WO2015061683A1 - Pronucléotides thiophosphoramidates à acide aminé d et pronucléotides thiophosphoramidates à d-analine de composés de nucléoside utiles pour le traitement du vhc - Google Patents

Pronucléotides thiophosphoramidates à acide aminé d et pronucléotides thiophosphoramidates à d-analine de composés de nucléoside utiles pour le traitement du vhc Download PDF

Info

Publication number
WO2015061683A1
WO2015061683A1 PCT/US2014/062174 US2014062174W WO2015061683A1 WO 2015061683 A1 WO2015061683 A1 WO 2015061683A1 US 2014062174 W US2014062174 W US 2014062174W WO 2015061683 A1 WO2015061683 A1 WO 2015061683A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
certain embodiments
compounds
hydrogen
formula
Prior art date
Application number
PCT/US2014/062174
Other languages
English (en)
Inventor
Alistair James Stewart
Adel M. Moussa
Benjamin Alexander Mayes
Original Assignee
Idenix Pharmaceuticals, 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
Application filed by Idenix Pharmaceuticals, Inc. filed Critical Idenix Pharmaceuticals, Inc.
Publication of WO2015061683A1 publication Critical patent/WO2015061683A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals

Definitions

  • D-alanine thiophosphoramidate pronucleotides of nucleoside compounds are provided which can display remarkable efficacy and bioavailability for the treatment of, for example, HCV infection in a human.
  • HCV hepatitis C virus
  • HCV causes a slow growing viral infection and is the major cause of cirrhosis and hepatocellular carcinoma (Di Besceglie, A. M. and Bacon, B. R., Scientific American, Oct.: 80-85, 1999; Boyer, N. et al., J. Hepatol. 32:98- 112, 2000). It is estimated there are about 130-150 million people with chronic hepatitis C virus infection. Hepatitis C-related liver diseases cause approximately 350,000 to 500,000 deaths each year. HCV infection becomes chronic in about 55-85% of cases, with many patients initially being asymptomatic. About 15-30% of patients with chronic hepatitis due to HCV develop cirrhosis within about 20 years. (Hepatitis C Fact Sheet, World Health
  • the compounds are D-amino acid phosphoramidate or thiophosphoramidate nucleoside compounds.
  • the compounds display high tissue levels of active species, remarkable efficacy, or bioavailability, or all, for the treatment of, for example, liver disease and conditions in a human in need thereof.
  • Some of the compounds are based, in part, on the discovery that the active component of certain therapeutic moieties linked to D-amino acids can accumulate favorably in liver cells when the compounds are administered to subjects.
  • the compounds provided herein are useful in the prevention and treatment of Flaviviridae infections and other related conditions such as anti- Flaviviridae antibody positive and Flaviviridae-positivQ conditions, chronic liver inflammation caused by HCV, cirrhosis, fibrosis, acute hepatitis, fulminant hepatitis, chronic persistent hepatitis and fatigue.
  • These compounds or formulations can also be used prophylactically to prevent or retard the progression of clinical illness in individuals who are anti-Flaviviridae antibody or Flaviviridae-antigen positive or who have been exposed to a Flaviviridae.
  • the Flaviviridae is hepatitis C.
  • the compounds are used to treat any virus that replicates through an RNA-dependent R A polymerase.
  • a method for the treatment of a Flaviviridae infection in a host including a human, is also provided that includes administering an effective amount of a compound provided herein, administered either alone or in combination or alternation with another anti- Flaviviridae agent, optionally in a pharmaceutically acceptable carrier.
  • R A is hydrogen, hydroxyl, methyl or halo
  • R B is hydrogen, hydroxyl, halo, cyano, azido, amino, or ethynyl
  • R is hydrogen
  • R is hydrogen or hydroxyl
  • R E is hydrogen or halo; or, in the alternative, R B and R E join to form alkylene or heteroalkylene
  • Base is a nucleobase
  • Ar is aryl
  • R is hydrogen or alkyl.
  • R A is hydrogen, hydroxyl, methyl or halo
  • R B is hydrogen, hydroxyl, halo, cyano, azido or amino
  • R is hydrogen
  • R is hydrogen or hydroxyl
  • R E is hydrogen or halo; or, in the alternative, R B and R E join to form alkylene or heteroalkylene
  • Base is a nucleobase
  • Ar is aryl
  • R is hydrogen or alkyl.
  • L is -O- or -CH 2 -;
  • M is -0-, -S- or -CH 2 -;
  • Z is hydroxyl
  • X is hydroxyl or -O-Ar; or, in the alternative, X and Z combine to form a single divalent -0-;
  • W is O or S
  • Ar is aryl
  • Sc is a side chain of a naturally occurring or non-naturally occurring amino acid;
  • Q 1 is -SR Y , -NR Y R Y , hydroxyl or alkoxyl;
  • R A is hydroxyl or fluoro;
  • R B is hydrogen or methyl;
  • R Y is hydrogen or alkyl;
  • each R 4 is independently hydrogen, hydroxyl, amino, or alkoxyl;
  • each R 5 is independently hydrogen, hydroxyl, or alkoxyl;
  • each R 6 is independently hydrogen, halogen, or alkyl;
  • each R 7 is independently hydrogen, hydroxyl or amino; subject to the proviso that when: Base is 6-methoxyl guanine, L is -0-, M is -0-, W is O, Sc is methyl, R A is hydroxyl, Z is hydroxyl, R B is hydrogen, and Q 1 is -OCH 2 C(CH 2 )3; then X is other than naphthyloxyl; subject to the pro
  • diastereomerically pure compounds according to Formula 1001 useful, for example, for the treatment of flavivirus infections such as HCV infections.
  • the diastereomerically pure compounds display remarkable efficacy or bioavailability, or both, for the treatment of, for example, HCV infection in a human.
  • the diastereomerically pure compounds display increased efficacy or bioavailability, or both, for example, for HCV infection in a human when compared with racemic mixtures or other diastereomers of the same compounds.
  • the compounds provided herein are provided or administered in combination with a second therapeutic agent, such as one useful for the treatment or prevention of HCV infections.
  • a second therapeutic agent such as one useful for the treatment or prevention of HCV infections.
  • Exemplary second therapeutic agents are provided in detail elsewhere herein.
  • compositions, single unit dosage forms, and kits suitable for use in treating or preventing disorders such as HCV infections which comprise a therapeutically or prophylactically effective amount of a compound provided herein, e.g., of Formula AI, I-XVII, 101-152d, 1001-1024b and 2001- 2286b, and a therapeutically or prophylactically effective amount of a second therapeutic agent such as one useful for the treatment or prevention of HCV infections.
  • a compound provided herein e.g., of Formula AI, I-XVII, 101-152d, 1001-1024b and 2001- 2286b
  • a second therapeutic agent such as one useful for the treatment or prevention of HCV infections.
  • the compounds provided herein are useful in the prevention and treatment of Flaviviridae infections and other related conditions such as anti- Flaviviridae antibody positive and Flaviviridae-positive conditions, chronic liver
  • the Flaviviridae is hepatitis C.
  • the compounds are used to treat any virus that replicates through an R A-dependent RNA polymerase.
  • a method of treatment of a liver disorder comprising administering to an individual in need thereof a treatment effective amount of a D- alanine thiophosphoramidate pronucleotide of a nucleoside compound.
  • a method of treatment of a liver disorder comprising administering to an individual in need thereof a treatment effective amount of a compound of Formula AI, I-XVII, 101-152d, 1001-1024b and 2001 -2286b.
  • a method for the treatment of a host infected with a hepatitis C virus comprising the administration of an effective treatment amount of a compound of Formula AI, I-XVII, 101-152d, 1001-1024b and 2001 -2286b or a
  • a method for the treatment of a Flaviviridae infection in a host, including a human, includes administering an effective amount of a compound provided herein, administered either alone or in combination or alternation with another anti- Flaviviridae agent, optionally in a pharmaceutically acceptable carrier.
  • Flaviviridae which can be treated are, e.g., discussed generally in Fields Virology, Sixth Ed., Editors: Knipe, D. M., and Howley, P. M., Lippincott Williams & Wilkins Publishers, Philadelphia, PA, Chapters 25-27, 2013.
  • the Flaviviridae is HCV.
  • the Flaviviridae is a flavivirus or pestivirus.
  • the Flaviviridae can be from any class of Flaviviridae.
  • the Flaviviridae is a mammalian tick-borne virus.
  • the Flaviviridae is a seabird tick-borne virus.
  • the Flaviviridae is a mosquito-borne virus.
  • the Flaviviridae is an Aroa virus.
  • the Flaviviridae is a Dengue virus.
  • certain embodiments, the Flaviviridae is a Dengue virus.
  • the Flaviviridae is a Japanese encephalitis virus. In certain embodiments, the Flaviviridae is a Kokobera virus. In certain embodiments, the Flaviviridae is a Ntaya virus. In certain embodiments, the Flaviviridae is a Spondweni virus. In certain embodiments, the Flaviviridae is a Yellow fever virus. In certain embodiments, the Flaviviridae is a Entebbe virus. In certain embodiments, the Flaviviridae is a Modoc virus. In certain embodiments, the Flaviviridae is a Rio Bravo virus.
  • flaviviruses which can be treated include, without limitation: Absettarov, Aedes, Alfuy, Alkhurma, aba, Aroa, Bagaza, Banzi, Bukalasa bat, Bouboui, Bussuquara, Cacipacore, Calbertado, Carey Island, Cell fusing agent, Cowbone Ridge, Culex, Dakar bat, Dengue 1, Dengue 2, Dengue 3, Dengue 4, Edge Hill, Entebbe bat, Gadgets Gully,
  • leukoencephalitis Murray valley encephalitis, Nakiwogo, Naranjal, Negishi, Ntaya, Omsk hemorrhagic fever, Phnom-Penh bat, Powassan, Quang Binh, Rio Bravo, Rocio, Royal Farm, Russian spring-summer encephalitis, Saboya, St. Louis encephalitis, Sal Vieja, San Perlita, Saumarez Reef, Sepik, Sokuluk, Spondweni, Stratford, Tembusu, Tick-borne encephalitis, Turkish sheep encephalitis, Tyuleniy, Kenya S, Usutu, Wesselsbron, West Nile, Yaounde, Yellow fever, Yokose, and Zika.
  • Pestiviruses which can be treated are discussed generally in Fields Virology, Sixth Ed., Editors: Knipe, D. M., and Howley, P. M., Lippincott Williams & Wilkins Publishers, Philadelphia, PA, Chapter 25, 2013.
  • Specific pestiviruses which can be treated include, without limitation: bovine viral diarrhea virus ("BVDV”), classical swine fever virus
  • CSFV also called hog cholera virus
  • BDV border disease virus
  • compositions and methods useful for treating liver disorders such as HCV infection in a subject are provided herein. Further provided are dosage forms useful for such methods.
  • alkyl refers to a saturated straight or branched hydrocarbon.
  • the alkyl group is a primary, secondary, or tertiary hydrocarbon.
  • the alkyl group includes one to ten carbon atoms, i.e., C ⁇ to Cio alkyl.
  • the alkyl group is methyl, CF 3 , CC1 3 , CFC1 2 , CF 2 CI, ethyl, CH 2 CF 3 , CF 2 CF 3 , propyl, isopropyl, butyl, isobutyl, secbutyl, t- butyl, pentyl, isopentyl, neopentyl, hexyl, isohexyl, 3-methylpentyl, 2,2-dimethylbutyl, or 2,3- dimethylbutyl.
  • the term includes both substituted and unsubstituted alkyl groups, including halogenated alkyl groups.
  • the alkyl group is a fluorinated alkyl group.
  • moieties with which the alkyl group can be substituted include halogen (fluoro, chloro, bromo or iodo), oxo, epoxy, hydroxyl, carbonyl, cycloalkyl, aralkyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al. , Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991 , hereby incorporated by reference.
  • lower alkyl refers to a saturated straight or branched hydrocarbon having one to six carbon atoms, i.e., C ⁇ to C 6 alkyl.
  • the lower alkyl group is a primary, secondary, or tertiary hydrocarbon. The term includes both substituted and unsubstituted moieties.
  • upper alkyl refers to a saturated straight or branched hydrocarbon having seven to thirty carbon atoms, i.e., C 7 to C30 alkyl.
  • the upper alkyl group is a primary, secondary, or tertiary hydrocarbon. The term includes both substituted and unsubstituted moieties.
  • cycloalkyl refers to a saturated cyclic hydrocarbon.
  • the cycloalkyl group may be a saturated, and/or bridged, and/or non-bridged, and/or a fused bicyclic group.
  • the cycloalkyl group includes three to ten carbon atoms, i.e., C 3 to C 10 cycloalkyl.
  • the cycloalkyl has from 3 to 15 (C 3-15 ), from 3 to 10 (C3-10), or from 3 to 7 (C3_ 7 ) carbon atoms.
  • the cycloalkyl group is
  • bicyclo[2.1.1]hexyl bicyclo[2.2.1]heptyl, decalinyl or adamantyl.
  • the term includes both substituted and unsubstituted cycloalkyl groups, including halogenated cycloalkyl groups.
  • the cycloalkyl group is a fluorinated cycloalkyl group.
  • Non-limiting examples of moieties with which the cycloalkyl group can be substituted include halogen (fluoro, chloro, bromo or iodo), oxo, epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • halogen fluoro, chloro, bromo or iodo
  • oxo epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • Cyclopropylene refers to a divalent cyclopropane group.
  • a cyclopropylene group is of formula [0028] "Oxiranylene,” as used herein, refers to a divalent oxirane group.
  • a oxiranylene group is of formula
  • Alkylene refers to divalent saturated aliphatic hydrocarbon groups, including those having from one to eleven carbon atoms which can be straight-chained or branched. In certain embodiments, the alkylene group contains 1 to 10 carbon atoms. The term includes both substituted and unsubstituted moieties. This term is exemplified by groups such as methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), the propylene isomers ⁇ e.g., -CH 2 CH 2 CH 2 - and - CH(CH 3 )CH 2 -) and the like. The term includes halogenated alkylene groups.
  • the alkylene group is a fluorinated alkylene group.
  • moieties with which the alkylene group can be substituted include halogen (fluoro, chloro, bromo or iodo), oxo, epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, alkylaryl, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, and phosphonate, either unprotected, or protected as necessary.
  • alkenyl refers to monovalent olefmically unsaturated hydrocarbon groups, in certain embodiments, having up to about 11 carbon atoms, including from 2 to 8 carbon atoms, or from 2 to 6 carbon atoms, which can be straight-chained or branched and having at least 1 , including from 1 to 2, site of olefmic unsaturation. The term includes both substituted and unsubstituted moieties.
  • the term includes halogenated alkenyl groups.
  • the alkenyl group is a fluorinated alkenyl group.
  • moieties with which the alkenyl group can be substituted include halogen (fluoro, chloro, bromo or iodo), oxo, epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • cycloalkenyl refers to an unsaturated cyclic hydrocarbon.
  • cycloalkenyl refers to mono- or multicyclic ring systems that include at least one double bond.
  • the cycloalkenyl group may be a bridged, non-bridged, and/or a fused bicyclic group.
  • the cycloalkyl group includes at least three carbon atoms, including three to ten carbon atoms, i.e., C 3 to C 10 cycloalkyl.
  • the cycloalkenyl has from 3 to 10 (C3-10), or from 4 to 7 (C4-7) carbon atoms.
  • the term includes both substituted and unsubstituted cycloalkenyl groups, including halogenated cycloalkenyl groups.
  • the cycloalkenyl group is a fluorinated cycloalkenyl group.
  • Non- limiting examples of moieties with which the cycloalkenyl group can be substituted include halogen (fluoro, chloro, bromo or iodo), oxo, epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • halogen fluoro, chloro, bromo or iodo
  • oxo epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • Alkenylene refers to divalent olefmically unsaturated hydrocarbon groups, in certain embodiments, having up to about 11 carbon atoms or from 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 or from 1 to 2 sites of olefmic unsaturation.
  • the term includes both substituted and unsubstituted alkenylene groups, including halogenated alkenylene groups.
  • the alkenylene group is a fluorinated alkenylene group.
  • moieties with which the alkenylene group can be substituted include halogen (fluoro, chloro, bromo or iodo), oxo, epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • Alkynyl refers to acetylenically unsaturated hydrocarbon groups, in certain embodiments, having up to about 11 carbon atoms or from 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 or from 1 to 2 sites of alkynyl unsaturation.
  • alkynyl groups include acetylenic, ethynyl (-C ⁇ CH), propargyl (-CH 2 C ⁇ CH), and the like.
  • the term includes both substituted and unsubstituted alkynyl groups, including halogenated alkynyl groups.
  • the alkynyl group is a fluorinated alkynyl group.
  • Non- limiting examples of moieties with which the alkynyl group can be substituted include halogen (fluoro, chloro, bromo or iodo), oxo, epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • halogen fluoro, chloro, bromo or iodo
  • oxo epoxy, hydroxyl, carbonyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • aryl refers to a substituent derived from an aromatic ring.
  • an aryl group is a C 6 -Ci 2 aryl group.
  • an aryl group is phenyl, biphenyl or naphthyl. The term includes both substituted and unsubstituted moieties.
  • An aryl group can be substituted with any described moiety, including, but not limited to, one or more moieties selected from the group halogen (fluoro, chloro, bromo or iodo), alkyl, haloalkyl, hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al. , Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • halogen fluoro, chloro, bromo or iodo
  • Alkoxy and alkoxyl refer to the group -OR' where R' is alkyl or cycloalkyl as defined herein. Alkoxy and alkoxyl groups include, by way of example, methoxy, ethoxy, n- propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1 ,2- dimethylbutoxy, and the like.
  • Alkylcarbonyloxy refers to a radical -0-C(0)-alkyl, wherein alkyl is as defined herein.
  • Alkoxylcarbonyl refers to a radical -C(0)-alkoxyl where alkoxyl is as defined herein.
  • Alkoxylalkylcarbonyl refers to a radical -C(0)-alkyl-alkoxyl where alkoxyl and alkyl are as defined herein.
  • Alkoxylcarbonylalkyl refers to a radical -alkyl-C(0)-alkoxyl where alkoxyl and alkyl are as defined herein.
  • Alkoxylcarbonylamino refers to a radical -amino-C(0)-alkoxyl where alkoxyl and amino are as defined herein.
  • alkylcarbonylthioalkyl refers to a radical -alkyl-S-C(0)-alkyl, where alkyl is as defined herein.
  • (alkoxylcarbonyl)(alkoxylcarbonylamino)alkyl refers to an alkyl radical substituted with both an alkoxylcarbonyl and an alkoxylcarbonylamino group, where “alkoxylcarbonyl” and “alkoxylcarbonylamino” are as described herein.
  • the alkoxylcarbonyl and “alkoxylcarbonylamino” are as described herein.
  • each of R 100 and R 101 is independently lower alkyl. In an embodiment, each of R 100 and R 101 is independently C1-C5 alkyl.
  • Amino refers to the group -NR r R 2 or -NR 1' -, wherein R 1' and R 2' are
  • alkylamino or "arylamino” refers to an amino group that has one or two alkyl or aryl substituents, respectively.
  • the alkyl substituent is an upper alkyl.
  • the alkyl substituent is a lower alkyl.
  • the alkyl, upper alkyl, or lower alkyl is unsubstituted.
  • Epoxy refers to an oxygen atom bonded to two carbon atoms, where the two carbon atoms are also bonded to each other.
  • Amino alcohol refers to the radical -NHLOH, wherein L is alkylene.
  • Carboxyl or “carboxy” refers to the radical -C(0)OH.
  • Halogen or "halo” refers to chloro, bromo, fluoro, or iodo.
  • “Monoalkylamino” refers to the group -NR'-alkyl, wherein R' is selected from hydrogen, alkyl, and cycloalkyl.
  • Thioalkoxyl refers to the group -SR' where R' is alkyl or cycloalkyl.
  • heterocyclo refers to a monovalent monocyclic non- aromatic ring system and/or multicyclic ring system that contains at least one non-aromatic ring, wherein one or more of the non-aromatic ring atoms are heteroatoms independently selected from O, S, or N; and the remaining ring atoms are carbon atoms.
  • the heterocyclo or heterocyclic group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms.
  • Heterocyclo groups are bonded to the rest of the molecule through the non-aromatic ring.
  • the heterocyclo is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include a fused or bridged ring system, and in which the nitrogen or sulfur atoms may be optionally oxidized, the nitrogen atoms may be optionally quaternized, and some rings may be partially or fully saturated, or aromatic.
  • the heterocyclo may be attached to the main structure at any heteroatom or carbon atom which results in the creation of a stable compound. Examples of such heterocyclic radicals include, but are not limited to, azepinyl, benzodioxanyl,
  • benzodioxolyl benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl, benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, ⁇ -carbolinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, decahydroisoquinolinyl, dihydrobenzisothiazinyl,
  • heterocyclic may also be optionally substituted as described herein.
  • Non-limiting examples of moieties with which the heterocyclic group can be substituted include halogen (fluoro, chloro, bromo or iodo), oxo, epoxy, hydroxyl, carbonyl, alkoxycarbonyl, alkoxycarbonylalkyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary.
  • halogen fluoro, chloro, bromo or iodo
  • oxo epoxy, hydroxyl, carbonyl, alkoxycarbonyl, alkoxycarbonylalkyl, sulfanyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate,
  • heteroaryl refers to a monovalent monocyclic aromatic group and/or multicyclic aromatic group that contain at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from O, S and N in the ring. Heteroaryl groups are bonded to the rest of the molecule through the aromatic ring.
  • Each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, and/or one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom.
  • the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms.
  • Examples of monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl and triazolyl.
  • Examples of bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, benzimidazolyl,
  • benzotriazolyl benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl,
