole analogues for the treatment or prevention of flavivirus infections
CN102471324B (en) 2009-08-07 2014-12-17 泰博特克药品公司 Bis-benzimidazole derivatives as hepatitis c virus inhibitors
EP2473503B1 (en) 2009-09-03 2014-07-23 Janssen R&D Ireland Bis-benzimidazole derivatives
CA2771327A1 (en) 2009-09-04 2011-03-10 Glaxosmithkline Llc Chemical compounds
US8703938B2 (en) 2009-09-11 2014-04-22 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8815928B2 (en) 2009-09-11 2014-08-26 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
WO2011031934A1 (en) 2009-09-11 2011-03-17 Enanta Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US8927709B2 (en) 2009-09-11 2015-01-06 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8822700B2 (en) 2009-09-11 2014-09-02 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
WO2011031904A1 (en) 2009-09-11 2011-03-17 Enanta Pharmaceuticals, Inc Hepatitis c virus inhibitors
US8759332B2 (en) 2009-09-11 2014-06-24 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
WO2011050146A1 (en) 2009-10-23 2011-04-28 Glaxosmithkline Llc Chemical compounds
UA108211C2 (en) 2009-11-04 2015-04-10 Янссен Рід Айрленд Benzimidazole imidazole derivatives
US20110269956A1 (en) 2009-11-11 2011-11-03 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20110274648A1 (en) 2009-11-11 2011-11-10 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20110281910A1 (en) 2009-11-12 2011-11-17 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
JP2013512246A (en) 2009-11-25 2013-04-11 メルク・シャープ・アンド・ドーム・コーポレーション Condensed tricyclic compounds and derivatives useful for the treatment of viral diseases
KR101768946B1 (en) 2009-12-04 2017-08-17 내셔날 헬스 리서치 인스티튜트 Proline derivatives
EP2512480A4 (en) 2009-12-14 2013-05-15 Enanta Pharm Inc Hepatitis c virus inhibitors
US8377980B2 (en) 2009-12-16 2013-02-19 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
TW201136942A (en) 2009-12-18 2011-11-01 Idenix Pharmaceuticals Inc 5,5-fused arylene or heteroarylene hepatitis C virus inhibitors
CN104530079B (en) 2009-12-18 2017-10-20 北京凯因科技股份有限公司 The new inhibitor that hepatitis C virus is replicated
US20130156731A1 (en) 2009-12-22 2013-06-20 Kevin X. Chen Fused tricyclic compounds and methods of use thereof for the treatment of viral diseas
CN102883718A (en) 2009-12-24 2013-01-16 顶点制药公司 Analogues for the treatment or prevention of flavivirus infections
US8362020B2 (en) 2009-12-30 2013-01-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2011091446A1 (en) 2010-01-22 2011-07-28 Glaxosmithkline Llc Chemical compounds
PE20130244A1 (en) 2010-01-25 2013-03-10 Enanta Pharm Inc HEPATITIS C VIRUS INHIBITORS
JP2013518062A (en) 2010-01-28 2013-05-20 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Hepatitis C inhibitor compound
US8178531B2 (en) 2010-02-23 2012-05-15 Enanta Pharmaceuticals, Inc. Antiviral agents
EA201201235A1 (en) 2010-03-04 2013-04-30 Энанта Фармасьютиклз, Инк. PHARMACEUTICAL AGENT COMBINATIONS AS HCV REPLICATION INHIBITORS
JP2013522202A (en) 2010-03-09 2013-06-13 メルク・シャープ・エンド・ドーム・コーポレイション Fused tricyclic silyl compounds and methods of use thereof for the treatment of viral diseases
TW201139438A (en) 2010-03-24 2011-11-16 Vertex Pharma Analogues for the treatment or prevention of flavivirus infections
CN102869657A (en) 2010-03-24 2013-01-09 沃泰克斯药物股份有限公司 Analogues for the treatment or prevention of flavivirus infections
TW201200522A (en) 2010-03-24 2012-01-01 Vertex Pharma Analogues for the treatment or prevention of flavivirus infections
TW201141857A (en) 2010-03-24 2011-12-01 Vertex Pharma Analogues for the treatment or prevention of flavivirus infections
WO2011127350A1 (en) 2010-04-09 2011-10-13 Enanta Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US20110312996A1 (en) 2010-05-17 2011-12-22 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
EP2575475A4 (en) 2010-05-28 2013-11-27 Presidio Pharmaceuticals Inc Inhibitors of hcv ns5a
EP2575819A4 (en) 2010-06-04 2013-11-27 Enanta Pharm Inc Hepatitis c virus inhibitors
AR081848A1 (en) 2010-06-09 2012-10-24 Presidio Pharmaceuticals Inc HCV NS5A PROTEIN INHIBITORS
NZ605440A (en) 2010-06-10 2014-05-30 Abbvie Bahamas Ltd Solid compositions comprising an hcv inhibitor
WO2012040389A2 (en) 2010-09-22 2012-03-29 Presidio Pharmaceuticals, Inc. Substituted bicyclic hcv inhibitors
WO2012039717A1 (en) 2010-09-24 2012-03-29 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2012040923A1 (en) 2010-09-29 2012-04-05 Merck Sharp & Dohme Corp. Tetracyclic indole derivatives and methods of use thereof for the treatment of viral diseases
WO2012050850A1 (en) 2010-09-29 2012-04-19 Merck Sharp & Dohme Corp. Polycyclic heterocycle derivatives and methods of use thereof for the treatment of viral diseases
WO2012041227A1 (en) 2010-09-29 2012-04-05 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds for treating hepatitis c viral infection
WO2012040924A1 (en) 2010-09-29 2012-04-05 Merck Sharp & Dohme Corp. Fused tetracyclic heterocycle compounds and methods of use thereof for treatment of viral diseases
AU2011314168A1 (en) 2010-09-29 2013-04-04 Merck Sharp & Dohme Corp. Fused tetracycle derivatives and methods of use thereof for the treatment of viral diseases
JP2013538831A (en) 2010-09-29 2013-10-17 メルク・シャープ・エンド・ドーム・コーポレイション Tetracyclic indole derivatives for the treatment of hepatitis C virus infection
CA2814534A1 (en) 2010-11-04 2012-05-10 Theravance, Inc. Novel inhibitors of hepatitis c virus
SG190785A1 (en) 2010-11-17 2013-07-31 Gilead Sciences Inc Antiviral compounds
RU2452735C1 (en) 2010-11-30 2012-06-10 Александр Васильевич Иващенко Substituted azoles, antiviral active component, pharmaceutical composition, synthesis and application method
WO2012087976A2 (en) 2010-12-21 2012-06-28 Intermune, Inc. Novel inhibitors of hepatitis c virus replication