  • heteroaryl examples include, but are not limited to, acridinyl, benzindolyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl and xanthenyl.
  • heteroaryl may also be optionally substituted as described herein.
  • alkylaryl refers to an aryl group with an alkyl substituent, wherein aryl and alkyl are as defined herein.
  • aralkyl or arylalkyl refers to an alkyl group with an aryl substituent, wherein aryl and alkyl are as defined herein.
  • alkylheterocyclo refers to a heterocyclo group with an alkyl substituent.
  • heterocycloalkyl refers to an alkyl group with a heterocyclo substituent.
  • alkylheteroaryl refers to a heteroaryl group with an alkyl substituent.
  • heteroarylalkyl refers to an alkyl group with a heteroaryl substituent.
  • hydantoinyl refers to the group , where
  • R and R are each independently hydrogen or lower alkyl.
  • hydantoinylalkyl refers to the group -alkyl -hydantoinyl, where alkyl and hydantoinyl are as described herein.
  • protecting group refers to a group that is added to an oxygen, nitrogen or phosphorus atom to prevent its further reaction or for other purposes.
  • oxygen and nitrogen protecting groups are known to those skilled in the art of organic synthesis.
  • “Pharmaceutically acceptable salt” refers to any salt of a compound provided herein which retains its biological properties and which is not toxic or otherwise undesirable for pharmaceutical use. Such salts may be derived from a variety of organic and inorganic counter- ions well known in the art. Such salts include, but are not limited to: (1) acid addition salts formed with organic or inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, sulfamic, acetic, trifluoroacetic, trichloroacetic, propionic, hexanoic,
  • cyclopentylpropionic glycolic, glutaric, pyruvic, lactic, malonic, succinic, sorbic, ascorbic, malic, maleic, fumaric, tartaric, citric, benzoic, 3-(4-hydroxybenzoyl)benzoic, picric, cinnamic, mandelic, phthalic, lauric, methanesulfonic, ethanesulfonic, 1 ,2-ethane-disulfonic, 2- hydroxyethanesulfonic, benzenesulfonic, 4-chlorobenzenesulfonic, 2-naphthalenesulfonic, 4- toluenesulfonic, camphoric, camphorsulfonic, 4-methylbicyclo[2.2.2]-oct-2-ene-l-carboxylic, glucoheptonic, 3-phenylpropionic, trimethylacetic, tert-butylacetic, lauryl sulfuric, glu
  • Pharmaceutically acceptable salts further include, by way of example only and without limitation, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium and the like, and when the compound contains a basic functionality, salts of non-toxic organic or inorganic acids, such as hydrohalides, e.g.
  • benzenesulfonate (besylate), 4-chlorobenzenesulfonate, 2-naphthalenesulfonate, 4- toluenesulfonate, camphorate, camphorsulfonate, 4-methylbicyclo[2.2.2]-oct-2-ene-l- carboxylate, glucoheptonate, 3-phenylpropionate, trimethylacetate, tert-butyl acetate, lauryl sulfate, gluconate, benzoate, glutamate, hydroxynaphthoate, salicylate, stearate,
  • nucleobase refers to the base portion of a nucleoside or nucleotide.
  • a nucleobase is a purine or pyrimidine base, as defined herein.
  • purine or "pyrimidine” base refers to, but is not limited to, adenine, N 6 - alkylpurines, N 6 -acylpurines (wherein acyl is C(0)(alkyl, aryl, alkylaryl, or arylalkyl), N 6 - benzylpurine, N 6 -halopurine, N 6 -vinylpurine, N 6 -acetylenic purine, N 6 -acyl purine,
  • Purine bases include, but are not limited to, guanine, adenine, hypoxanthine, 7-deazaguanine, 7-fluoro-7- deazaguanine, 7-deazaadenine, 7-fluoro-7-deazaadenine, 2,6-diaminopurine, 2-amino-6- chloropurine, 6-ethoxypurine, 6-methoxylpurine and 6-chloropurine.
  • Functional oxygen and nitrogen groups on the base can be protected as necessary or desired. Suitable protecting groups are well known to those skilled in the art, and include trimethylsilyl,
  • acyl refers to a group of the formula C(0)R', wherein R' is alkyl or cycloalkyl (including lower alkyl), carboxylate reside of amino acid, aryl including phenyl, alkaryl, arylalkyl including benzyl, alkoxyalkyl including methoxymethyl, aryloxyalkyl such as phenoxymethyl; or substituted alkyl (including lower alkyl), aryl including phenyl optionally substituted with chloro, bromo, fluoro, iodo, Ci to C 4 alkyl, or Ci to C 4 alkoxy, sulfonate esters such as alkyl or arylalkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxy-trityl, substituted benzyl, alkaryl, arylalkyl including
  • Aryl groups in the esters optimally comprise a phenyl group.
  • acyl groups include acetyl, trifluoroacetyl, methylacetyl, cyclpropylacetyl, propionyl, butyryl, hexanoyl, heptanoyl, octanoyl, neo-heptanoyl, phenylacetyl, 2-acetoxy-2-phenylacetyl, diphenylacetyl, ⁇ -methoxy-a-trifluoromethyl-phenylacetyl, bromoacetyl, 2-nitro- benzeneacetyl, 4-chloro-benzeneacetyl, 2-chloro-2,2-diphenylacetyl, 2-chloro-2 -phenylacetyl, trimethylacetyl, chlorodifluoroacetyl, perfluoroacetyl, fluoroacetyl, bromodi
  • amino acid refers to naturally occurring and synthetic ⁇ , ⁇ , ⁇ , or ⁇ amino acids, and includes but is not limited to, amino acids found in proteins, i.e. glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine.
  • the amino acid is in the L-configuration.
  • the amino acid is in the D-configuration.
  • the amino acid is provided as a substituent of a compound described herein, wherein the amino acid is a residue selected from alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, ⁇ -alanyl, ⁇ -valinyl, ⁇ -leucinyl, ⁇ - isoleuccinyl, ⁇ -prolinyl, ⁇ -phenylalaninyl, ⁇ -tryptophanyl, ⁇ -methioninyl, ⁇ -glycinyl, ⁇ -
  • amino acid derivative refers to a group derivable from a naturally or non-naturally occurring amino acid, as described and exemplified herein.
  • Amino acid derivatives are apparent to those of skill in the art and include, but are not limited to, ester, amino alcohol, amino aldehyde, amino lactone, and N-methyl derivatives of naturally and non-naturally occurring amino acids.
  • an amino acid derivative is provided as a substituent of a compound described herein, wherein the substituent is -NR X - wherein Q 1 is -SR Y , -NR Y R Y , or alkoxyl, R Y is hydrogen or alkyl, S c is a side chain of a naturally occurring or non-naturally occurring amino acid, G is C 1 -C 2 alkylene, and R x is hydrogen or R x and Sc, together with the atoms to which they are attached, combine to form a five-membered heterocyclic ring.
  • an amino acid derivative is provided as a substituent of a compound described herein, wherein the
  • substituent is -0-C(0)-G(Sc)-NH-Q , wherein Q is hydrogen or alkoxyl, Sc is a side chain of a naturally occurring or non-naturally occurring amino acid and G is C1-C2 alkylene.
  • Q and Sc together with the atoms to which they are attached, combine to form a five-membered heterocyclic ring.
  • G is Ci alkylene and Sc is hydrogen, alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl, aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl, alkylsulfanylalkyl, or hydroxylarylalkyl.
  • an amino acid derivative is provided as a substituent of a compound described herein, wherein the amino acid derivative is in the D-configuration.
  • an amino acid derivative is provided as a substituent of a compound described herein, wherein the amino acid derivative is in the L- configuration.
  • aminoalkyl refers to an alkyl group with an amino substituent, where alkyl and amino are as described herein.
  • hydroxylalkyl and “hydroxyalkyl” refer to an alkyl group with a hydroxyl substituent, where alkyl is as described herein.
  • carboxylalkyl refers to the group -alkyl-C(0)OH, where alkyl is as described herein.
  • aminoaminoalkyl refers to the group -alkyl- amino-C(NH)-amino, where alkyl and amino are as described herein.
  • aminocarbonylalkyl refers to the group -alkyl-C(O)- amino, where alkyl and amino are as described herein.
  • sulfanylalkyl refers to the group -alkyl-SH, where alkyl is as described herein.
  • carbamoylalkyl refers to the group -alkyl-C(O)- amino, where alkyl and amino are as described herein.
  • alkylsulfanylalkyl refers to the group -alkyl-S-alkyl, where alkyl is as described herein.
  • hydroxylarylalkyl refers to the group -alkyl-aryl-OH, where alkyl and aryl are as described herein.
  • alpha refers to a substituent on the same side of the plane of the sugar ring as the 5' carbon and the term “beta” refers to a substituent on the opposite side of the plane of the sugar ring from the 5' carbon.
  • substituent “A” is in the “alpha” position
  • substituent “B” is in the "beta” position with respect to the 5' carbon:
  • the term “substantially free of or “substantially in the absence of with respect to a nucleoside composition can refer to a nucleoside composition that includes at least 85% or 90%> by weight, in certain embodiments, 95%, 98%>, 99%, or 100%) by weight, of the designated stereoisomer of that nucleoside.
  • the compounds are substantially free of undesignated stereoisomers or other compounds.
  • the term “substantially free of or “substantially in the absence of” when used in connection with an article (including, but not limited to, a compound, a salt thereof, a solvate thereof,
  • substantially free of or substantially in the absence of with respect to a solid form can refer to a solid form that includes at least 85% or 90% by weight, in certain embodiments, 95%), 98%), 99%), or 100% by weight, of the designated solid form.
  • the solid form is substantially free of other solid forms.
  • nucleoside composition refers to a nucleoside composition that includes at least 85%, 90%, 95%, 98%, or 99% to 100% by weight, of the nucleoside, the remainder comprising other chemical species or stereoisomers.
  • isolated with respect to a solid form of a compound refers to a solid that includes at least 85%, 90%, 95%, 98%, or 99% to 100% by weight, of such solid form of the compound, the remainder comprising other solid forms of the compound, other compounds, solvents, and/or other impurities.
  • solvent refers to a compound provided herein or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
  • Isotopic composition refers to the amount of each isotope present for a given atom
  • naturally occurring isotopic composition refers to the naturally occurring isotopic composition or abundance for a given atom
  • Atoms containing their natural isotopic composition may also be referred to herein as "non-enriched" atoms.
  • the atoms of the compounds recited herein are meant to represent any stable isotope of that atom.
  • FT or "hydrogen” the position is understood to have hydrogen at its natural isotopic composition.
  • Isotopic enrichment refers to the percentage of incorporation of an amount of a specific isotope at a given atom in a molecule in the place of that atom's natural isotopic abundance. For example, deuterium enrichment of 1% at a given position means that 1% of the molecules in a given sample contain deuterium at the specified position. Because the naturally occurring distribution of deuterium is about 0.0156%, deuterium enrichment at any position in a compound synthesized using non-enriched starting materials is about 0.0156%).
  • the isotopic enrichment of the compounds provided herein can be determined using conventional analytical methods known to one of ordinary skill in the art, including mass spectrometry and nuclear magnetic resonance spectroscopy.
  • “Isotopically enriched” refers to an atom having an isotopic composition other than the natural isotopic composition of that atom. “Isotopically enriched” may also refer to a compound containing at least one atom having an isotopic composition other than the natural isotopic composition of that atom.
  • alkyl As used herein, “alkyl,” “cycloalkyl,” “alkenyl,” “cycloalkenyl,” “alkynyl,” “aryl,” “alkoxy,” “alkoxycarbonyl,” “alkoxycarbonylalkyl,” “amino,” “carboxyl,” “alkylamino,” “arylamino,” “thioalkyoxy,” “heterocyclo,” “heteroaryl,” “alkylheterocyclo,”
  • alkylheteroaryl optionally comprise deuterium at one or more positions where hydrogen atoms are present, and wherein the deuterium composition of the atom or atoms is other than the natural isotopic composition.
  • alkyl cycloalkyl
  • alkenyl cycloalkenyl
  • alkynyl aryl
  • alkoxy alkoxycarbonyl
  • alkoxycarbonylalkyl alkoxycarbonylalkyl
  • carbboxyl alkylamino
  • arylamino thioalkyoxy
  • alkylheteroaryl optionally comprise carbon- 13 at an amount other than the natural isotopic composition.
  • EC50 refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.
  • the IC 50 refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a maximal response in an assay that measures such response.
  • the term "host,” as used herein, refers to any unicellular or multicellular organism in which the virus can replicate, including cell lines and animals, and in certain embodiments, a human. Alternatively, the host can be carrying a part of the Flaviviridae viral genome, whose replication or function can be altered by the compounds of the present invention. The term host specifically includes infected cells, cells transfected with all or part of the Flaviviridae genome and animals, in particular, primates (including chimpanzees) and humans. In most animal applications of the present invention, the host is a human patient. Veterinary applications, in certain indications, however, are clearly anticipated by the present invention (such as chimpanzees).
  • the terms “subject” and “patient” are used interchangeably herein.
  • the terms “subject” and “subjects” refer to an animal, such as a mammal including a non- primate ⁇ e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate ⁇ e.g., a monkey such as a cynomolgous monkey, a chimpanzee and a human), and for example, a human.
  • the subject is refractory or non-responsive to current treatments for hepatitis C infection.
  • the subject is a farm animal ⁇ e.g., a horse, a cow, a pig, etc.) or a pet ⁇ e.g., a dog or a cat). In certain embodiments, the subject is a human.
  • terapéutica agent refers to any agent(s) which can be used in the treatment or prevention of a disorder or one or more symptoms thereof.
  • therapeutic agent includes a compound provided herein.
  • a therapeutic agent is an agent which is known to be useful for, or has been or is currently being used for the treatment or prevention of a disorder or one or more symptoms thereof.
  • “Therapeutically effective amount” refers to an amount of a compound or composition that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease.
  • a “therapeutically effective amount” can vary depending on, inter alia, the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.
  • Treating” or “treatment” of any disease or disorder refers, in certain embodiments, to ameliorating a disease or disorder that exists in a subject. In another embodiment, “treating” or “treatment” includes ameliorating at least one physical parameter, which may be
  • treating includes modulating the disease or disorder, either physically ⁇ e.g., stabilization of a discernible symptom) or physiologically (e.g., stabilization of a physical parameter) or both.
  • treating or “treatment” includes delaying the onset of the disease or disorder.
  • prophylactic agent and “prophylactic agents” as used refer to any agent(s) which can be used in the prevention of a disorder or one or more symptoms thereof.
  • the term “prophylactic agent” includes a compound provided herein.
  • the term “prophylactic agent” does not refer a compound provided herein.
  • a prophylactic agent is an agent which is known to be useful for, or has been or is currently being used to prevent or impede the onset, development, progression and/or severity of a disorder.
  • prophylactically effective amount refers to the amount of a therapy (e.g. , prophylactic agent) which is sufficient to result in the prevention or reduction of the development, recurrence or onset of one or more symptoms associated with a disorder, or to enhance or improve the prophylactic effect(s) of another therapy (e.g., another prophylactic agent).
  • a therapy e.g. , prophylactic agent
  • another therapy e.g., another prophylactic agent
  • D-alanine thiophosphoramidate pronucleotides of nucleoside compounds useful for the treatment of Flaviviridae infections such as HCV infection.
  • the D- alanine thiophosphoramidate pronucleotides of nucleoside compounds can be formed as described herein and used for the treatment of Flaviviridae infections such as HCV infection.
  • R A is hydrogen, hydroxyl, methyl or halo
  • R B is hydrogen, hydroxyl, halo, cyano, azido, amino, or ethynyl
  • R is hydrogen
  • R is hydrogen or hydroxyl
  • R E is hydrogen or halo; or, in the alternative, R B and R E join to form alkylene or heteroalkylene
  • Base is a nucleobase
  • Ar is aryl
  • R is hydrogen or alkyl.
  • R A is hydrogen, hydroxyl, methyl or halo
  • R B is hydrogen, hydroxyl, halo, cyano, azido or amino
  • R c is hydrogen
  • R D is hydrogen or hydroxyl
  • R E is hydrogen or halo; or, in the alternative, R B and R E join to form alkylene or heteroalkylene
  • Base is a nucleobase
  • Ar is aryl
  • R is hydrogen or alkyl.
  • R A is hydrogen, hydroxyl, methyl or halo
  • R B is hydrogen hydroxyl, halo, cyano, azido, amino, or ethynyl
  • R is hydrogen
  • R is hydrogen or hydroxyl
  • R E is hydrogen or halo; or, in the alternative, R B and R E join to form alkylene or
  • Base is a nucleobase
  • Ar is aryl
  • R is hydrogen or alkyl
  • R A is hydrogen, hydroxyl, methyl or halo
  • R B is hydrogen hydroxyl, halo, cyano, azido or amino
  • R c is hydrogen
  • R D is hydrogen or hydroxyl
  • R E is hydrogen or halo; or, in the alternative, R B and R E join to form alkylene or heteroalkylene
  • Base is a nucleobase
  • Ar is aryl
  • R is hydrogen or alkyl.
  • compounds according to Formula Ala, Alb, Ia or lb are provided which are substantially free of corresponding diastereomers and corresponding L- alanine compounds.
  • a composition is provided which is 85%-100% by weight compounds according to Formula Ala, Alb, Ia or lb and 0%-15% by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition is provided which is 90%- 100% by weight compounds according to Formula Ala, Alb, Ia or lb and 0%>-10%> by weight the corresponding diastereomers and/or
  • a composition which is 95%- 100%) by weight compounds according to Formula Ala, Alb, Ia or lb and 0%>-5%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition is provided which is 97%>-100%> by weight compounds according to Formula Ala, Alb, Ia or lb and 0%>-3%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • weight percent is relative to the total weight of compound Ala, Alb, la or lb, the corresponding diastereomers and corresponding L-alanine compounds.
  • R B is hydroxyl; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6- methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2 , 6-diaminopurine .
  • R B is fluoro
  • R is lower alkyl
  • Ar is phenyl
  • Base guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6- ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R B is cyano
  • R is lower alkyl
  • Ar is phenyl
  • Base is guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6- ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R B is amino; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6- ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R B is azido; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6- ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R is ethynyl
  • R is lower alkyl
  • Ar is phenyl
  • Base is guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6- ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • compounds according to Formula Ila or lib are provided which are substantially free of corresponding diastereomers and corresponding L-alanine compounds.
  • a composition is provided which is 85%- 100% by weight compounds according to Formula Ila or lib and 0%>-15 > by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition is provided which is 90%>-100%> by weight compounds according to Formula Ila or lib and 0%)-10%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition which is 95%>-100%> by weight compounds according to Formula Ila or lib and 0%>-5%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition which is 97%>-100%> by weight compounds according to Formula Ila or lib and 0%>-3%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • weight percent is relative to the total weight of compound Ila or lib, the corresponding diastereomers and corresponding L-alanine compounds.
  • R, Ar, Base and R B are as described in the context of Formula I or AI.
  • R, Ar, Base and R B are as described in the context of Formula I or AI.
  • R, Ar, Base and R B are as described in the context of Formula I or AI.
  • R, Ar, Base and R B are as described in the context of Formula I or AI.
  • R, Ar, Base and R B are as described in the context of Formula I or AI.
  • R, Ar, Base and R B are as described in the context of Formula I or AI.
  • R B is cyano.
  • R B is cyano
  • R is lower alkyl.
  • R B is cyano
  • R is lower alkyl
  • Ar is phenyl
  • Base is guanosine, 6-deoxyguanosine, 6- methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2 , 6-diaminopurine .
  • R B is amino; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6- methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2 , 6-diaminopurine .
  • R B is azido; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6- methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2 , 6-diaminopurine .
  • compounds of Formula III, Ilia or Illb wherein R B is ethynyl; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6- methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2 , 6-diaminopurine .
  • compounds according to Formula Ilia or Illb are provided which are substantially free of corresponding diastereomers and corresponding L-alanine compounds.
  • a composition which is 85%- 100% by weight compounds according to Formula Ilia or Illb and 0%>-15 > by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition which is 90%>-100%> by weight compounds according to Formula Ilia or Illb and 0%)-10%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition is provided which is 95%>-100%> by weight compounds according to Formula Ilia or Illb and 0%>-5%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition is provided which is 97%>-100%> by weight compounds according to Formula Ilia or Illb and 0%>-3%> by weight the corresponding diastereomers and/or corresponding L-alanine
  • weight percent is relative to the total weight of compound Ilia or Illb, the corresponding diastereomers and corresponding L-alanine compounds.
  • R, Ar, Base, R A , R B and R E are as described in the context of Formula I or AI.
  • R A is fluoro
  • R B is hydrogen
  • R E is fluoro
  • R is lower alkyl
  • Ar is phenyl
  • Base guanosine, 6- deoxyguanosine, 6-methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R A is hydrogen; R B is fluoro; R E is fluoro; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6- deoxyguanosine, 6-methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R A is hydrogen; R B is hydroxyl; R E is fluoro; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • compounds according to Formula IVa or IVb are provided which are substantially free of corresponding diastereomers and corresponding L-alanine compounds.
  • a composition is provided which is 85%- 100% by weight compounds according to Formula IVa or IVb and 0%>-15 > by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition is provided which is 90%>-100%> by weight compounds according to Formula IVa or IVb and 0%)-10%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition which is 95%>-100%> by weight compounds according to Formula IVa or IVb and 0%>-5%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition which is 97%>-100%> by weight compounds according to Formula IVa or IVb and 0%>-3%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • weight percent is relative to the total weight of compound IVa or IVb, the corresponding diastereomers and corresponding L-alanine compounds.
  • R, Ar, Base, and R A are as described in the context of Formula I or AI, and W is -CH 2 - -O- or -NH-.
  • W is -CH 2 - -O- or -NH-.
  • R A is methyl
  • W is -0-
  • R is lower alkyl
  • Ar is phenyl
  • Base guanosine, 6- deoxyguanosine, 6-methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R A is hydrogen; W is -0-; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R A is hydrogen
  • W is -CH 2 -
  • R is lower alkyl
  • Ar is phenyl
  • Base guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R A is methyl
  • W is -NH-
  • R is lower alkyl
  • Ar is phenyl
  • Base guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • R A is hydrogen; W is -NH-; R is lower alkyl; Ar is phenyl; and Base is guanosine, 6-deoxyguanosine, 6-methoxyguanosine, 6-ethoxyguanosine, adenine, thymine, cytosine, uracil, 5-fluoro-uracil or 2,6-diaminopurine.
  • compounds according to Formula Va or Vb are provided which are substantially free of corresponding diastereomers and corresponding L-alanine compounds.
  • a composition is provided which is 85%- 100% by weight compounds according to Formula Va or Vb and 0%>-15 > by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition is provided which is 90%>-100%> by weight compounds according to Formula Va or Vb and 0%)-10%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds.
  • a composition which is 95%>-100%> by weight compounds according to Formula Va or Vb and 0%>-5%> by weight the corresponding diastereomers and/or corresponding L-alanine compounds. In an embodiment, a composition is provided which is 97%>-100%> by weight compounds according to Formula Va or Vb and 0%>-3%> by weight the corresponding diastereomers and/or corresponding L-alanine
  • weight percent is relative to the total weight of compound Va or Vb, the corresponding diastereomers and corresponding L-alanine
  • each Base is independently: or tautomeric form thereof, wherein: each R is independently hydrogen, hydroxyl, alkoxyl, amino or aminoalkyl; each R 5 is independently hydrogen, hydroxyl, amino, or alkoxyl; each R 6 is independently hydrogen, halogen, or alkyl; and each R 7 is independently hydrogen or amino.
  • R A , R B , R c , R D , R E , Ar, and R are as described in the context of Formula I or AI; each R is independently hydrogen, halogen, or alkyl; and each R is independently hydrogen or -NH 2 .
  • each row provides five structures - one according to the top left structure (1-56), one according to the top middle structure (la-56a), one according to the top right structure (lb-56b), one according to the bottom left structure (lc-56c), and one according to the bottom right structure (lc-56d).
  • the first row provides compound 1 according to the top left structure, compound la according to the top middle structure, compound lb according to the top right structure, compound lc according to the bottom left structure, and compound Id according to the bottom right structure, each with the indicated variables in the row.
  • each row provides five structures - one according to the top left structure (57-104), one according to the top middle structure 57- 104a), one according to the top right structure (57b -104b), one according to the bottom left structure (57c- 104c), and one according to the bottom right structure 57-104d).
  • the first row provides compound 57 according to the top left structure, compound 57a according to the top middle structure, compound 57b according to the top right structure, compound 57c according to the bottom left structure, and compound 57d according to the bottom right structure, each with the indicated variables in the row.
  • each row provides five structures - one according to the top left structure (105-152), one according to the top middle structure (105a-152a), one according to the top right structure (105b-152b), one according to the bottom left structure (lc-56c), and one according to the bottom right structure (105c-152d).
  • the first row provides compound 105 according to the top left structure, compound 105 a according to the top middle structure, compound 105b according to the top right structure, compound 105 c according to the bottom left structure, and compound 105d according to the bottom right structure, each with the indicated variables in the row.
  • compounds of Formula I useful for the treatment of Flaviviridae infections such as HCV infection in a subject in need thereof.
  • the compounds of Formula I can be formed as described herein and used for the treatment of Flaviviridae infections such as HCV infection.
  • L is -O- or -CH 2 -;
  • M is -0-, -S- or -CH 2 -;
  • Z is hydroxyl
  • X is hydroxyl or -O-Ar; or, in the alternative, X and Z combine to form a single divalent -0-;
  • W is O or S
  • Ar is aryl
  • Sc is a side chain of a naturally occurring or non-naturally occurring amino acid;
  • Q 1 is -SR Y , -NR Y R Y , hydroxyl or alkoxyl;
  • R A is hydroxyl or fluoro;
  • R B is hydrogen or methyl;
  • R Y is hydrogen or alkyl;
  • each R 4 is independently hydrogen, hydroxyl, amino, or alkoxyl;
  • each R 5 is independently hydrogen, hydroxyl, or alkoxyl;
  • each R 6 is independently hydrogen, halogen, or alkyl;
  • each R 7 is independently hydrogen, hydroxyl or amino; subject to the proviso that when: Base is 6-methoxyl guanine, L is -0-, M is -0-, W is O, Sc is methyl, R A is hydroxyl, Z is hydroxyl, R B is hydrogen, and Q 1 is -OCH 2 C(CH 2 ) 3 ; then X is other than naphthyloxyl; subject to the
  • R Ci-Cio alkyl
  • Ar is aryl
  • Base Sc and Q 1 are as described in the context of Formula I.
  • a compound according to Formula 1007-1007b wherein Ar is phenyl and Q 1 is isopropyloxyl.
  • each Base is independently: , or a tautomeric form thereof; and Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl,
  • each Base is independently:
  • Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl, aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl, alkylsulfanylalkyl and hydroxylarylalkyl; and wherein Q 1 is alkoxyl.
  • each Base is independently:
  • Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl, aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl, alkylsulfanylalkyl and hydroxylarylalkyl; and wherein Q 1 is Ci-Cio alkoxyl.
  • each Base is independently:
  • Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl, aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl, alkylsulfanylalkyl and hydroxylarylalkyl; and wherein Q 1 is C1-C5 alkoxyl.
  • each Base is independently:
  • Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl, aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl, alkylsulfanylalkyl and hydroxylarylalkyl; and wherein Q 1 is -OCH(CH 3 ) 2 .
  • each Base is independently:
  • each Base is independently:
  • Sc is alkyl; and wherein Q is C 1 -C5 alkoxyl.
  • each Base is independently: , or a tautomeric form thereof;
  • Sc is alkyl; and wherein Q 1 is -OCH(CH 3 ) 2 .
  • each Base is independently:
  • each Base is independently:
  • Sc is C1-C3 alkyl; and wherein Q 1 is -OCH(CH 3 ) 2 .
  • each Base is independently: , , or , or a tautomeric form thereof; and Sc is methyl.
  • each Base is independently:
  • each Base is independently: , , or , or a tautomeric form thereof;
  • R B , L, M, W, Ar, Sc and Q 1 are as described in the context of Formula 1001.
  • a compound according to any of Formulas 1001-1024b wherein Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl,
  • aminoiminoaminoalkyl aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl,
  • alkylsulfanylalkyl and hydroxylarylalkyl are independently selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl,
  • hydroxylalkyl aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl, alkylsulfanylalkyl and hydroxylarylalkyl, and wherein Q 1 is alkoxyl.
  • a compound according to any of Formulas 1001-1024b wherein Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl, aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl, alkylsulfanylalkyl and hydroxylarylalkyl, and wherein Q 1 is Ci-Cio alkoxyl.
  • a compound according to any of Formulas 1001-1024b wherein Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl,
  • aminoiminoaminoalkyl aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl,
  • a compound according to any of Formulas 1001-1024b wherein Sc is selected from the group consisting of alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl, aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl, carbamoylalkyl, alkylsulfanylalkyl and hydroxylarylalkyl, and wherein Q 1 is -OCH(CH 3 ) 2 .
  • provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is alkyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is alkyl and wherein Q 1 is alkoxyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is alkyl and wherein Q 1 is Ci-Cio alkoxyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is alkyl and wherein Q 1 is C 1 -C5 alkoxyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein S c is alkyl and wherein Q 1 is -OCH(CH 3 ) 2 .
  • provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is C 1 -C 3 alkyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is C 1 -C 3 alkyl wherein Q 1 is alkoxyl In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is C 1 -C 3 alkyl and wherein Q 1 is C 1 -C 10 alkoxyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is C 1 -C 3 alkyl and wherein Q 1 is C 1 -C5 alkoxyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is C 1 -C 3 alkyl and wherein Q 1 is -OCH(CH 3 ) 2 .
  • provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is methyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is methyl and wherein Q 1 is alkoxyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is methyl and wherein Q 1 is C 1 -C 10 alkoxyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is methyl and wherein Q 1 is C 1 -C5 alkoxyl. In an embodiment, provided herein is a compound according to any of Formulas 1001-1024b, wherein Sc is methyl and wherein Q 1 is -OCH(CH 3 ) 2 .
  • Formulas 1001-1024b include a chiral amino acid residue linked to a 5'- phosphoramidate group.
  • the amino acid residue has R stereochemistry at the carbon bonded to Sc; i.e. , that it is a D-amino acid residue.
  • the compounds provided herein are based, at least in part, on the discovery that D-amino acid phosphoramidate prodrugs can provide superior human pharmacokinetics including superior accumulation of active nucleoside and nucleotide analogs in target cells, such as liver cells.
  • the compounds provided herein are D-amino acid
  • the compounds provided herein are D-amino acid, Sp phosphoramidate compounds.
  • Any compound provided herein is preferably in the form of a composition that is substantially free of other stereoisomers of the compound, as described herein.
  • each row provides four structures - one according to the left structure (2001-2095), one according to the middle structure (2001a-2095a), and one according to the right structure (2001b-2095b).
  • the first row provides compound 2001 according to the left structure, compound 2001a according to the middle structure, and compound 2001b according to the right structure, each with the indicated variables in the row.
  • each row provides four structures - one according to the left structure (2096-2190), one according to the middle structure (2096a-2190a), and one according to the right structure (2096b-2190b).
  • the first row provides compound 2096according to the left structure, compound 2096a according to the middle structure, and compound 2096b according to the right structure, each with the indicated variables in the row.
  • each row provides four structures - one according to the left structure (2191-2286), one according to the middle structure (2191a-2286a), and one according to the right structure (2191b-2286b). For instance, the first row provides
  • compound 2191 according to the left structure compound 2191a according to the middle structure, and compound 2191b according to the right structure, each with the indicated variables in the row.
  • the 1 ' and 4' carbons of a nucleoside are chiral, their non- hydrogen substituents (the base and the CHOR groups, respectively) can be either cis (on the same side) or trans (on opposite sides) with respect to the sugar ring system.
  • the four optical isomers therefore are represented by the following configurations (when orienting the sugar moiety in a horizontal plane such that the oxygen atom is in the back): cis (with both groups “up”, which corresponds to the configuration of naturally occurring ⁇ -D nucleosides), cis (with both groups “down”, which is a non-naturally occurring ⁇ -L configuration), trans (with the CT substituent "up” and the C4' substituent "down”), and trans (with the CT substituent "down” and the C4' substituent "up”).
  • the "D-nucleosides” are cis nucleosides in a natural configuration and the "L-nucleosides” are cis nucleosides in the non-naturally occurring configuration.
  • optically active materials examples include at least the following. i) physical separation of crystals - a technique whereby macroscopic
  • crystals of the individual enantiomers are manually separated. This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct; ii) simultaneous crystallization - a technique whereby the individual
  • enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state; iii) enzymatic resolutions - a technique whereby partial or complete
  • enzymatic asymmetric synthesis - a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer
  • chemical asymmetric synthesis - a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries
  • the resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer; first- and second-order asymmetric transformations - a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer.
  • kinetic resolutions - this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions; enantiospecific synthesis from non-racemic precursors - a synthetic technique whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis; chiral liquid chromatography - a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase.
  • the stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions; chiral gas chromatography - a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase; xii) extraction with chiral solvents - a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent; xiii) transport across chiral membranes - a technique whereby a racemate is placed in contact with a thin membrane barrier.
  • the barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
  • thiophosphoramidate pronucleotide of a nucleoside compound that comprises a substantially pure designated enantiomer of the D-alanine thiophosphoramidate pronucleotide of a nucleoside compound.
  • the compounds are substantially free of other enantiomers.
  • a composition includes a compound that is at least 85%, 90%, 95%, 98%, 99% or 100% by weight, of the compound, the remainder comprising other chemical species or enantiomers.
  • isotopically enriched compounds including but not limited to isotopically enriched D-alanine thiophosphoramidate pronucleotides of nucleoside compounds.
  • Isotopic enrichment of a drug can be used, for example, to (1) reduce or eliminate unwanted metabolites, (2) increase the half-life of the parent drug, (3) decrease the number of doses needed to achieve a desired effect, (4) decrease the amount of a dose necessary to achieve a desired effect, (5) increase the formation of active metabolites, if any are formed, and/or (6) decrees the production of deleterious metabolites in specific tissues and/or create a more effective drug and/or a safer drug for combination therapy, whether the combination therapy is intentional or not.
  • KIE Kinetic Isotope Effect
  • DKIE Deuterium Kinetic Isotope Effect
  • the magnitude of the DKIE can be expressed as the ratio between the rates of a given reaction in which a C-H bond is broken, and the same reaction where deuterium is substituted for hydrogen.
  • the DKIE can range from about 1 (no isotope effect) to very large numbers, such as 50 or more, meaning that the reaction can be fifty, or more, times slower when deuterium is substituted for hydrogen.
  • High DKIE values may be due in part to a phenomenon known as tunneling, which is a consequence of the uncertainty principle.
  • Tunneling is ascribed to the small mass of a hydrogen atom, and occurs because transition states involving a proton can sometimes form in the absence of the required activation energy. Because deuterium has more mass than hydrogen, it statistically has a much lower probability of undergoing this phenomenon.
  • substitution of tritium (“T”) for hydrogen results in yet a stronger bond than deuterium and gives numerically larger isotope effects.
  • substitution of isotopes for other elements including, but not limited to, 13 C or 14 C for carbon, 33 S, 34 S, or 36 S for sulfur, 15 N for nitrogen, and 17 0 or 18 0 for oxygen, may lead to a similar kinetic isotope effect.
  • the DKIE was used to decrease the hepatotoxicity of halothane by presumably limiting the production of reactive species such as trifluoroacetyl chloride.
  • the animal body expresses a variety of enzymes for the purpose of eliminating foreign substances, such as therapeutic agents, from its circulation system.
  • enzymes include the cytochrome P450 enzymes ("CYPs"), esterases, proteases, reductases, dehydrogenases, and monoamine oxidases, to react with and convert these foreign substances to more polar intermediates or metabolites for renal excretion.
  • CYPs cytochrome P450 enzymes
  • esterases enzymes
  • proteases e.g., reductases, dehydrogenases, and monoamine oxidases
  • Some of the most common metabolic reactions of pharmaceutical compounds involve the oxidation of a carbon-hydrogen (C-H) bond to either a carbon-oxygen (C-O) or carbon-carbon (C-C) pi-bond.
  • C-H carbon-hydrogen
  • C-O carbon-oxygen
  • C-C carbon-carbon
  • the resultant metabolites may be stable or unstable under physiological conditions, and can have
  • isotopic enrichment at certain positions of a compound provided herein will produce a detectable KIE that will affect the pharmacokinetic, pharmacologic, and/or toxicological profiles of a compound provided herein in comparison with a similar compound having a natural isotopic composition.
  • R Base, R A , R B , R c , R D , R E and Ar are as described herein in the context of Formula I or AI.
  • Nucleosides can be prepared according to international application nos. PCT/US 13/063731 and PCT/US13/065892, and U.S. provisional application nos. 61/807,266, 61/807,268, 61/726,521, 61/726,522, 61/889,384, and 61/807,259, the contents of which are hereby incorporated by reference in their entireties.
  • one or more protection or deprotection steps may be included in the methods of preparation described in Exemplary Preparation Scheme 1.
  • provided herein is a compound prepared according to the above Exemplary Preparation Scheme 1.
  • D-alanine thiophosphoramidate pronucleotides of nucleoside compounds can be formulated into pharmaceutical compositions using methods available in the art and those disclosed herein. Any of the compounds disclosed herein can be provided in the appropriate pharmaceutical composition and be administered by a suitable route of administration.
  • compositions containing at least one compound as described herein including a compound of general Formula AI, I-XVII, 101-152d, 1001-1024b and 2001 -2286b, if appropriate in the salt form, either used alone or in the form of a combination with one or more compatible and pharmaceutically acceptable carriers, such as diluents or adjuvants, or with another anti-HCV agent.
  • the second agent can be formulated or packaged with the compound provided herein.
  • the second agent will only be formulated with the compound provided herein when, according to the judgment of those of skill in the art, such co-formulation should not interfere with the activity of either agent or the method of administration.
  • the compound provided herein and the second agent are formulated separately. They can be packaged together, or packaged separately, for the convenience of the practitioner of skill in the art.
  • the active agents provided herein may be administered by any conventional route, in particular orally, parenterally, rectally or by inhalation ⁇ e.g. in the form of aerosols).
  • the compound provided herein is administered orally.
  • compositions for oral administration may be made, as solid compositions for oral administration, of tablets, pills, hard gelatin capsules, powders or granules.
  • the active product is mixed with one or more inert diluents or adjuvants, such as sucrose, lactose or starch.
  • compositions can comprise substances other than diluents, for example a lubricant, such as magnesium stearate, or a coating intended for controlled release.
  • a lubricant such as magnesium stearate
  • Use may be made, as liquid compositions for oral administration, of solutions which are pharmaceutically acceptable, suspensions, emulsions, syrups and elixirs containing inert diluents, such as water or liquid paraffin.
  • These compositions can also comprise substances other than diluents, for example wetting, sweetening or flavoring products.
  • compositions for parenteral administration can be emulsions or sterile solutions. Use may be made, as solvent or vehicle, of propylene glycol, a polyethylene glycol, vegetable oils, in particular olive oil, or injectable organic esters, for example ethyl oleate. These compositions can also contain adjuvants, in particular wetting, isotonizing, emulsifying, dispersing and stabilizing agents. Sterilization can be carried out in several ways, for example using a bacteriological filter, by radiation or by heating. They can also be prepared in the form of sterile solid compositions which can be dissolved at the time of use in sterile water or any other injectable sterile medium.
  • compositions for rectal administration are suppositories or rectal capsules which contain, in addition to the active principle, excipients such as cocoa butter, semisynthetic glycerides or polyethylene glycols.
  • compositions can also be aerosols.
  • the compositions can be stable sterile solutions or solid compositions dissolved at the time of use in apyrogenic sterile water, in saline or any other pharmaceutically acceptable vehicle.
  • the active principle is finely divided and combined with a water-soluble solid diluent or vehicle, for example dextran, mannitol or lactose.
  • compositions provided herein is a pharmaceutical composition or a single unit dosage form.
  • Pharmaceutical compositions and single unit dosage forms provided herein comprise a prophylactically or therapeutically effective amount of one or more prophylactic or therapeutic agents (e.g. , a compound provided herein, or other prophylactic or therapeutic agent), and a typically one or more pharmaceutically acceptable carriers or excipients.
  • prophylactic or therapeutic agents e.g. , a compound provided herein, or other prophylactic or therapeutic agent
  • “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier includes a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water can be used as a carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Examples of suitable pharmaceutical carriers are described in
  • Typical pharmaceutical compositions and dosage forms comprise one or more excipients.
  • Suitable excipients are well-known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • composition or dosage form Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a subject and the specific active ingredients in the dosage form.
  • the composition or single unit dosage form if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • Lactose free compositions can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmocopia (USP) SP (XXI)/NF (XVI).
  • lactose free compositions comprise an active ingredient, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts.
  • Exemplary lactose free dosage forms comprise an active ingredient, microcrystalline cellulose, pre gelatinized starch, and magnesium stearate.
  • anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water e.g., 5%
  • Anhydrous pharmaceutical compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine can be anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions can be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose.
  • compounds which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • compositions and single unit dosage forms can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • Oral formulation can include standard carriers such as
  • compositions and dosage forms will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent, in certain embodiments, in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject.
  • the formulation should suit the mode of administration.
  • the pharmaceutical compositions or single unit dosage forms are sterile and in suitable form for administration to a subject, for example, an animal subject, such as a mammalian subject, for example, a human subject.
  • a pharmaceutical composition is formulated to be compatible with its intended route of administration.
  • routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, intramuscular, subcutaneous, oral, buccal, sublingual, inhalation, intranasal, transdermal, topical, transmucosal, intra-tumoral, intra-synovial and rectal administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal or topical administration to human beings.
  • a pharmaceutical composition is formulated in accordance with routine procedures for subcutaneous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocamne to ease pain at the site of the injection.
  • Examples of dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a subject, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil in water emulsions, or a water in oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a subject; and sterile solids (e.g. , crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a subject.
  • suspensions e.g., aqueous or non
  • composition, shape, and type of dosage forms provided herein will typically vary depending on their use.
  • a dosage form used in the initial treatment of viral infection may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the maintenance treatment of the same infection.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease or disorder.
  • compositions are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • Typical dosage forms comprise a compound provided herein, or a pharmaceutically acceptable salt, solvate or hydrate thereof lie within the range of from about 0.1 mg to about 1000 mg per day, given as a single once-a-day dose in the morning or as divided doses throughout the day taken with food.
  • Particular dosage forms can have about 0.1 , 0.2, 0.3, 0.4, 0.5, 1.0, 2.0, 2.5, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 100, 200, 250, 500 or 1000 mg of the active compound.
  • compositions that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets ⁇ e.g., chewable tablets), caplets, capsules, and liquids ⁇ e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences, 20th ed., Mack Publishing, Easton PA (2000).
  • the oral dosage forms are solid and prepared under anhydrous conditions with anhydrous ingredients, as described in detail herein.
  • anhydrous ingredients as described in detail herein.
  • the scope of the compositions provided herein extends beyond anhydrous, solid oral dosage forms. As such, further forms are described herein.
  • Typical oral dosage forms are prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms include, but are not limited to, starches, sugars, micro crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or non-aqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms include, but are not limited to, binders, fillers, disintegrants, and lubricants. Binders suitable for use in
  • compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl
  • fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre gelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL PH 101 , AVICEL PH 103 AVICEL RC 581 , AVICEL PH 105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof.
  • a specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC 581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL PH 103TM and Starch 1500 LM.
  • Disintegrants are used in the compositions to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. Typical
  • compositions comprise from about 0.5 to about 15 weight percent of disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms include, but are not limited to, agar, alginic acid, calcium carbonate, micro crystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB O SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • AEROSIL 200 a syloid silica gel
  • a coagulated aerosol of synthetic silica marketed by Degussa Co. of Piano, TX
  • CAB O SIL a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA
  • lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • Active ingredients such as the compounds provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.:
  • Such dosage forms can be used to provide slow or controlled release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients provided herein.
  • single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled release.
  • controlled release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts.
  • the use of an optimally designed controlled release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled release formulations include extended activity of the drug, reduced dosage frequency, and increased subject compliance.
  • controlled release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • the drug may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of
  • a pump may be used (see, Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321 :574 (1989)).
  • polymeric materials can be used.
  • a controlled release system can be placed in a subject at an appropriate site determined by a practitioner of skill, i.e., thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release, vol. 2, pp. 115-138 (1984)).
  • the active ingredient can be dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethyl ene/ethyl acrylate copoly
  • a solid inner matrix e.g., polymethylmethacrylate, polybutyl
  • parenteral dosage forms can be administered to subjects by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial.
  • parenteral dosage forms are typically, sterile or capable of being sterilized prior to administration to a subject.
  • parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
  • Suitable vehicles that can be used to provide parenteral dosage forms are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • Transdermal, topical, and mucosal dosage forms include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences, 16 th , 18th and 20 th eds., Mack Publishing, Easton PA (1980, 1990 & 2000); and Introduction to
  • Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels.
  • transdermal dosage forms include "reservoir type” or “matrix type” patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
  • Suitable excipients ⁇ e.g. , carriers and diluents
  • other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed herein are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
  • excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane 1,3 diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are nontoxic and pharmaceutically acceptable.
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington's Pharmaceutical Sciences, 16 th , 18th and 20 th eds., Mack Publishing, Easton PA (1980, 1990 & 2000).
  • penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
  • Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various water soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery enhancing or penetration enhancing agent.
  • Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
  • doses are from about 1 to about 1000 mg per day for an adult, or from about 5 to about 250 mg per day or from about 10 to 50 mg per day for an adult. In certain embodiments, doses are from about 5 to about 400 mg per day or 25 to 200 mg per day per adult. In certain embodiments, dose rates of from about 50 to about 500 mg per day are also contemplated.
  • kits for treating or preventing an HCV infection in a subject by administering, to a subject in need thereof, an effective amount of a compound provided herein, or a pharmaceutically acceptable salt thereof.
  • the amount of the compound or composition which will be effective in the prevention or treatment of a disorder or one or more symptoms thereof will vary with the nature and severity of the disease or condition, and the route by which the active ingredient is administered.
  • the frequency and dosage will also vary according to factors specific for each subject depending on the specific therapy ⁇ e.g., therapeutic or prophylactic agents) administered, the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the subject.
  • exemplary doses of a composition include milligram or microgram amounts of the active compound per kilogram of subject or sample weight (e.g., about 10 micrograms per kilogram to about 50 milligrams per kilogram, about 100 micrograms per kilogram to about 25 milligrams per kilogram, or about 100 microgram per kilogram to about 10 milligrams per kilogram).
  • the dosage administered to a subject is 0.140 mg/kg to 3 mg/kg of the subject's body weight, based on weight of the active compound.
  • the dosage administered to a subject is between 0.20 mg/kg and 2.00 mg/kg, or between 0.30 mg/kg and 1.50 mg/kg of the subject's body weight.
  • the recommended daily dose range of a composition provided herein for the conditions described herein lie within the range of from about 0.1 mg to about 1000 mg per day, given as a single once-a-day dose or as divided doses throughout a day.
  • the daily dose is administered twice daily in equally divided doses.
  • a daily dose range should be from about 10 mg to about 200 mg per day, in other embodiments, between about 10 mg and about 150 mg per day, in further embodiments, between about 25 and about 100 mg per day. It may be necessary to use dosages of the active ingredient outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art.
  • the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with subject response.
  • the dosage of the composition provided herein, based on weight of the active compound, administered to prevent, treat, manage, or ameliorate a disorder, or one or more symptoms thereof in a subject is 0.1 mg/kg, 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 10 mg/kg, or 15 mg/kg or more of a subject's body weight.
  • the dosage of the composition or a composition provided herein administered to prevent, treat, manage, or ameliorate a disorder, or one or more symptoms thereof in a subject is a unit dose of 0.1 mg to 200 mg, 0.1 mg to 100 mg, 0.1 mg to 50 mg, 0.1 mg to 25 mg, 0.1 mg to 20 mg, 0.1 mg to 15 mg, 0.1 mg to 10 mg, 0.1 mg to 7.5 mg, 0.1 mg to 5 mg, 0.1 to 2.5 mg, 0.25 mg to 20 mg, 0.25 to 15 mg, 0.25 to 12 mg, 0.25 to 10 mg, 0.25 mg to 7.5 mg, 0.25 mg to 5 mg, 0.5 mg to 2.5 mg, 1 mg to 20 mg, 1 mg to 15 mg, 1 mg to 12 mg, 1 mg to 10 mg, 1 mg to 7.5 mg, 1 mg to 5 mg, or 1 mg to 2.5 mg.
  • treatment or prevention can be initiated with one or more loading doses of a compound or composition provided herein followed by one or more maintenance doses.
  • the loading dose can be, for instance, about 60 to about 400 mg per day, or about 100 to about 200 mg per day for one day to five weeks.
  • the loading dose can be followed by one or more maintenance doses.
  • each maintenance does is, independently, about from about 10 mg to about 200 mg per day, between about 25 mg and about 150 mg per day, or between about 25 and about 80 mg per day.
  • Maintenance doses can be administered daily and can be administered as single doses, or as divided doses.
  • a dose of a compound or composition provided herein can be administered to achieve a steady-state concentration of the active ingredient in blood or serum of the subject.
  • the steady-state concentration can be determined by measurement according to techniques available to those of skill or can be based on the physical
  • a sufficient amount of a compound or composition provided herein is administered to achieve a steady- state concentration in blood or serum of the subject of from about 300 to about 4000 ng/mL, from about 400 to about 1600 ng/mL, or from about 600 to about 1200 ng/mL.
  • loading doses can be administered to achieve steady-state blood or serum concentrations of about 1200 to about 8000 ng/mL, or about 2000 to about 4000 ng/mL for one to five days.
  • maintenance doses can be administered to achieve a steady- state concentration in blood or serum of the subject of from about 300 to about 4000 ng/mL, from about 400 to about 1600 ng/mL, or from about 600 to about 1200 ng/mL.
  • administration of the same composition may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • administration of the same prophylactic or therapeutic agent may be repeated and the administration may be separated by at least at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • unit dosages comprising a compound, or a pharmaceutically acceptable salt thereof, in a form suitable for administration. Such forms are described in detail herein.
  • the unit dosage comprises 1 to 1000 mg, 5 to 250 mg or 10 to 50 mg active ingredient.
  • the unit dosages comprise about 1, 5, 10, 25, 50, 100, 125, 250, 500 or 1000 mg active ingredient.
  • Such unit dosages can be prepared according to techniques familiar to those of skill in the art.
  • the dosages of the second agents are to be used in the combination therapies provided herein. In certain embodiments, dosages lower than those which have been or are currently being used to prevent or treat HCV infection are used in the combination therapies provided herein.
  • the recommended dosages of second agents can be obtained from the knowledge of those of skill. For those second agents that are approved for clinical use, recommended dosages are described in, for example, Hardman et ah, eds., 1996, Goodman & Gilman's The Pharmacological Basis Of Basis Of Therapeutics 9 th Ed, Mc-Graw-Hill, New York; Physician's Desk Reference (PDR) 57 th Ed., 2003, Medical Economics Co., Inc., Montvale, NJ, which are incorporated herein by reference in its entirety.
  • the therapies are administered less than 5 minutes apart, less than 30 minutes apart, 1 hour apart, at about 1 hour apart, at about 1 to about 2 hours apart, at about 2 hours to about 3 hours apart, at about 3 hours to about 4 hours apart, at about 4 hours to about 5 hours apart, at about 5 hours to about 6 hours apart, at about 6 hours to about 7 hours apart, at about 7 hours to about 8 hours apart, at about 8 hours to about 9 hours apart, at about 9 hours to about 10 hours apart, at about 10 hours to about 11 hours apart, at about 11 hours to about 12 hours apart, at about 12 hours to 18 hours apart, 18 hours to 24 hours apart, 24 hours to 36 hours apart, 36 hours to 48 hours apart, 48 hours to 52 hours apart, 52 hours to 60 hours apart, 60 hours to 72 hours apart, 72 hours to 84 hours apart, 84 hours to 96 hours apart, or 96 hours to 120 hours apart.
  • the therapies are administered no more than 24 hours apart or no more than 48 hours apart. In certain embodiments, two or more therapies are administered within the same patient visit. In other embodiments, the compound provided herein and the second agent are administered concurrently. [00242] In other embodiments, the compound provided herein and the second agent are administered at about 2 to 4 days apart, at about 4 to 6 days apart, at about 1 week part, at about 1 to 2 weeks apart, or more than 2 weeks apart.
  • administration of the same agent may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • administration of the same agent may be repeated and the administration may be separated by at least at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • a compound provided herein and a second agent are administered to a patient, for example, a mammal, such as a human, in a sequence and within a time interval such that the compound provided herein can act together with the other agent to provide an increased benefit than if they were administered otherwise.
  • the second active agent can be administered at the same time or sequentially in any order at different points in time; however, if not administered at the same time, they should be administered sufficiently close in time so as to provide the desired therapeutic or prophylactic effect.
  • the compound provided herein and the second active agent exert their effect at times which overlap.
  • Each second active agent can be administered separately, in any appropriate form and by any suitable route.
  • the compound provided herein is administered before, concurrently or after administration of the second active agent.
  • the compound provided herein and the second agent are cyclically administered to a patient.
  • Cycling therapy involves the administration of a first agent (e.g., a first prophylactic or therapeutic agents) for a period of time, followed by the administration of a second agent and/or third agent (e.g., a second and/or third prophylactic or therapeutic agents) for a period of time and repeating this sequential administration. Cycling therapy can reduce the development of resistance to one or more of the therapies, avoid or reduce the side effects of one of the therapies, and/or improve the efficacy of the treatment.
  • a first agent e.g., a first prophylactic or therapeutic agents
  • second agent and/or third agent e.g., a second and/or third prophylactic or therapeutic agents
  • the compound provided herein and the second active agent are administered in a cycle of less than about 3 weeks, about once every two weeks, about once every 10 days or about once every week.
  • One cycle can comprise the administration of a compound provided herein and the second agent by infusion over about 90 minutes every cycle, about 1 hour every cycle, about 45 minutes every cycle.
  • Each cycle can comprise at least 1 week of rest, at least 2 weeks of rest, at least 3 weeks of rest.
  • the number of cycles administered is from about 1 to about 12 cycles, more typically from about 2 to about 10 cycles, and more typically from about 2 to about 8 cycles.
  • courses of treatment are administered concurrently to a patient, i.e., individual doses of the second agent are administered separately yet within a time interval such that the compound provided herein can work together with the second active agent.
  • one component can be administered once per week in combination with the other components that can be administered once every two weeks or once every three weeks.
  • the dosing regimens are carried out concurrently even if the
  • therapeutics are not administered simultaneously or during the same day.
  • the second agent can act additively or synergistically with the compound provided herein.
  • the compound provided herein is administered concurrently with one or more second agents in the same pharmaceutical composition.
  • a compound provided herein is administered concurrently with one or more second agents in separate pharmaceutical compositions.
  • a compound provided herein is administered prior to or subsequent to administration of a second agent.
  • administration of a compound provided herein and a second agent by the same or different routes of administration, e.g., oral and parenteral.
  • the second active agent when the compound provided herein is administered concurrently with a second agent that potentially produces adverse side effects including, but not limited to, toxicity, can advantageously be administered at a dose that falls below the threshold that the adverse side effect is elicited.
  • kits for use in methods of treatment of a liver disorder such as HCV infections.
  • the kits can include a compound or composition provided herein, a second agent or composition, and instructions providing information to a health care provider regarding usage for treating the disorder. Instructions may be provided in printed form or in the form of an electronic medium such as a floppy disc, CD, or DVD, or in the form of a website address where such instructions may be obtained.
  • a unit dose of a compound or composition provided herein, or a second agent or composition can include a dosage such that when administered to a subject, a therapeutically or prophylactically effective plasma level of the compound or composition can be maintained in the subject for at least 1 days.
  • a compound or composition can be included as a sterile aqueous pharmaceutical composition or dry powder (e.g., lyophilized) composition.
  • suitable packaging includes a solid matrix or material customarily used in a system and capable of holding within fixed limits a compound provided herein and/or a second agent suitable for administration to a subject.
  • materials include glass and plastic (e.g., polyethylene, polypropylene, and polycarbonate) bottles, vials, paper, plastic, and plastic-foil laminated envelopes and the like. If e-beam sterilization techniques are employed, the packaging should have sufficiently low density to permit sterilization of the contents.
  • a method for inhibiting replication of a virus in a host which comprises contacting the host with a therapeutically effective amount of a 3 '-substituted methyl nucleoside disclosed herein, e.g., a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound according to Formula AI, I-XVII, 101-152d, 1001-1024b and 2001- 2286b, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or a tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, prodrug, phosphate, or active metabolite thereof.
  • a 3 '-substituted methyl nucleoside disclosed herein, e.g., a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound according to Formula AI,
  • a method for inhibiting replication of a virus in a cell which comprises contacting the cell with a therapeutically effective amount of a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound disclosed herein, e.g. , a compound of Formula AI, I-XVII, 101-152d, 1001-1024b and 2001 -2286b, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or a tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, prodrug, phosphate, or active metabolite thereof.
  • a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound disclosed herein e.g. , a compound of Formula AI, I-XVII, 101-152d, 1001-1024b and 2001 -2286b, including a single en
  • a method for inhibiting replication of a virus which comprises contacting the virus with a therapeutically effective amount of a D-alanine
  • thiophosphoramidate pronucleotide of a nucleoside compound disclosed herein e.g. , a D- alanine thiophosphoramidate pronucleotide of a nucleoside compound of Formula AI, I- XVII, 101-152d, 1001-1024b and 2001 -2286b, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or a tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, prodrug, phosphate, or active metabolite thereof.
  • a method for inhibiting the activity of a polymerase which comprises contacting the polymerase with a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound disclosed herein, e.g. , a D-alanine thiophosphoramidate
  • pronucleotide of a nucleoside compound of Formula AI, I-XVII, 101-152d, 1001-1024b and 2001 -2286b including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or a tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, prodrug, phosphate, or active metabolite thereof.
  • kits for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound disclosed herein, e.g., a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound of Formula AI, I-XVII, 101-152d, 1001-1024b and 2001 -2286b, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or a tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, prodrug, phosphate, or active metabolite thereof.
  • a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound disclosed herein e.g., a D-alan
  • kits for treating an HCV infection in a subject encompass the step of administering to the subject in need thereof an amount of a compound effective for the treatment or prevention of an HCV infection in combination with a second agent effective for the treatment or prevention of the infection.
  • the compound can be any compound as described herein, and the second agent can be any second agent described in the art or herein.
  • the compound is in the form of a pharmaceutical composition or dosage form, as described elsewhere herein.
  • provided herein are methods for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of a compounds provided herein, or a pharmaceutically acceptable salt thereof.
  • methods for treating an HCV infection in a subject encompass the step of administering to the subject in need thereof an amount of a compound effective for the treatment or prevention of an HCV infection in combination with a second agent effective for the treatment or prevention of the infection.
  • the compound can be any compound as described herein, and the second agent can be any second agent described in the art or herein.
  • the compound is in the form of a pharmaceutical composition or dosage form, as described elsewhere herein.
  • Flaviviridae which can be treated are, e.g., discussed generally in Fields Virology, Fifth Ed., Editors: Knipe, D. M., and Howley, P. M., Lippincott Williams & Wilkins
  • the Flaviviridae is HCV.
  • the Flaviviridae is a flavivirus or pestivirus.
  • the Flaviviridae can be from any class of Flaviviridae.
  • the Flaviviridae is a mammalian tick-borne virus.
  • the Flaviviridae is a seabird tick-borne virus.
  • the Flaviviridae is a mosquito-borne virus.
  • the Flaviviridae is an Aroa virus.
  • the Flaviviridae is a Dengue virus.
  • certain embodiments are a mammalian tick-borne virus.
  • the Flaviviridae is a seabird tick-borne virus.
  • the Flaviviridae is a mosquito-borne virus.
  • the Flaviviridae is an Aroa virus.
  • the Flaviviridae is a Dengue virus.
  • the Flaviviridae is a Japanese encephalitis virus. In certain embodiments, the Flaviviridae is a Kokobera virus. In certain embodiments, the Flaviviridae is a Ntaya virus. In certain embodiments, the Flaviviridae is a Spondweni virus. In certain embodiments, the Flaviviridae is a Yellow fever virus. In certain embodiments, the Flaviviridae is a Entebbe virus. In certain embodiments, the Flaviviridae is a Modoc virus. In certain embodiments, the Flaviviridae is a Rio Bravo virus.
  • flaviviruses which can be treated include, without limitation: Absettarov, Aedes, Alfuy, Alkhurma, aba, Aroa, Bagaza, Banzi, Bukalasa bat, Bouboui, Bussuquara, Cacipacore, Calbertado, Carey Island, Cell fusing agent, Cowbone Ridge, Culex, Dakar bat, Dengue 1, Dengue 2, Dengue 3, Dengue 4, Edge Hill, Entebbe bat, Gadgets Gully, Hanzalova, Hypr, Ilheus, Israel turkey meningoencephalitis, Japanese encephalitis, Jugra, Jutiapa, Kadam, Kamiti River, Karshi, Kedougou, Kokobera, Koutango, Kumlinge, Kunjin, Kyasanur Forest disease, Langat, Louping ill, Meaban, Modoc, Montana myotis leukoencephalitis, Murray valley encephalitis, Nakiwogo, Naranjal, Neg
  • Pestiviruses which can be treated are discussed generally in Fields Virology, Fifth Ed., Editors: Knipe, D. M., and Howley, P. M., Lippincott Williams & Wilkins Publishers, Philadelphia, PA, Chapters 33-35, 2006. Specific pestiviruses which can be treated include, without limitation: bovine viral diarrhea virus ("BVDV”), classical swine fever virus (“CSFV,” also called hog cholera virus), and border disease virus (“BDV”).
  • BVDV bovine viral diarrhea virus
  • CSFV classical swine fever virus
  • BDV border disease virus
  • the subject can be any subject infected with, or at risk for infection with, HCV. Infection or risk for infection can be determined according to any technique deemed suitable by the practitioner of skill in the art.
  • subjects are humans infected with HCV.
  • the subject has never received therapy or prophylaxis for an HCV infection.
  • the subject has previously received therapy or prophylaxis for an HCV infection.
  • the subject has not responded to an HCV therapy.
  • the subject can be a subject that received therapy but continued to suffer from viral infection or one or more symptoms thereof.
  • the subject can be a subject that received therapy but failed to achieve a sustained virologic response.
  • the subject has received therapy for an HCV infection but has failed to show, for example, a 2 logio decline in HCV R A levels after 12 weeks of therapy. It is believed that subjects who have not shown more than 2 logio reduction in serum HCV RNA after 12 weeks of therapy have a 97-100% chance of not responding.
  • the subject is a subject that discontinued an HCV therapy because of one or more adverse events associated with the therapy.
  • the subject is a subject where current therapy is not indicated.
  • certain therapies for HCV are associated with neuropsychiatric events.
  • Interferon (IFN)-alfa plus ribavirin is associated with a high rate of depression.
  • Depressive symptoms have been linked to a worse outcome in a number of medical disorders. Life-threatening or fatal neuropsychiatric events, including suicide, suicidal and homicidal ideation, depression, relapse of drug
  • Interferon-induced depression is a limitation for the treatment of chronic hepatitis C, especially for subjects with psychiatric disorders.
  • Psychiatric side effects are common with interferon therapy and responsible for about 10% to 20% of discontinuations of current therapy for HCV infection.
  • methods of treating or preventing HCV infection in subjects where a neuropsychiatric event, such as depression, or risk of such indicates dose reduction of current HCV therapy.
  • a hemoglobinopathy for instance thalassemia major subjects and sickle-cell anemia subjects
  • the subject has received an HCV therapy and discontinued that therapy prior to administration of a method provided herein. In further embodiments, the subject has received therapy and continues to receive that therapy along with administration of a method provided herein.
  • the methods can be co-administered with other therapy for HBC and/or HCV according to the judgment of one of skill in the art. In certain embodiments, the methods or compositions provided herein can be co-administered with a reduced dose of the other therapy for HBC and/or HCV.
  • the subject can be a subject that has failed to respond to treatment with one or more agents selected from the group consisting of interferon, interferon a, pegylated interferon a, interferon plus ribavirin, interferon a plus ribavirin and pegylated interferon a plus ribavirin.
  • the subject can be a subject that has responded poorly to treatment with one or more agents selected from the group consisting of interferon, interferon a, pegylated interferon a, interferon plus ribavirin, interferon a plus ribavirin and pegylated interferon a plus ribavirin.
  • a pro-drug form of ribavirin such as taribavirin, may also be used.
  • the subject has, or is at risk for, co-infection of HCV with HIV.
  • 30% of HIV subjects are co-infected with HCV and evidence indicates that people infected with HIV have a much more rapid course of their hepatitis C infection.
  • the methods provided herein can be used to treat or prevent HCV infection in such subjects. It is believed that elimination of HCV in these subjects will lower mortality due to end-stage liver disease. Indeed, the risk of progressive liver disease is higher in subjects with severe AIDS-defining immunodeficiency than in those without.
  • kits for treating or preventing HIV infection and HCV infection in subjects in need thereof are provided.
  • the compounds or compositions are administered to a subject following liver transplant.
  • Hepatitis C is a leading cause of liver transplantation in the U.S., and many subjects that undergo liver transplantation remain HCV positive following transplantation.
  • Compounds can be assayed for HCV activity according to any assay known to those of skill in the art.
  • compounds can be assayed for accumulation in liver cells of a subject according to any assay known to those of skill in the art.
  • a compound can be administered to the subject, and a liver cell of the subject can be assayed for the compound or a derivative thereof, e.g. a nucleoside, nucleoside phosphate or nucleoside triphosphate derivative thereof.
  • a D-alanine thiophosphoramidate pronucleotide of a nucleoside compound is administered to cells, such as liver cells, in vivo or in vitro, and the nucleoside triphosphate levels delivered intracellularly are measured, to indicate delivery of the compound and triphosphorylation in the cell.
  • triphosphate can be measured using analytical techniques known in the art. Methods of detecting ddATP are described herein below by way of example, but other nucleoside triphosphates can be readily detected using the appropriate controls, calibration samples and assay techniques. [00273] In certain embodiments, ddATP concentrations are measured in a sample by comparison to calibration standards made from control samples. The ddATP concentrations in a sample can be measured using an analytical method such as HPLC LC MS. In certain embodiments, a test sample is compared to a calibration curve created with known
  • the samples are manipulated to remove impurities such as salts (Na + , K + , etc.) before analysis.
  • the lower limit of quantitation is about - 0.2 pmol / mL for hepatocyte cellular extracts particularly where reduced salt is present.
  • the method allows successfully measuring triphosphate nucleotides formed at levels of 1 - 10,000 pmol per million cells in e.g. cultured hepatocytes and HepG2 cells.
  • the compounds and compositions provided herein are useful in methods of treatment of a liver disorder, that comprise further administration of a second agent effective for the treatment of the disorder, such as HCV infection in a subject in need thereof.
  • a second agent effective for the treatment of the disorder, such as HCV infection in a subject in need thereof.
  • the second agent can be any agent known to those of skill in the art to be effective for the treatment of the disorder, including those currently approved by the FDA.
  • a compound provided herein is administered in
  • a second agent is administered in combination with two second agents.
  • a second agent is administered in combination with two or more second agents.
  • the term "in combination” includes the use of more than one therapy (e.g., one or more prophylactic and/or therapeutic agents).
  • the use of the term “in combination” does not restrict the order in which therapies ⁇ e.g., prophylactic and/or therapeutic agents) are administered to a subject with a disorder.
  • a first therapy ⁇ e.g., a prophylactic or therapeutic agent such as a compound provided herein
  • a prophylactic or therapeutic agent such as a compound provided herein
  • a second therapy e.g., a prophylactic
  • the term "synergistic” includes a combination of a compound provided herein and another therapy (e.g. , a prophylactic or therapeutic agent) which has been or is currently being used to prevent, manage or treat a disorder, which is more effective than the additive effects of the therapies.
  • a synergistic effect of a combination of therapies permits the use of lower dosages of one or more of the therapies and/or less frequent administration of said therapies to a subject with a disorder.
  • a therapy e.g., a prophylactic or therapeutic agent
  • a synergistic effect can result in improved efficacy of agents in the prevention or treatment of a disorder.
  • a synergistic effect of a combination of therapies e.g., a combination of prophylactic or therapeutic agents
  • the active compounds provided herein can be administered in combination or alternation with another therapeutic agent, in particular an anti-HCV agent.
  • combination therapy effective dosages of two or more agents are administered together, whereas in alternation or sequential-step therapy, an effective dosage of each agent is administered serially or sequentially.
  • the dosages given will depend on absorption, inactivation and excretion rates of the drug as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens and schedules should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions.
  • an anti-HCV (or anti-pestivirus or anti-flavivirus) compound that exhibits an EC50 of 10-15 ⁇ . In certain embodiments, less than 1-5 ⁇ , is desirable.
  • Non-limiting examples of second agents include:
  • HCV Protease inhibitors examples include Medivir HCV Protease Inhibitor (HCV- PI or TMC435) (Medivir/Tibotec); MK-7009 (Merck), RG7227 (ITMN-191)
  • narlaprevir SCH900518 (Schering), narlaprevir SCH900518 (Schering/Merck), ABT-450 (Abbott/Enanta), ACH-1625 (Achillion), BI 201335 (Boehringer Ingelheim), PHX1766 (Phenomix), VX-500 (Vertex) and telaprevir (VX-950) (Vertex).
  • protease inhibitors include substrate-based NS3 protease inhibitors (Attwood et al., Antiviral peptide derivatives, PCT WO 98/22496, 1998; Attwood et al., Antiviral Chemistry and Chemotherapy 1999, 10, 259-273; Attwood et al., Preparation and use of amino acid derivatives as anti-viral agents, German Patent Pub.
  • SCH 351633 isolated from the fungus Penicillium griseofulvum, was identified as a protease inhibitor (Chu M. et al., Bioorganic and Medicinal Chemistry Letters 9: 1949-1952).
  • Eglin c isolated from leech, is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, a-chymotrypsin, chymase and subtilisin. Qasim M.A. et al, Biochemistry 36: 1598-1607, 1997; [00285]
  • U.S. patents disclosing protease inhibitors for the treatment of HCV include, for example, U.S. Patent No.
  • Diaryl peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/48172 and US 6,911,428 to Schering Corporation.
  • Imidazoleidinones as NS3 serine protease inhibitors of HCV are disclosed in WO 02/08198 and US 6,838,475 to Schering Corporation and WO 02/48157 and US 6,727,366 to Bristol Myers Squibb.
  • HCV protease inhibitors include US 6,872,805 (Bristol- Myers Squibb); WO 2006000085 (Boehringer Ingelheim); US 7,208,600 (Vertex); US
  • HCV polymerase inhibitors including nucleoside and non-nucleoside polymerase inhibitors, such as ribavirin, viramidine, clemizole, filibuvir (PF-00868554), HCV POL, NM 283 (valopicitabine), MK-0608, 7-Fluoro-MK-0608, MK-3281, IDX-375, ABT-072, ABT- 333, ANA598, BI 207127, GS 9190, PSI-6130, R1626, PSI-6206, PSI-938, PSI-7851, PSI- 7977, RG1479, RG7128, HCV-796 VCH-759 or VCH-916;
  • nucleoside and non-nucleoside polymerase inhibitors such as ribavirin, viramidine, clemizole, filibuvir (PF-00868554), HCV POL, NM 283 (valopicitabine), MK-0608
  • Interfering RNA (iRNA) based antivirals including short interfering RNA (siRNA) based antivirals, such as Sirna-034 and others described in International Patent Publication Nos. WO/03/070750 and WO 2005/012525, and US Patent Publication No. US 2004/0209831;
  • HCV NS5A inhibitors such as BMS-790052 (daclatasvir, Bristol-Myers Squibb), PPI-461 (Presidio Pharmaceuticals), PPI-1301 (Presidio Pharmaceuticals), IDX-719 (Idenix Pharmaceuticals), AZD7295 (Arrow Therapeutics, AstraZeneca), EDP-239 (Enanta), ACH- 2928 (Achillion), ACH-3102 (Achillion), ABT-267 (Abbott), or GS-5885 (Gilead);
  • BMS-790052 daclatasvir, Bristol-Myers Squibb
  • PPI-461 Presidio Pharmaceuticals
  • PPI-1301 Presidio Pharmaceuticals
  • IDX-719 Idenix Pharmaceuticals
  • AZD7295 Arrow Therapeutics, AstraZeneca
  • EDP-239 Enanta
  • ACH- 2928 Achillion
  • ACH-3102 Achillion
  • ABT-267 Abbott
  • GS-5885 Gilead
  • S-ODN Antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5' non-coding region (NCR) of the virus (Alt M. et al, Hepatology, 1995, 22, 707-717), or nucleotides 326-348 comprising the 3' end of the NCR and nucleotides 371-388 located in the core coding region of the HCV RNA (Alt M. et al, Archives of
  • Inhibitors of IRES-dependent translation (Ikeda N et al. , Agent for the prevention and treatment of hepatitis C, Japanese Patent Pub. JP-08268890; Kai Y. et al, Prevention and treatment of viral diseases, Japanese Patent Pub. JP-10101591);
  • HCV entry inhibitors such as celgosivir (MK-3253) (MIGENIX Inc.), SP-30 (Samaritan Pharmaceuticals), ITX4520 (iTherX), ITX5061 (iTherX), PRO-206 (Progenies Pharmaceuticals) and other entry inhibitors by Progenies Pharmaceuticals, e.g., as disclosed in U.S. Patent Publication No. 2006/0198855;
  • Ribozymes such as nuclease-resistant ribozymes (Maccjak, D. J. et al, Hepatology 1999, 30, abstract 995) and those disclosed in U.S. Patent No. 6,043,077 to Barber et al, and U.S. Patent Nos. 5,869,253 and 5,610,054 to Draper et al; and
  • the compounds provided herein can be administered in combination with any of the compounds described by Idenix Pharmaceuticals in International Publication Nos. WO 01/90121, WO 01/92282, WO 2004/003000, 2004/002422 and WO 2004/002999.