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050084529A1 (en) * 2003-08-28 2005-04-21 Joerg Rosenberg Solid pharmaceutical dosage form
US8691938B2 (en) * 2009-06-11 2014-04-08 Abbvie Inc. Anti-viral compounds
US20140315792A1 (en) * 2009-06-11 2014-10-23 Abbvie, Inc. Anti-viral compounds

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Koev, Gennadiy et al, "Antiviral interactions of an hcv polymerase inhibitor with an hcv protease inhibitor or interferon in vitro." Antiviral Res. (2007) 73 p78-83 *
Wu, Stephen Hong Wei and Hopkins, Warren K.; "Characteristics of d-alpha-tocoperyl peg 1000 succinate for applications as an absorption enhancer in drug delivery systems." Pharmaceutical Tech. (1999) 23 p52-68 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9598407B2 (en) 2011-08-10 2017-03-21 Janssen Sciences Ireland Uc Antibacterial piperidinyl substituted 3,4-dihydro-1H-[1,8]naphthyridinones
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10639281B2 (en) 2013-08-12 2020-05-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10792254B2 (en) 2013-12-17 2020-10-06 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
WO2016010771A1 (en) * 2014-07-17 2016-01-21 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US20180185399A1 (en) * 2015-07-03 2018-07-05 Zhejiang Hisun Pharmaceutical Co., Ltd. Ginsenoside c-k oral solid preparation and preparation method thereof