Abstract

La présente invention concerne des composés, des compositions et des méthodes de traitement d'infections par les Flaviviridae, dont les infections par le VHC. Dans certains modes de réalisation, des composés et des compositions de dérivés de nucléoside, qui peuvent être administrés soit seuls, soit en combinaison avec d'autres agents antiviraux, sont divulgués. Dans certains modes de réalisation, les composés sont de Formule AI ou 1001 ou un sel pharmaceutiquement acceptable, un solvate, une forme stéréoisomère, une forme tautomère ou une forme polymorphe de ceux-ci.
PCT/US2014/062174 2013-10-25 2014-10-24 Pronucléotides thiophosphoramidates à acide aminé d et pronucléotides thiophosphoramidates à d-analine de composés de nucléoside utiles pour le traitement du vhc WO2015061683A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361895992P 2013-10-25 2013-10-25
US61/895,992 2013-10-25
US201361909338P 2013-11-26 2013-11-26
US61/909,338 2013-11-26

Publications (1)

Publication Number Publication Date
WO2015061683A1 true WO2015061683A1 (fr) 2015-04-30

Family

ID=51905394

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/062174 WO2015061683A1 (fr) 2013-10-25 2014-10-24 Pronucléotides thiophosphoramidates à acide aminé d et pronucléotides thiophosphoramidates à d-analine de composés de nucléoside utiles pour le traitement du vhc

Country Status (1)

Country Link
WO (1) WO2015061683A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9422323B2 (en) 2012-05-25 2016-08-23 Janssen Sciences Ireland Uc Uracyl spirooxetane nucleosides
WO2018013937A1 (fr) 2016-07-14 2018-01-18 Atea Pharmaceuticals, Inc. Nucléotides de purine beta-d-2'-deoxy-2'-alpha-fluoro-2'-beta-c-substitué-4'-fluoro-n6-substitué-6-amino-2-substitué pour le traitement de l'infection par le virus de l'hépatite c
WO2018048937A1 (fr) 2016-09-07 2018-03-15 Atea Pharmaceuticals, Inc. Nucléotides de purine substitués en position 2'-n 6 pour le traitement du virus à arn
US10202411B2 (en) 2014-04-16 2019-02-12 Idenix Pharmaceuticals Llc 3′-substituted methyl or alkynyl nucleosides nucleotides for the treatment of HCV
US10231986B2 (en) 2013-03-13 2019-03-19 Idenix Pharmaceuticals Llc Amino acid phosphoramidate pronucleotides of 2′-cyano, azido and amino nucleosides for the treatment of HCV
US10238680B2 (en) 2013-08-01 2019-03-26 Idenix Pharmaceuticals Llc D-amino acid phosphoramidate pronucleotides of halogeno pyrimidine compounds for liver disease
US10513534B2 (en) 2012-10-08 2019-12-24 Idenix Pharmaceuticals Llc 2′-chloro nucleoside analogs for HCV infection
US10717758B2 (en) 2012-05-22 2020-07-21 Idenix Pharmaceuticals Llc D-amino acid compounds for liver disease

Citations (112)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US5026687A (en) 1990-01-03 1991-06-25 The United States Of America As Represented By The Department Of Health And Human Services Treatment of human retroviral infections with 2',3'-dideoxyinosine alone and in combination with other antiviral compounds
US5059595A (en) 1989-03-22 1991-10-22 Bioresearch, S.P.A. Pharmaceutical compositions containing 5-methyltetrahydrofolic acid, 5-formyltetrahydrofolic acid and their pharmaceutically acceptable salts in controlled-release form active in the therapy of organic mental disturbances
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5354556A (en) 1984-10-30 1994-10-11 Elan Corporation, Plc Controlled release powder and process for its preparation
US5496546A (en) 1993-02-24 1996-03-05 Jui H. Wang Compositions and methods of application of reactive antiviral polyadenylic acid derivatives
US5538865A (en) 1990-04-06 1996-07-23 Genelabs Technologies, Inc. Hepatitis C virus epitopes
JPH08268890A (ja) 1995-03-31 1996-10-15 Eisai Co Ltd C型肝炎の予防・治療剤
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5610054A (en) 1992-05-14 1997-03-11 Ribozyme Pharmaceuticals, Inc. Enzymatic RNA molecule targeted against Hepatitis C virus
US5633358A (en) 1994-09-14 1997-05-27 Huels Aktiengesellschaft Process for bleaching aqueous surfactant solutions
US5633388A (en) 1996-03-29 1997-05-27 Viropharma Incorporated Compounds, compositions and methods for treatment of hepatitis C
US5639480A (en) 1989-07-07 1997-06-17 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5674533A (en) 1994-07-07 1997-10-07 Recordati, S.A., Chemical And Pharmaceutical Company Pharmaceutical composition for the controlled release of moguisteine in a liquid suspension
WO1997036554A1 (fr) 1996-03-29 1997-10-09 Viropharma Incorporated Derives de la piperidine, compositions pharmaceutiques issues desdits derives et procedes d'utilisation dans le traitement de l'hepatite c
US5725859A (en) 1994-05-03 1998-03-10 Omer; Osama L.M. Plant-based therapeutic agent with virustatic and antiviral effect
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5739108A (en) 1984-10-04 1998-04-14 Monsanto Company Prolonged release of biologically active polypeptides
JPH10101591A (ja) 1996-09-27 1998-04-21 Eisai Co Ltd ウイルス感染症の予防・治療剤
WO1998017679A1 (fr) 1996-10-18 1998-04-30 Vertex Pharmaceuticals Incorporated Inhibiteurs de serines proteases, notamment de ns3 protease du virus de l'hepatite c
WO1998022496A2 (fr) 1996-11-18 1998-05-28 F. Hoffmann-La Roche Ag Derives peptidiques antiviraux
US5837257A (en) 1996-07-09 1998-11-17 Sage R&D Use of plant extracts for treatment of HIV, HCV and HBV infections
US5846964A (en) 1993-07-19 1998-12-08 Tokyo Tanabe Company Limited Hepatitis C virus proliferation inhibitor
WO1999007734A2 (fr) 1997-08-11 1999-02-18 Boehringer Ingelheim (Canada) Ltd. Analogues de peptides inhibiteurs de l'hepatite c
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US5891874A (en) 1996-06-05 1999-04-06 Eli Lilly And Company Anti-viral compound
US5922757A (en) 1996-09-30 1999-07-13 The Regents Of The University Of California Treatment and prevention of hepatic disorders
US5922356A (en) 1996-10-09 1999-07-13 Sumitomo Pharmaceuticals Company, Limited Sustained release formulation
WO1999043691A1 (fr) 1998-02-25 1999-09-02 Emory University 2'-fluoronucleosides
DE19914474A1 (de) 1998-03-30 1999-10-07 Hoffmann La Roche Aminosäurederivate
US5972891A (en) 1992-12-07 1999-10-26 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US5990276A (en) 1996-05-10 1999-11-23 Schering Corporation Synthetic inhibitors of hepatitis C virus NS3 protease
US5993855A (en) 1995-09-18 1999-11-30 Shiseido Company, Ltd. Delayed drug-releasing microspheres
US6004933A (en) 1997-04-25 1999-12-21 Cortech Inc. Cysteine protease inhibitors
WO2000009543A2 (fr) 1998-08-10 2000-02-24 Boehringer Ingelheim (Canada) Ltd. Tri-peptides inhibiteurs de l'hepatite c
US6034134A (en) 1997-06-30 2000-03-07 Merz + Co. Gmbh & Co. 1-Amino-alkylcyclohexane NMDA receptor antagonists
US6043077A (en) 1996-02-29 2000-03-28 Immusol Inc. Hepatitis C virus ribozymes
US6045830A (en) 1995-09-04 2000-04-04 Takeda Chemical Industries, Ltd. Method of production of sustained-release preparation
US6056961A (en) 1996-12-15 2000-05-02 Lavie; David Plant extracts for the preparation of pharmaceutical compositions for the treatment of hepatitis
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6113943A (en) 1996-10-31 2000-09-05 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
US6197350B1 (en) 1996-12-20 2001-03-06 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
WO2001032153A2 (fr) 1999-11-04 2001-05-10 Shire Biochem Inc. Procede de traitement ou de prevention de l'infection virale par flaviviridae faisant appel a des analogues des nucleosides
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6267981B1 (en) 1995-06-27 2001-07-31 Takeda Chemical Industries, Ltd. Method of producing sustained-release preparation
WO2001060315A2 (fr) 2000-02-18 2001-08-23 Shire Biochem Inc. Methode de traitement ou de prevention d'infections a flavivirus a l'aide d'analogues nucleosidiques
WO2001079246A2 (fr) 2000-04-13 2001-10-25 Pharmasset, Ltd. Derives de nucleoside substitues par 3'- ou 2'-hydroxymethyle utilises dans le traitement des infections imputables au virus de l'hepatite
WO2001090121A2 (fr) 2000-05-23 2001-11-29 Idenix (Cayman) Limited Methodes et compositions permettant de traiter le virus de l'hepatite c
WO2001092282A2 (fr) 2000-05-26 2001-12-06 Idenix (Cayman) Limited Procedes et compositions de traitement des flavivirus et des pestivirus
WO2002008251A2 (fr) 2000-07-21 2002-01-31 Corvas International, Inc. Nouveaux peptides utilises comme inhibiteurs de ns3-serine protease du virus de l'hepatite c
WO2002008198A2 (fr) 2000-07-21 2002-01-31 Schering Corporation Nouveaux imidazolidinones comme inhibiteurs de la protease ns3-serine du virus de l'hepatite c
WO2002008256A2 (fr) 2000-07-21 2002-01-31 Schering Corporation Nouveaux peptides utilises comme inhibiteurs de serine ns3 protease du virus de l'hepatite c
WO2002008187A1 (fr) 2000-07-21 2002-01-31 Schering Corporation Nouveaux peptides utilises comme inhibiteurs de la serine protease ns3 du virus de l'hepatite c
WO2002018404A2 (fr) 2000-08-30 2002-03-07 F. Hoffmann-La Roche Ag Derives de nucleosides
WO2002032920A2 (fr) 2000-10-18 2002-04-25 Pharmasset Limited Nucleosides modifies pour traiter des infections virales et une proliferation cellulaire anormale
WO2002048165A2 (fr) 2000-12-15 2002-06-20 Pharmasset Ltd. Agents antiviraux utilises dans le traitement des infections par les flaviviridae
WO2002048157A2 (fr) 2000-12-13 2002-06-20 Bristol-Myers Squibb Pharma Company Imidazolidinones et leurs derives associes, utiles en tant qu'inhibiteurs des proteases ns3 du virus de l'hepatite c
WO2002048116A2 (fr) 2000-12-13 2002-06-20 Bristol-Myers Squibb Pharma Company Inhibiteurs de la protease ns3 du virus de l'hepatite c
WO2002048172A2 (fr) 2000-12-12 2002-06-20 Schering Corporation Peptides diaryliques utilises comme inhibiteurs de ns3-serine protease du virus de l'hepatite c
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
WO2002057287A2 (fr) 2001-01-22 2002-07-25 Merck & Co., Inc. Derives de nucleoside servant d'inhibiteurs de l'arn polymerase virale arn dependante
WO2002060926A2 (fr) 2000-11-20 2002-08-08 Bristol-Myers Squibb Company Inhibiteurs du virus de l'hepatite c
WO2003053349A2 (fr) 2001-12-20 2003-07-03 Bristol-Myers Squibb Company Inhibiteurs de virus de l'hepatite c
US6589548B1 (en) 1998-05-16 2003-07-08 Mogam Biotechnology Research Institute Controlled drug delivery system using the conjugation of drug to biodegradable polyester
WO2003064416A1 (fr) 2002-02-01 2003-08-07 Boehringer Ingelheim International Gmbh Tripeptides heterocycliques utiles en tant qu'inhibiteurs de l'hepatite c
WO2003064456A1 (fr) 2002-02-01 2003-08-07 Boehringer Ingelheim International Gmbh Tripeptides comprenant un hydroxyproline ether d'une quinoline substituee destines a inhiber ns3 (hepatite c)
WO2003070750A2 (fr) 2002-02-20 2003-08-28 Sirna Therapeutics, Inc Inhibition de l'expression du gene du virus de l'hepatite c (vhc) induite par l'interference d'arn au moyen d'acide nucleique interferant court (sina)
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
US6642204B2 (en) 2002-02-01 2003-11-04 Boehringer Ingelheim International Gmbh Hepatitis C inhibitor tri-peptides
US6653295B2 (en) 2000-12-13 2003-11-25 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus NS3 protease
WO2003099274A1 (fr) 2002-05-20 2003-12-04 Bristol-Myers Squibb Company Inhibiteurs du virus de l'hepatite c
WO2003099316A1 (fr) 2002-05-20 2003-12-04 Bristol-Myers Squibb Company Sulfamides heterocycliques en tant qu'inhibiteurs du virus de l'hepatite c
US6660721B2 (en) 2001-05-23 2003-12-09 Hoffmann-La Roche Inc. Anti-HCV nucleoside derivatives
WO2004003000A2 (fr) 2002-06-28 2004-01-08 Idenix (Cayman) Limited Promedicaments 2' et 3' de nucleoside permettant de traiter des infections par les flaviviridae
WO2004002422A2 (fr) 2002-06-28 2004-01-08 Idenix (Cayman) Limited Ester 3'-l-valine de ?-d-2'-c-methyl-ribofuranosyl cytidine pour le traitement d'infections par des flaviviridae
WO2004002999A2 (fr) 2002-06-28 2004-01-08 Idenix (Cayman) Limited Promedicaments a nucleosides 2' et 3' destines a traiter les infections aux flavivirus
WO2004032827A2 (fr) 2002-05-20 2004-04-22 Bristol-Myers Squibb Company Inhibiteurs du virus de l'hepatite c
WO2004043339A2 (fr) 2002-05-20 2004-05-27 Bristol-Myers Squibb Company Inhibiteurs du virus de l'hepatite c a base de cycloalkyle p1' substitue
US20040121980A1 (en) 2002-11-19 2004-06-24 Roche Palo Alto Llc Antiviral nucleoside derivatives
US6784166B2 (en) 2001-06-12 2004-08-31 Syntex (U.S.A.) Llc 4′-substituted nucleoside derivatives as inhibitors of HCV RNA replication.
US20040209831A1 (en) 2002-02-20 2004-10-21 Mcswiggen James RNA interference mediated inhibition of hepatitis C virus (HCV) gene expression using short interfering nucleic acid (siNA)
US20050009737A1 (en) 2003-05-30 2005-01-13 Jeremy Clark Modified fluorinated nucleoside analogues
WO2005012525A1 (fr) 2003-07-25 2005-02-10 Amgen Inc Petit arn interferant utilise en tant qu'agent antiviral pour l'hepatite c
US20050038240A1 (en) 2003-06-19 2005-02-17 Roche Palo Alto Llc Processes for preparing 4'-azido-nucleoside derivatives
US20050153877A1 (en) 2003-02-07 2005-07-14 Zhenwei Miao Macrocyclic hepatitis C serine protease inhibitors
US6927291B2 (en) 2001-03-01 2005-08-09 Pharmasset, Ltd. Method for the synthesis of 2′,3′-dideoxy-2′,3′-didehydronucleosides
WO2006000085A1 (fr) 2004-06-28 2006-01-05 Boehringer Ingelheim International Gmbh Analogues peptidiques d'inhibiteurs de l'hepatite c
US20060040890A1 (en) 2004-08-23 2006-02-23 Roche Palo Alto Llc Anti-viral nucleosides
US20060046956A1 (en) 2004-08-27 2006-03-02 Schering Corporation Acylsulfonamide compounds as inhibitors of hepatitis C virus NS3 serine protease
US7091184B2 (en) 2002-02-01 2006-08-15 Boehringer Ingelheim International Gmbh Hepatitis C inhibitor tri-peptides
US20060198855A1 (en) 2001-06-26 2006-09-07 Progenics Pharmaceuticals, Inc. Uses of DC-SIGN and DC-SIGNR for inhibiting hepatitis C virus infection
US7105499B2 (en) 2001-01-22 2006-09-12 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
WO2006119061A2 (fr) 2005-05-02 2006-11-09 Merck & Co., Inc. Inhibiteurs de la protease ns3 du vhc
WO2007001406A2 (fr) 2004-10-05 2007-01-04 Chiron Corporation Composes macrocycliques contenant un aryle
US7208600B2 (en) 2003-10-10 2007-04-24 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly HCV NS3-NS4A proteases
WO2008121634A2 (fr) * 2007-03-30 2008-10-09 Pharmasset, Inc. Promédicaments de phosphoramidate de nucléoside
WO2010075554A1 (fr) * 2008-12-23 2010-07-01 Pharmasset, Inc. Synthèse de nucléosides de type purine
WO2012088155A1 (fr) * 2010-12-22 2012-06-28 Alios Biopharma, Inc. Analogues de nucléotides cycliques
WO2012142523A2 (fr) * 2011-04-13 2012-10-18 Gilead Sciences, Inc. Analogues de n-nucléoside de pyrimidine 1'-substitués pour un traitement antiviral
WO2013092481A1 (fr) * 2011-12-20 2013-06-27 F. Hoffmann-La Roche Ag Dérivés de nucléoside substitué 2',4'-difluoro-2'-méthyle en tant qu'inhibiteurs de la réplication de l'arn du vhc (virus de l'hépatite c)
WO2013177219A1 (fr) * 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. Composés d'acide d-aminé contre les maladies hépatiques
WO2014078427A1 (fr) * 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. Ester de d-alanine d'analogue de rp-nucléoside
WO2014078436A1 (fr) * 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. Ester de d-alanine d'analogue de sp-nucléoside
WO2014148949A1 (fr) * 2013-03-22 2014-09-25 Асави, Ллс Alkyl 2-{[(2r,3s,5r)-5-(4-amino-2-oxo-2n-pyrimidin-1-yl)-3-hydroxy-tétrahydro-furan-2-ilméthoxy]-phénoxy-phosphorilamino}-propionates, inhibiteurs de nucléosides d'arn-polymérase hcv ns5b, procédés de fabrication et d'utilisation

Patent Citations (138)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US5739108A (en) 1984-10-04 1998-04-14 Monsanto Company Prolonged release of biologically active polypeptides
US5354556A (en) 1984-10-30 1994-10-11 Elan Corporation, Plc Controlled release powder and process for its preparation
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5059595A (en) 1989-03-22 1991-10-22 Bioresearch, S.P.A. Pharmaceutical compositions containing 5-methyltetrahydrofolic acid, 5-formyltetrahydrofolic acid and their pharmaceutically acceptable salts in controlled-release form active in the therapy of organic mental disturbances
US5639480A (en) 1989-07-07 1997-06-17 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5026687A (en) 1990-01-03 1991-06-25 The United States Of America As Represented By The Department Of Health And Human Services Treatment of human retroviral infections with 2',3'-dideoxyinosine alone and in combination with other antiviral compounds
US5538865A (en) 1990-04-06 1996-07-23 Genelabs Technologies, Inc. Hepatitis C virus epitopes
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5610054A (en) 1992-05-14 1997-03-11 Ribozyme Pharmaceuticals, Inc. Enzymatic RNA molecule targeted against Hepatitis C virus
US5869253A (en) 1992-05-14 1999-02-09 Ribozyme Pharmaceuticals, Inc. Method and reagent for inhibiting hepatitis C virus replication
US5972891A (en) 1992-12-07 1999-10-26 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5496546A (en) 1993-02-24 1996-03-05 Jui H. Wang Compositions and methods of application of reactive antiviral polyadenylic acid derivatives
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6376461B1 (en) 1993-06-24 2002-04-23 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5846964A (en) 1993-07-19 1998-12-08 Tokyo Tanabe Company Limited Hepatitis C virus proliferation inhibitor
US5725859A (en) 1994-05-03 1998-03-10 Omer; Osama L.M. Plant-based therapeutic agent with virustatic and antiviral effect
US5674533A (en) 1994-07-07 1997-10-07 Recordati, S.A., Chemical And Pharmaceutical Company Pharmaceutical composition for the controlled release of moguisteine in a liquid suspension
US5633358A (en) 1994-09-14 1997-05-27 Huels Aktiengesellschaft Process for bleaching aqueous surfactant solutions
JPH08268890A (ja) 1995-03-31 1996-10-15 Eisai Co Ltd C型肝炎の予防・治療剤
US6267981B1 (en) 1995-06-27 2001-07-31 Takeda Chemical Industries, Ltd. Method of producing sustained-release preparation
US6045830A (en) 1995-09-04 2000-04-04 Takeda Chemical Industries, Ltd. Method of production of sustained-release preparation
US5993855A (en) 1995-09-18 1999-11-30 Shiseido Company, Ltd. Delayed drug-releasing microspheres
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US6043077A (en) 1996-02-29 2000-03-28 Immusol Inc. Hepatitis C virus ribozymes
US5633388A (en) 1996-03-29 1997-05-27 Viropharma Incorporated Compounds, compositions and methods for treatment of hepatitis C
WO1997036554A1 (fr) 1996-03-29 1997-10-09 Viropharma Incorporated Derives de la piperidine, compositions pharmaceutiques issues desdits derives et procedes d'utilisation dans le traitement de l'hepatite c
US5830905A (en) 1996-03-29 1998-11-03 Viropharma Incorporated Compounds, compositions and methods for treatment of hepatitis C
US5990276A (en) 1996-05-10 1999-11-23 Schering Corporation Synthetic inhibitors of hepatitis C virus NS3 protease
US5891874A (en) 1996-06-05 1999-04-06 Eli Lilly And Company Anti-viral compound
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5837257A (en) 1996-07-09 1998-11-17 Sage R&D Use of plant extracts for treatment of HIV, HCV and HBV infections
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
JPH10101591A (ja) 1996-09-27 1998-04-21 Eisai Co Ltd ウイルス感染症の予防・治療剤
US5922757A (en) 1996-09-30 1999-07-13 The Regents Of The University Of California Treatment and prevention of hepatic disorders
US5922356A (en) 1996-10-09 1999-07-13 Sumitomo Pharmaceuticals Company, Limited Sustained release formulation
US6265380B1 (en) 1996-10-18 2001-07-24 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis C virus NS3 protease
WO1998017679A1 (fr) 1996-10-18 1998-04-30 Vertex Pharmaceuticals Incorporated Inhibiteurs de serines proteases, notamment de ns3 protease du virus de l'hepatite c
US6113943A (en) 1996-10-31 2000-09-05 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
US6699500B2 (en) 1996-10-31 2004-03-02 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
WO1998022496A2 (fr) 1996-11-18 1998-05-28 F. Hoffmann-La Roche Ag Derives peptidiques antiviraux
US6056961A (en) 1996-12-15 2000-05-02 Lavie; David Plant extracts for the preparation of pharmaceutical compositions for the treatment of hepatitis
US6197350B1 (en) 1996-12-20 2001-03-06 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US6004933A (en) 1997-04-25 1999-12-21 Cortech Inc. Cysteine protease inhibitors
US6034134A (en) 1997-06-30 2000-03-07 Merz + Co. Gmbh & Co. 1-Amino-alkylcyclohexane NMDA receptor antagonists
WO1999007734A2 (fr) 1997-08-11 1999-02-18 Boehringer Ingelheim (Canada) Ltd. Analogues de peptides inhibiteurs de l'hepatite c
WO1999043691A1 (fr) 1998-02-25 1999-09-02 Emory University 2'-fluoronucleosides
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
DE19914474A1 (de) 1998-03-30 1999-10-07 Hoffmann La Roche Aminosäurederivate
US6589548B1 (en) 1998-05-16 2003-07-08 Mogam Biotechnology Research Institute Controlled drug delivery system using the conjugation of drug to biodegradable polyester
WO2000009543A2 (fr) 1998-08-10 2000-02-24 Boehringer Ingelheim (Canada) Ltd. Tri-peptides inhibiteurs de l'hepatite c
US6323180B1 (en) 1998-08-10 2001-11-27 Boehringer Ingelheim (Canada) Ltd Hepatitis C inhibitor tri-peptides
US6534523B1 (en) 1998-08-10 2003-03-18 Boehringer Ingelheim (Canada) Ltd. Hepatitis C inhibitor tri-peptides
US6420380B2 (en) 1998-08-10 2002-07-16 Boehringer Ingelheim (Canada) Ltd. Hepatitis C inhibitor tri-peptides
US6410531B1 (en) 1998-08-10 2002-06-25 Boehringer Ingelheim (Canada) Ltd. Hepatitis C inhibitor tri-peptides
WO2001032153A2 (fr) 1999-11-04 2001-05-10 Shire Biochem Inc. Procede de traitement ou de prevention de l'infection virale par flaviviridae faisant appel a des analogues des nucleosides
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
WO2001060315A2 (fr) 2000-02-18 2001-08-23 Shire Biochem Inc. Methode de traitement ou de prevention d'infections a flavivirus a l'aide d'analogues nucleosidiques
US7094770B2 (en) 2000-04-13 2006-08-22 Pharmasset, Ltd. 3′-or 2′-hydroxymethyl substituted nucleoside derivatives for treatment of hepatitis virus infections
WO2001079246A2 (fr) 2000-04-13 2001-10-25 Pharmasset, Ltd. Derives de nucleoside substitues par 3'- ou 2'-hydroxymethyle utilises dans le traitement des infections imputables au virus de l'hepatite
WO2001090121A2 (fr) 2000-05-23 2001-11-29 Idenix (Cayman) Limited Methodes et compositions permettant de traiter le virus de l'hepatite c
WO2001092282A2 (fr) 2000-05-26 2001-12-06 Idenix (Cayman) Limited Procedes et compositions de traitement des flavivirus et des pestivirus
WO2002008187A1 (fr) 2000-07-21 2002-01-31 Schering Corporation Nouveaux peptides utilises comme inhibiteurs de la serine protease ns3 du virus de l'hepatite c
WO2002008256A2 (fr) 2000-07-21 2002-01-31 Schering Corporation Nouveaux peptides utilises comme inhibiteurs de serine ns3 protease du virus de l'hepatite c
WO2002008198A2 (fr) 2000-07-21 2002-01-31 Schering Corporation Nouveaux imidazolidinones comme inhibiteurs de la protease ns3-serine du virus de l'hepatite c
WO2002008251A2 (fr) 2000-07-21 2002-01-31 Corvas International, Inc. Nouveaux peptides utilises comme inhibiteurs de ns3-serine protease du virus de l'hepatite c
US20050176648A1 (en) 2000-07-21 2005-08-11 Schering-Plough Corporation Novel peptides as NS3-serine protease inhibitors of hepatitis C virus
US6838475B2 (en) 2000-07-21 2005-01-04 Schering Corporation Imidazolidinones as NS3-serine protease inhibitors of hepatitis C virus
US7169760B2 (en) 2000-07-21 2007-01-30 Schering Corporation Peptides as NS3-serine protease inhibitors of hepatitis C virus
WO2002018404A2 (fr) 2000-08-30 2002-03-07 F. Hoffmann-La Roche Ag Derives de nucleosides
WO2002032920A2 (fr) 2000-10-18 2002-04-25 Pharmasset Limited Nucleosides modifies pour traiter des infections virales et une proliferation cellulaire anormale
US6872805B2 (en) 2000-11-20 2005-03-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2002060926A2 (fr) 2000-11-20 2002-08-08 Bristol-Myers Squibb Company Inhibiteurs du virus de l'hepatite c
US6911428B2 (en) 2000-12-12 2005-06-28 Schering Corporation Diaryl peptides as NS3-serine protease inhibitors of hepatitis C virus
WO2002048172A2 (fr) 2000-12-12 2002-06-20 Schering Corporation Peptides diaryliques utilises comme inhibiteurs de ns3-serine protease du virus de l'hepatite c
WO2002048157A2 (fr) 2000-12-13 2002-06-20 Bristol-Myers Squibb Pharma Company Imidazolidinones et leurs derives associes, utiles en tant qu'inhibiteurs des proteases ns3 du virus de l'hepatite c
US6727366B2 (en) 2000-12-13 2004-04-27 Bristol-Myers Squibb Pharma Company Imidazolidinones and their related derivatives as hepatitis C virus NS3 protease inhibitors
US6653295B2 (en) 2000-12-13 2003-11-25 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus NS3 protease
WO2002048116A2 (fr) 2000-12-13 2002-06-20 Bristol-Myers Squibb Pharma Company Inhibiteurs de la protease ns3 du virus de l'hepatite c
WO2002048165A2 (fr) 2000-12-15 2002-06-20 Pharmasset Ltd. Agents antiviraux utilises dans le traitement des infections par les flaviviridae
WO2002057425A2 (fr) 2001-01-22 2002-07-25 Merck & Co., Inc. Derives de nucleoside comme inhibiteurs de l'arn polymerase virale arn-dependante
US7202224B2 (en) 2001-01-22 2007-04-10 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
US7125855B2 (en) 2001-01-22 2006-10-24 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
US7105499B2 (en) 2001-01-22 2006-09-12 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
WO2002057287A2 (fr) 2001-01-22 2002-07-25 Merck & Co., Inc. Derives de nucleoside servant d'inhibiteurs de l'arn polymerase virale arn dependante
US6777395B2 (en) 2001-01-22 2004-08-17 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase of hepatitis C virus
US6927291B2 (en) 2001-03-01 2005-08-09 Pharmasset, Ltd. Method for the synthesis of 2′,3′-dideoxy-2′,3′-didehydronucleosides
US6660721B2 (en) 2001-05-23 2003-12-09 Hoffmann-La Roche Inc. Anti-HCV nucleoside derivatives
US6784166B2 (en) 2001-06-12 2004-08-31 Syntex (U.S.A.) Llc 4′-substituted nucleoside derivatives as inhibitors of HCV RNA replication.
US20060198855A1 (en) 2001-06-26 2006-09-07 Progenics Pharmaceuticals, Inc. Uses of DC-SIGN and DC-SIGNR for inhibiting hepatitis C virus infection
US6867185B2 (en) 2001-12-20 2005-03-15 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus
WO2003053349A2 (fr) 2001-12-20 2003-07-03 Bristol-Myers Squibb Company Inhibiteurs de virus de l'hepatite c
US7091184B2 (en) 2002-02-01 2006-08-15 Boehringer Ingelheim International Gmbh Hepatitis C inhibitor tri-peptides
WO2003064456A1 (fr) 2002-02-01 2003-08-07 Boehringer Ingelheim International Gmbh Tripeptides comprenant un hydroxyproline ether d'une quinoline substituee destines a inhiber ns3 (hepatite c)
WO2003064416A1 (fr) 2002-02-01 2003-08-07 Boehringer Ingelheim International Gmbh Tripeptides heterocycliques utiles en tant qu'inhibiteurs de l'hepatite c
US6642204B2 (en) 2002-02-01 2003-11-04 Boehringer Ingelheim International Gmbh Hepatitis C inhibitor tri-peptides
WO2003070750A2 (fr) 2002-02-20 2003-08-28 Sirna Therapeutics, Inc Inhibition de l'expression du gene du virus de l'hepatite c (vhc) induite par l'interference d'arn au moyen d'acide nucleique interferant court (sina)
US20040209831A1 (en) 2002-02-20 2004-10-21 Mcswiggen James RNA interference mediated inhibition of hepatitis C virus (HCV) gene expression using short interfering nucleic acid (siNA)
WO2004043339A2 (fr) 2002-05-20 2004-05-27 Bristol-Myers Squibb Company Inhibiteurs du virus de l'hepatite c a base de cycloalkyle p1' substitue
WO2003099316A1 (fr) 2002-05-20 2003-12-04 Bristol-Myers Squibb Company Sulfamides heterocycliques en tant qu'inhibiteurs du virus de l'hepatite c
US6878722B2 (en) 2002-05-20 2005-04-12 Bristol-Myers Squibb Company Substituted cycloalkyl P1′ hepatitis C virus inhibitors
WO2004032827A2 (fr) 2002-05-20 2004-04-22 Bristol-Myers Squibb Company Inhibiteurs du virus de l'hepatite c
US7041698B2 (en) 2002-05-20 2006-05-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US6869964B2 (en) 2002-05-20 2005-03-22 Bristol-Myers Squibb Company Heterocyclicsulfonamide hepatitis C virus inhibitors
US6995174B2 (en) 2002-05-20 2006-02-07 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2003099274A1 (fr) 2002-05-20 2003-12-04 Bristol-Myers Squibb Company Inhibiteurs du virus de l'hepatite c
WO2004002422A2 (fr) 2002-06-28 2004-01-08 Idenix (Cayman) Limited Ester 3'-l-valine de ?-d-2'-c-methyl-ribofuranosyl cytidine pour le traitement d'infections par des flaviviridae
WO2004002999A2 (fr) 2002-06-28 2004-01-08 Idenix (Cayman) Limited Promedicaments a nucleosides 2' et 3' destines a traiter les infections aux flavivirus
WO2004003000A2 (fr) 2002-06-28 2004-01-08 Idenix (Cayman) Limited Promedicaments 2' et 3' de nucleoside permettant de traiter des infections par les flaviviridae
US20040121980A1 (en) 2002-11-19 2004-06-24 Roche Palo Alto Llc Antiviral nucleoside derivatives
US6846810B2 (en) 2002-11-19 2005-01-25 Roche Palo Alto Llc Antiviral nucleoside derivatives
US20050153877A1 (en) 2003-02-07 2005-07-14 Zhenwei Miao Macrocyclic hepatitis C serine protease inhibitors
US20050009737A1 (en) 2003-05-30 2005-01-13 Jeremy Clark Modified fluorinated nucleoside analogues
US20050038240A1 (en) 2003-06-19 2005-02-17 Roche Palo Alto Llc Processes for preparing 4'-azido-nucleoside derivatives
WO2005012525A1 (fr) 2003-07-25 2005-02-10 Amgen Inc Petit arn interferant utilise en tant qu'agent antiviral pour l'hepatite c
US7208600B2 (en) 2003-10-10 2007-04-24 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly HCV NS3-NS4A proteases
WO2006000085A1 (fr) 2004-06-28 2006-01-05 Boehringer Ingelheim International Gmbh Analogues peptidiques d'inhibiteurs de l'hepatite c
US20060040890A1 (en) 2004-08-23 2006-02-23 Roche Palo Alto Llc Anti-viral nucleosides
US20060046956A1 (en) 2004-08-27 2006-03-02 Schering Corporation Acylsulfonamide compounds as inhibitors of hepatitis C virus NS3 serine protease
WO2007001406A2 (fr) 2004-10-05 2007-01-04 Chiron Corporation Composes macrocycliques contenant un aryle
WO2006119061A2 (fr) 2005-05-02 2006-11-09 Merck & Co., Inc. Inhibiteurs de la protease ns3 du vhc
WO2008121634A2 (fr) * 2007-03-30 2008-10-09 Pharmasset, Inc. Promédicaments de phosphoramidate de nucléoside
WO2010075554A1 (fr) * 2008-12-23 2010-07-01 Pharmasset, Inc. Synthèse de nucléosides de type purine
WO2012088155A1 (fr) * 2010-12-22 2012-06-28 Alios Biopharma, Inc. Analogues de nucléotides cycliques
WO2012142523A2 (fr) * 2011-04-13 2012-10-18 Gilead Sciences, Inc. Analogues de n-nucléoside de pyrimidine 1'-substitués pour un traitement antiviral
WO2013092481A1 (fr) * 2011-12-20 2013-06-27 F. Hoffmann-La Roche Ag Dérivés de nucléoside substitué 2',4'-difluoro-2'-méthyle en tant qu'inhibiteurs de la réplication de l'arn du vhc (virus de l'hépatite c)
WO2013177219A1 (fr) * 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. Composés d'acide d-aminé contre les maladies hépatiques
WO2014078427A1 (fr) * 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. Ester de d-alanine d'analogue de rp-nucléoside
WO2014078436A1 (fr) * 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. Ester de d-alanine d'analogue de sp-nucléoside
WO2014148949A1 (fr) * 2013-03-22 2014-09-25 Асави, Ллс Alkyl 2-{[(2r,3s,5r)-5-(4-amino-2-oxo-2n-pyrimidin-1-yl)-3-hydroxy-tétrahydro-furan-2-ilméthoxy]-phénoxy-phosphorilamino}-propionates, inhibiteurs de nucléosides d'arn-polymérase hcv ns5b, procédés de fabrication et d'utilisation

Non-Patent Citations (38)

* Cited by examiner, † Cited by third party
Title
"Chronic Hepatitis", THE MERCK MANUAL ONLINE, - February 2014 (2014-02-01), Retrieved from the Internet <URL:www.merckmanuals.com/professional/hepatic_and biliary_disorders/hepatitis/ chronic - hepatitis.html>
"Hepatitis C Fact Sheet", WORLD HEALTH ORGANIZATION FACT SHEET NO. 164, April 2014 (2014-04-01)
"Physician's Desk Reference (PDR) 57th Ed.", 2003, MEDICAL ECONOMICS CO., INC.
"Remington's Pharmaceutical Sciences, 16th, 18th and 20th eds", 1980, MACK PUBLISHING
"Remington's Pharmaceutical Sciences, 20th ed.", 2000, MACK PUBLISHING
ALT M. ET AL., ARCHIVES OF VIROLOGY, vol. 142, 1997, pages 589 - 599
ALT M. ET AL., HEPATOLOGY, vol. 22, 1995, pages 707 - 717
ATTWOOD ET AL., ANTIVIRAL CHEMISTRY AND CHEMOTHERAPY, vol. 10, 1999, pages 259 - 273
BOYER, N. ET AL., J. HEPATOL., vol. 32, 2000, pages 98 - 112
BUCHWALD ET AL., SURGERY, vol. 88, 1980, pages 507
CHU M. ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, vol. 9, pages 1949 - 1952
CHU M. ET AL., TETRAHEDRON LETTERS, vol. 37, 1996, pages 7229 - 7232
DI BESCEGLIE; A. M.; BACON, B. R., SCIENTIFIC AMERICAN, October 1999 (1999-10-01), pages 80 - 85
E.W. MARTIN: "Remington's Pharmaceutical Sciences"
FERRARI R. ET AL., JOURNAL OF VIROLOP, vol. 73, 1999, pages 1649 - 1654
FOSTER ET AL., ADV. DRUG RES., vol. 14, 1985, pages 1 - 36
GALDERISI U. ET AL., JOURNAL OF CELLULAR PHYSIOLOGY, vol. 181, 1999, pages 251 - 257
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115 - 138
GREENE ET AL.: "Protective Groups in Organic Synthesis, 2nd ed.", 1991, JOHN WILEY AND SONS
HARDMAN ET AL.: "Goodman & Gilman's The Pharmacological Basis Of Basis Of Therapeutics, 9th ed.", 1996, MC-GRAW-HILL
JENS T. CARSTENSEN: "Drug Stability: Principles & Practice, 2nd ed.", 1995, MARCEL DEKKER, pages: 379 - 380
KAKIUCHI N. ET AL., J. EBS LETTERS, vol. 421, pages 217 - 220
KNIPE, D. M., AND HOWLEY, P. M.: "Fields Virology, 5th ed.", 2006, LIPPINCOTT WILLIAMS & WILKINS PUBLISHERS, article "Chapters 33-35"
KNIPE, D. M., AND HOWLEY, P. M.: "Fields Virology, 6th ed.", 2013, LIPPINCOTT WILLIAMS & WILKINS PUBLISHERS, article "Chapters 25-27"
KUSHNER ET AL., CAN. J. PHYSIOL. PHARMACOL., vol. 77, 1999, pages 79 - 88
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LEA & FEBIGER: "Introduction to Pharmaceutical Dosage Forms, 4th ed.", 1985, LEA & FEBIGER
LESENS ET AL., J1U/ECT DIS, vol. 179, 1999, pages 1254 - 1258
LOHMANN V. ET AL., VIROLOGY, vol. 249, 1998, pages 108 - 118
MACCJAK, D. J. ET AL., HEPATOLOGY, vol. 30, 1999
MAIER; WU, WORLD J CASTRØEMERØ/, vol. 8, 2002, pages 577 - 57
QASIM M.A. ET AL., BIOCHEMISTRY, vol. 36, 1997, pages 1598 - 1607
SAUDEK ET AL., N. ENGL. J. MED., vol. 321, 1989, pages 574
SEFTON, CRC CRIT. REF. BIOMED. ENG., vol. 14, 1987, pages 201
SUDO K. ET AL., ANTIVIRAL CHEMISTRY AND CHEMOTHERAPY, vol. 9, 1998, pages 186
SUDO K. ET AL., ANTIVIRAL RESEARCH, vol. 32, 1996, pages 9 - 18
SUDO K. ET AL., BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 238, 1997, pages 643 - 647
TAKESHITA N. ET AL., ANALYTICAL BIOCHEMISTRY, vol. 247, 1997, pages 242 - 246

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10717758B2 (en) 2012-05-22 2020-07-21 Idenix Pharmaceuticals Llc D-amino acid compounds for liver disease
US10301347B2 (en) 2012-05-25 2019-05-28 Janssen Sciences Ireland Unlimited Company Uracyl spirooxetane nucleosides
US9422323B2 (en) 2012-05-25 2016-08-23 Janssen Sciences Ireland Uc Uracyl spirooxetane nucleosides
US10774106B2 (en) 2012-05-25 2020-09-15 Janssen Sciences Ireland Unlimited Company Uracyl spirooxetane nucleosides
US10040814B2 (en) 2012-05-25 2018-08-07 Janssen Sciences Ireland Uc Uracyl spirooxetane nucleosides
US9845336B2 (en) 2012-05-25 2017-12-19 Janssen Sciences Ireland Uc Uracyl spirooxetane nucleosides
US10544184B2 (en) 2012-05-25 2020-01-28 Janssen Sciences Ireland Unlimited Company Uracyl spirooxetane nucleosides
US10513534B2 (en) 2012-10-08 2019-12-24 Idenix Pharmaceuticals Llc 2′-chloro nucleoside analogs for HCV infection
US10231986B2 (en) 2013-03-13 2019-03-19 Idenix Pharmaceuticals Llc Amino acid phosphoramidate pronucleotides of 2′-cyano, azido and amino nucleosides for the treatment of HCV
US10238680B2 (en) 2013-08-01 2019-03-26 Idenix Pharmaceuticals Llc D-amino acid phosphoramidate pronucleotides of halogeno pyrimidine compounds for liver disease
US10202411B2 (en) 2014-04-16 2019-02-12 Idenix Pharmaceuticals Llc 3′-substituted methyl or alkynyl nucleosides nucleotides for the treatment of HCV
WO2018013937A1 (fr) 2016-07-14 2018-01-18 Atea Pharmaceuticals, Inc. Nucléotides de purine beta-d-2'-deoxy-2'-alpha-fluoro-2'-beta-c-substitué-4'-fluoro-n6-substitué-6-amino-2-substitué pour le traitement de l'infection par le virus de l'hépatite c
WO2018048937A1 (fr) 2016-09-07 2018-03-15 Atea Pharmaceuticals, Inc. Nucléotides de purine substitués en position 2'-n 6 pour le traitement du virus à arn
US10946033B2 (en) 2016-09-07 2021-03-16 Atea Pharmaceuticals, Inc. 2′-substituted-N6-substituted purine nucleotides for RNA virus treatment
EP3865136A1 (fr) 2016-09-07 2021-08-18 ATEA Pharmaceuticals, Inc. Nucléotides de purine substitués en position 2'-n 6 pour le traitement du virus corona
EP4088725A1 (fr) 2016-09-07 2022-11-16 ATEA Pharmaceuticals, Inc. Nucléotides de purine substitués en position 2'-n6 pour le traitement du picornaviridae

Similar Documents

Publication Publication Date Title
US9339541B2 (en) Thiophosphate nucleosides for the treatment of HCV
US9192621B2 (en) Esters and malonates of SATE prodrugs
US9211300B2 (en) 4′-fluoro nucleosides for the treatment of HCV
EP2981542B1 (fr) 2&#39;,4&#39;-fluoronucléosides pour le traitement du vhc
EP2970358B1 (fr) 3&#39;-désoxynucléosides utilisables en vue du traitement d&#39;une infection par le vhc
EP3004130B1 (fr) 1&#39;,4&#39;-thio nucléosides pour le traitement du virus de l&#39;hépatite c (vhc)
EP2909223B1 (fr) Composés dinucléotides contre une infection par le vhc
EP3027636B1 (fr) Phosphoramidate de d-acides aminés pronucleotidiques dérivés de pyrimidine halogenée pour des maladies du foie
WO2014160484A1 (fr) Pronucléotides de phosphoramidate d&#39;acide aminé de 2&#39;-cyano, azido et amino nucléosides pour le traitement du virus de l&#39;hépatite c (vhc)
EP3074399A1 (fr) Analogues de nucléoside 2&#39;-dichloro et 2&#39;-fluoro-2&#39;-chloro analogues pour l&#39;infection par le vhc
EP2920195A1 (fr) Ester de d-alanine d&#39;analogue de rp-nucléoside
WO2014078436A1 (fr) Ester de d-alanine d&#39;analogue de sp-nucléoside
WO2013177188A1 (fr) Promédicaments de 3&#39;,5&#39;-phosphoramidate cyclique pour traiter une infection par le virus de l&#39;hépatite c
WO2014066239A1 (fr) Nucléosides 2&#39;, 4&#39;-pontés pour l&#39;infection par le vhc
WO2015077360A2 (fr) Analogues nucléosidiques renfermant un cyclopentane ou un cyclopentène pour le traitement de l&#39;hépatite c
WO2015061683A1 (fr) Pronucléotides thiophosphoramidates à acide aminé d et pronucléotides thiophosphoramidates à d-analine de composés de nucléoside utiles pour le traitement du vhc
WO2015095419A1 (fr) Nucléosides 4&#39;-or pour le traitement du vhc

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14799922

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14799922

Country of ref document: EP

Kind code of ref document: A1