Also Published As

Publication number Publication date
SI2579854T2 (en) 2018-10-30
HRP20150926T4 (en) 2018-11-30
JP2013528225A (en) 2013-07-08
BR112012031500A8 (en) 2022-11-01
ES2546767T3 (en) 2015-09-28
HUE025758T2 (en) 2016-04-28
CY1116748T1 (en) 2017-03-15
RS57982B1 (en) 2019-01-31
PE20131036A1 (en) 2013-10-20
SG186251A1 (en) 2013-01-30
TW201204713A (en) 2012-02-01
PT2954892T (en) 2018-11-09
KR20180023014A (en) 2018-03-06
CR20120650A (en) 2013-05-16
EP2579854B1 (en) 2015-07-15
SI2579854T1 (en) 2015-09-30
BR112012031500A2 (en) 2015-11-17
SI2954892T1 (en) 2019-01-31
US20120258909A1 (en) 2012-10-11
RS54282B2 (en) 2018-08-31
DK2579854T4 (en) 2018-10-22
AR083240A1 (en) 2013-02-13
US8686026B2 (en) 2014-04-01
MY164607A (en) 2018-01-30
ECSP13012382A (en) 2013-03-28
EP3456318A1 (en) 2019-03-20
JP5814356B2 (en) 2015-11-17
KR102059386B1 (en) 2019-12-26
CA2802180A1 (en) 2011-12-15
PL2579854T5 (en) 2018-12-31
CN103209686A (en) 2013-07-17
EP2954892A1 (en) 2015-12-16
KR20130053440A (en) 2013-05-23
BR212012031500U2 (en) 2015-11-17
EA024538B1 (en) 2016-09-30
RS54282B1 (en) 2016-02-29
CA2802180C (en) 2018-12-18
DOP2012000307A (en) 2013-01-31
EA201291394A1 (en) 2013-05-30
TR201815161T4 (en) 2018-11-21
IL223535A (en) 2017-02-28
CO6660490A2 (en) 2013-04-30
TWI487700B (en) 2015-06-11
SMT201500263B (en) 2016-01-08
PL2579854T3 (en) 2015-12-31
CN103209686B (en) 2015-06-17
NZ605440A (en) 2014-05-30
HRP20181658T1 (en) 2019-01-11
WO2011156578A1 (en) 2011-12-15
UA105434C2 (en) 2014-05-12
LT2954892T (en) 2018-12-10
KR20150008151A (en) 2015-01-21
EP2954892B1 (en) 2018-07-18
ES2546767T5 (en) 2018-07-27
DK2954892T3 (en) 2018-10-29
AU2011264823B2 (en) 2014-03-13
MX2012014384A (en) 2013-05-01
ZA201300112B (en) 2013-09-25
ES2691625T3 (en) 2018-11-28
HRP20150926T1 (en) 2015-10-09
AU2011264823A1 (en) 2013-01-24
EP2579854A1 (en) 2013-04-17
EP2579854B2 (en) 2018-07-04
UY33446A (en) 2012-01-31
KR101481395B1 (en) 2015-01-13
CY1122267T1 (en) 2020-07-31
DK2579854T3 (en) 2015-10-19
PT2579854E (en) 2015-10-16
ME02201B (en) 2016-02-20
GT201200332A (en) 2014-11-06
HK1184068A1 (en) 2014-01-17
CL2012003470A1 (en) 2013-02-15
PL2954892T3 (en) 2019-02-28
HUE039719T2 (en) 2019-01-28

Similar Documents

Publication Publication Date Title
US8686026B2 (en) Solid compositions
AU2011224558B2 (en) Solid compositions
US8716454B2 (en) Solid compositions
US20150322108A1 (en) Crystalline polymorphs
AU2014200725B2 (en) Solid compositions
WO2015171162A1 (en) Anti-viral compound
TW201311245A (en) Pharmaceutical compositions

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBOTT LABORATORIES, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SEVER, NANCY E.;LAFOUNTAINE, JUSTIN S.;SINCLAIR, BRENT D.;AND OTHERS;SIGNING DATES FROM 20110819 TO 20120314;REEL/FRAME:033000/0293

Owner name: ABBOTT GMBH & CO. KG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIEPOLD, BERND;JUNG, TINA;HOLIG, PETER;AND OTHERS;SIGNING DATES FROM 20110826 TO 20110829;REEL/FRAME:033000/0229

Owner name: ABBVIE INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABBOTT LABORATORIES;REEL/FRAME:033000/0301

Effective date: 20120801

Owner name: ABBOTT LABORATORIES, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABBOTT GMBH & CO.KG;REEL/FRAME:033000/0270

Effective date: 20110823

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION