US20180235993A1 - Methods and compositions for treating flaviviruses and pestiviruses - Google Patents

Methods and compositions for treating flaviviruses and pestiviruses Download PDF

Info

Publication number
US20180235993A1
US20180235993A1 US15/952,117 US201815952117A US2018235993A1 US 20180235993 A1 US20180235993 A1 US 20180235993A1 US 201815952117 A US201815952117 A US 201815952117A US 2018235993 A1 US2018235993 A1 US 2018235993A1
Authority
US
United States
Prior art keywords
alkyl
purine
acyl
phosphate
monophosphate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/952,117
Inventor
Jean-Pierre Sommadossi
Paolo La Colla
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universita degli Studi di Cagliari
Idenix Pharmaceuticals LLC
Original Assignee
Universita degli Studi di Cagliari
Idenix Pharmaceuticals LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26902464&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20180235993(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Universita degli Studi di Cagliari, Idenix Pharmaceuticals LLC filed Critical Universita degli Studi di Cagliari
Priority to US15/952,117 priority Critical patent/US20180235993A1/en
Publication of US20180235993A1 publication Critical patent/US20180235993A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/7056Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing five-membered rings with nitrogen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • C07H19/073Pyrimidine radicals with 2-deoxyribosyl as the saccharide radical
    • 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
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • 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
    • 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
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • 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
    • C07H19/23Heterocyclic radicals containing two or more heterocyclic rings condensed among themselves or condensed with a common carbocyclic ring system, not provided for in groups C07H19/14 - C07H19/22
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention is in the area of pharmaceutical chemistry, and in particular, is a compound, method and composition for the treatment of flaviviruses and pestiviruses.
  • Pestiviruses and flaviviruses belong to the Flaviviridae family of viruses along with hepatitis C virus.
  • the pestivirus genus includes bovine viral diarrhea virus (BVDV), classical swine fever virus (CSFV, also called hog cholera virus) and border disease virus (BDV) of sheep (Moennig, V. et al. Adv. Vir. Res. 1992, 41, 53-98).
  • Pestivirus infections of domesticated livestock (cattle, pigs and sheep) cause significant economic losses worldwide.
  • BVDV causes mucosal disease in cattle and is of significant economic importance to the livestock industry (Meyers, G. and Thiel, H.-J., Advances in Virus Research, 1996, 47, 53-118; Moennig V., et al, Adv. Vir. Res. 1992, 41, 53-98).
  • Pestivirus infections in man have been implicated in several diseases including congenital brain injury, infantile gastroenteritis and chronic diarrhea in human immunodeficiency virus (HIV) positive patients.
  • HAV human immunodeficiency virus
  • the flavivirus genus includes more than 68 members separated into groups on the basis of serological relatedness (Calisher et al., J. Gen. Virol, 1993, 70, 37-43). Clinical symptoms vary and include fever, encephalitis and hemorrhagic fever. Fields Virology , Editors: Fields, B. N., Knipe, D. M., and Howley, P. M., Lippincott-Raven Publishers, Philadelphia, Pa., 1996, Chapter 31, 931-959. Flaviviruses of global concern that are associated with human disease include the dengue hemorrhagic fever viruses (DHF), yellow fever virus, shock syndrome and Japanese encephalitis virus. Halstead, S. B., Rev. Infect. Dis., 1984, 6, 251-264; Halstead, S. B., Science, 239:476-481, 1988 ; Monath, T. P., New Eng. J. Med., 1988, 319, 641-643.
  • DHF dengue hemorrhagic fever viruses
  • antiviral agents that have been identified as active against the flavivirus or pestiviruses include:
  • a compound of Formula I or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • R 1 , R 2 and R 3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 and X 2 are independently H or
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; and Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 and
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; and Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 and
  • a compound of Formula IV or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • R 1 , R 2 and R 3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 is selected from the group consisting of
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; and Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 is selected from
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; and Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 is selected from
  • a compound selected from Formulas VII, VIII and IX, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano,
  • a compound of Formulas X, XI and XII, or a pharmaceutically acceptable salt or prodrug thereof is provided:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano,
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano,
  • the invention provides a compound of Formula XVI, or a pharmaceutically acceptable salt or prodrug thereof:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 and R 2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 or R 2 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkyn
  • the invention provides a compound of Formula XVII, or a pharmaceutically acceptable salt or prodrug thereof:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 and R 2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 or R 2 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkyn
  • the invention provides a compound of Formula XVIII, or a pharmaceutically acceptable salt or prodrug thereof:
  • Base is a purine or pyrimidine base as defined herein; R 1 and R 2 independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl);
  • sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 or R 2 is independently H or phosphate;
  • R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO 2 , NH 2 , —NH(lower alkyl), —NH(acyl), —N(lower alkyl) 2 , —N(acyl) 2 ;
  • R 7 and R 9 are independently hydrogen, OR 2 , alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO 2 , amino, loweralkylamino or di(lower-
  • the ⁇ -D- and ⁇ -L-nucleosides of this invention may inhibit flavivirus or pestivirus polymerase activity. These nucleosides can be assessed for their ability to inhibit flavivirus or pestivirus polymerase activity in vitro according to standard screening methods.
  • the efficacy of the anti-flavivirus or pestivirus compound is measured according to the concentration of compound necessary to reduce the plaque number of the virus in vitro, according to methods set forth more particularly herein, by 50% (i.e. the compound's EC 50 ).
  • the compound exhibits an EC 50 of less than 15 or preferably, less than 10 micromolar in vitro.
  • the active compound can be administered in combination or alternation with another anti-flavivirus or pestivirus agent.
  • combination therapy effective dosages of two or more agents are administered together, whereas during alternation therapy an effective dosage of each agent is administered serially.
  • the dosages 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.
  • HCV is a member of the Flaviviridae family; however, now, HCV has been placed in a new monotypic genus, hepacivirus. Therefore, in one embodiment, the flavivirus or pestivirus is not HCV.
  • antiviral agents that can be used in combination with the compounds disclosed herein include:
  • Inhibitors of serine proteases particularly hepatitis C virus NS3 protease, PCT WO 98/17679), including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an electrophile such as a boronic acid or phosphonate.
  • Llinas-Brunet et al Hepatitis C inhibitor peptide analogues, PCT WO 99/07734.
  • Non-substrate-based inhibitors such as 2,4,6-trihydroxy-3-nitro-benzamide derivatives(Sudo K. et al., Biochemical and Biophysical Research Communications, 238:643-647, 1997; Sudo K. et al. Antiviral Chemistry and Chemotherapy 9:186, 1998), including RD3-4082 and RD3-4078, the former substituted on the amide with a 14 carbon chain and the latter processing a para-phenoxyphenyl group;
  • Helicase inhibitors (Diana G. D. et al., Compounds, compositions and methods for treatment of hepatitis C, U.S. Pat. No. 5,633,358; Diana G. D. et al., Piperidine derivatives, pharmaceutical compositions thereof and their use in the treatment of hepatitis C, PCT WO 97/36554);
  • 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 22:707-717, 1995), or nucleotides 326-348 comprising the 3′ end of the NCR and nucleotides 371-388 located in the core coding region of the IICV RNA (Alt M. et al., Archives of Virology 142:589-599, 1997; Galderisi U. et al., Journal of Cellular Physiology 181:251-257, 1999);
  • Inhibitors of IRES-dependent translation (Ikeda N et al., Agent for the prevention and treatment of hepatitis C, Japanese Patent Publication JP-08268890; Kai Y. et al. Prevention and treatment of viral diseases, Japanese Patent Publication JP-10101591);
  • miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S. Pat. No. 6,034,134 to Gold et al.), alkyl lipids (U.S. Pat. No. 5,922,757 to Chojkier et al.), vitamin E and other antioxidants (U.S. Pat. No. 5,922,757 to Chojkier et al.), squalene, amantadine, bile acids (U.S. Pat. No. 5,846,964 to Ozeki et al.), N-(phosphonoacetyl)-L-aspartic acid, (U.S. Pat. No.
  • FIG. 1 provides the structure of various non-limiting examples of nucleosides of the present invention, as well as other known nucleosides, FIAU and Ribavirin, which are used as comparative examples in the text.
  • FIG. 2 is a line graph of the pharmacokinetics (plasma concentrations) of ⁇ -D-2′-CH 3 -riboG administered to Cynomolgus Monkeys over time after administration.
  • FIGS. 3 a and 3 b are line graphs of the pharmacokinetics (plasma concentrations) of ⁇ -D-2′-CH 3 -riboG administered to Cynomolgus Monkeys either intravenously (3a) or orally (3b) over time after administration.
  • FIG. 4 depicts line graphs of the results of the cell protection assay of ⁇ -D-2′-CH 3 -riboG against BVDV.
  • FIG. 5 depicts line graphs of the results of the cell protection assay of ribavirin against BVDV.
  • FIG. 6 are line graphs of the cell protection assay of ⁇ -D-2′-CH 3 -riboG, ⁇ -D-2′-CH 3 -riboC, ⁇ -D-2′-CH 3 -riboU, ⁇ -D-2′-CH 3 -riboA and ribavirin.
  • FIG. 7 are line graphs of the results of the plaque reduction assay for ⁇ -D-2′-CH 3 -riboU, ⁇ -D-2′-CH 3 -riboC and ⁇ -D-2′-CH 3 -riboG.
  • FIG. 8 is an illustration of plaque reduction based on increasing concentrations of ⁇ -D-2′-CH 3 -riboU.
  • FIG. 9 is a line graph of the results of the yield reduction assay for ⁇ -D-2′-CH 3 -riboG, depicting a 4 log reduction at 9 ⁇ M.
  • FIG. 10 is an illustration of the yield reduction based on increasing concentrations of ⁇ -D-2′-CH 3 -riboC.
  • the invention as disclosed herein is a compound, method and composition for the treatment of pestiviruses and flaviviruses in humans and other host animals, that includes the administration of an effective flavivirus or pestivirus treatment amount of an ⁇ -D- or ⁇ -L-nucleoside as described herein or a pharmaceutically acceptable salt or prodrug thereof, optionally in a pharmaceutically acceptable carrier.
  • the compounds of this invention either possess antiviral (i.e., anti-flavivirus or pestivirus) activity, or are metabolized to a compound that exhibits such activity.
  • the present invention includes the following features:
  • Flaviviruses included within the scope of this invention are discussed generally in Fields Virology , Editors: Fields, B. N., Knipe, D. M., and Howley, P. M., Lippincott-Raven Publishers, Philadelphia, Pa., Chapter 31, 1996.
  • flaviviruses include, without limitation: Absettarov, Alfuy, AIN, Aroa, Bagaza, Banzi, Bouboui, Bussuquara, Cacipacore, Carey Island, 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, Karshi, Kedougou, Kokobera, Koutango, Kumlinge, Kunjin, Kyasanur Forest disease, Langat, Louping ill, Meaban, Modoc, Montana myotis leukoencephalitis, Murray valley encephalitis, Naranjal, Negishi, Ntaya, Omsk hemorrhagic fever, Phnom-Penh bat, Powassan, Rio Bravo, Rocio, Royal Farm, Russian spring-summer encephalitis, Saboya
  • Pestiviruses included within the scope of this invention are discussed generally in Fields Virology , Editors: Fields, B. N., Knipe, D. M., and Howley, P. M., Lippincott-Raven Publishers, Philadelphia, Pa., Chapter 33, 1996.
  • Specific pestiviruses 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
  • a compound of Formula I or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • R 1 , R 2 and R 3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 and X 2 are independently H or
  • R 1 , R 2 and R 3 are independently H or phosphate (preferably H);
  • X 1 is H
  • X 2 is H or NH 2 ; and Y is hydrogen, bromo, chloro, fluoro, iodo, NH 2 or OH.
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; and Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 and
  • R 1 , R 2 and R 3 are independently H or phosphate (preferably H);
  • X 1 is H
  • X 2 is H or NH 2 ; and Y is hydrogen, bromo, chloro, fluoro, iodo, NH 2 or OH.
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; and Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 and
  • R 1 , R 2 and R 3 are independently H or phosphate (preferably H);
  • X 1 is H
  • X 2 is H or NH 2 ; and Y is hydrogen, bromo, chloro, fluoro, iodo, NH 2 or OH.
  • a compound of Formula IV or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • R 1 , R 2 and R 3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 is selected from the group consisting of
  • R 1 , R 2 and R 3 are independently H or phosphate (preferably H); X 1 is H or CH 3 ; and Y is hydrogen, bromo, chloro, fluoro, iodo, NH 2 or OH.
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; and Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ; X 1 is selected from
  • R 1 , R 2 and R 3 are independently H or phosphate (preferably H); X 1 is H or CH 3 ; and Y is hydrogen, bromo, chloro, fluoro, iodo, NH 2 or OH.
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; and
  • Y is hydrogen, bromo, chloro, fluoro, iodo, OR 4 , NR 4 R 5 or SR 4 ;
  • X 1 is selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR 4 , NR 4 NR 5 or SR 5 ; and R 4 and R 5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • R 1 , R 2 and R 3 are independently H or phosphate (preferably H); X 1 is H or CH 3 ; and Y is hydrogen, bromo, chloro, fluoro, iodo, NH 2 or OH.
  • a compound selected from Formulas VII, VIII and IX, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano,
  • Base is a purine or pyrimidine base as defined herein; R 1 , R 2 and R 3 are independently hydrogen or phosphate; R 6 is alkyl; and X is O, S, SO 2 or CH 2 .
  • Base is a purine or pyrimidine base as defined herein; R 1 , R 2 and R 3 are hydrogens; R 6 is alkyl; and X is O, S, SO 2 or CH 2 .
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently hydrogen or phosphate;
  • R 6 is alkyl;
  • X is O.
  • a compound of Formula X, XI or XII, or a pharmaceutically acceptable salt or prodrug thereof is provided:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano,
  • Base is a purine or pyrimidine base as defined herein; R 1 , R 2 and R 3 are independently hydrogen or phosphate; R 6 is alkyl; and X is O, S, SO 2 or CH 2 .
  • Base is a purine or pyrimidine base as defined herein; R 1 , R 2 and R 3 are hydrogens; R 6 is alkyl; and X is O, S, SO 2 or CH 2 .
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently H or phosphate;
  • R 6 is alkyl;
  • X is O.
  • Base is a purine or pyrimidine base as defined herein; optionally substituted with an amine or cyclopropyl (e.g., 2-amino, 2,6-diamino or cyclopropyl guanosine); and R 1 and R 2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 or R 3 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano,
  • Base is a purine or pyrimidine base as defined herein; R 1 , R 2 and R 3 are independently hydrogen or phosphate; R 6 is alkyl; and X is O, S, SO 2 or CH 2 .
  • Base is a purine or pyrimidine base as defined herein; R 1 , R 2 and R 3 are hydrogens; R 6 is alkyl; and X is O, S, SO 2 or CH 2 .
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 , R 2 and R 3 are independently hydrogen or phosphate;
  • R 6 is alkyl;
  • X is O.
  • the invention provides a compound of Formula XVI, or a pharmaceutically acceptable salt or prodrug thereof:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 and R 2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 and R 2 are independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkyn
  • a compound of Formula XVI is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R′ is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R′ is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R′ is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R′ is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R′ is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6 is independently H or phosphate;
  • a compound of Formula XVI is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 1 is independently H or phosphate; (3) R 1 is independently H or phosphate; (3) R 1 is independently
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO 2 , amino, loweralkylamino or di(loweralkyl)amino; (4) R 7 and R 9 are independently OR 2 ; (5) R 8 and R 10 are hydrogen; and (6) X is O, S, SO 2 or CH 2 .
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl; (4) R 7 and R 9 are independently OR 2 ; (5) R 8 and R 10 are hydrogen; and (6) X is O, S, SO 2 , or CH 2 .
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl; (4) R 7 and R 9 are independently OR 2 ; (5) R 8 and R 10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O.
  • a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl; (4) R 7 and R 9 are independently OR 2 , alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO 2 , amino, loweralkylamino or di(loweralkyl)amino; (5) R 8 and R 10 are hydrogen; and (6) X is O.
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is guanine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is cytosine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl;
  • R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is thymine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is uracil; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is phosphate; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is ethyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is propyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is butyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 is hydrogen and R 9 is hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is S;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is SO 2 ;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 and R 10 are hydrogen; and (6) X is CH 2 ;
  • the invention provides a compound of Formula XVII, or a pharmaceutically acceptable salt or prodrug thereof:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 is H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-viny
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl; (4) R 7 and R 9 are independently hydrogen, OR 2 , alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO 2 , amino, loweralkylamino or di(loweralkyl)-amino; (5) R 10 is H, alkyl (including lower alkyl), chlorine, bromine or iodine; and (6) X is O, S, SO 2 , or CH 2 .
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO 2 , amino, loweralkylamino, or di(loweralkyl)amino; (4) R 7 and R 9 are independently OR 2 ; (5) R 10 is H; and (6) X is O, S, SO 2 , or CH 2 .
  • a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl; (4) R 7 and R 9 are independently OR 2 ; (5) R 10 is H; and (6) X is O, S, SO 2 , or CH 2 .
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is guanine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is cytosine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is thymine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is uracil; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is phosphate; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is ethyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is propyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is butyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is S;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 10 is hydrogen; and (6) X is SO 2 ; or
  • the invention provides a compound of Formula XVIII, or a pharmaceutically acceptable salt or prodrug thereof:
  • Base is a purine or pyrimidine base as defined herein;
  • R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; R 6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vin
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 is independently H or phosphate; (3) R 6
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO 2 , amino, loweralkylamino or di(loweralkyl)amino; (4) R 7 and R 9 are independently OR 2 ; (5) R 8 is H; and (6) X is O, S, SO 2 or CH 2 .
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl; (4) R 7 and R 9 are independently OR 2 ; (5) R 8 is H; and (6) X is O, S, SO 2 , or CH 2 .
  • a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R 1 is independently H or phosphate; (3) R 6 is alkyl; (4) R 7 and R 9 are independently OR 2 ; (5) R 8 is H; and (6) X is O.
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is O;
  • Base is guanine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is O;
  • Base is cytosine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is O;
  • Base is thymine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is O;
  • Base is uracil; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5)
  • R 8 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is phosphate; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is ethyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is propyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is butyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is O;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is S;
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is SO 2 ; or
  • Base is adenine; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R 8 is hydrogen; and (6) X is CH 2 .
  • the ⁇ -D- and ⁇ -L-nucleosides of this invention belong to a class of anti-flavivirus or pestivirus agents that may inhibit flavivirus or pestivirus polymerase activity.
  • Nucleosides can be screened for their ability to inhibit flavivirus or pestivirus polymerase activity in vitro according to screening methods set forth more particularly herein. One can readily determine the spectrum of activity by evaluating the compound in the assays described herein or with another confirmatory assay.
  • the efficacy of the anti-flavivirus or pestivirus compound is measured according to the concentration of compound necessary to reduce the plaque number of the virus in vitro, according to methods set forth more particularly herein, by 50% (i.e. the compound's EC 50 ). In preferred embodiments the compound exhibits an EC 50 of less than 15 or 10 micromolar.
  • HCV is a member of the Flaviviridae family; however, now, HCV has been placed in a new monotypic genus, hepacivirus. Therefore, in one embodiment, the flavivirus or pestivirus is not HCV.
  • the active compound can be administered as any salt or prodrug that upon administration to the recipient is capable of providing directly or indirectly the parent compound, or that exhibits activity itself.
  • Nonlimiting examples are the pharmaceutically acceptable salts (alternatively referred to as “physiologically acceptable salts”), and a compound, which has been alkylated or acylated at the 5′-position, or on the purine or pyrimidine base (a type of “pharmaceutically acceptable prodrug”).
  • physiologically acceptable salts alternatively referred to as “physiologically acceptable salts”
  • the modifications can affect the biological activity of the compound, in some cases increasing the activity over the parent compound. This can easily be assessed by preparing the salt or prodrug and testing its antiviral activity according to the methods described herein, or other methods known to those skilled in the art.
  • alkyl refers to a saturated straight, branched, or cyclic, primary, secondary, or tertiary hydrocarbon of typically C 1 to C 10 , and specifically includes methyl, trifluoromethyl, ethyl, propyl, isopropyl, cyclopropyl, butyl, isobutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 3-methylpentyl, 2,2-dimethylbutyl, and 2,3-dimethylbutyl.
  • the term includes both substituted and unsubstituted alkyl groups.
  • Moieties with which the alkyl group can be substituted are selected from the group consisting of 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, hereby incorporated by reference.
  • lower alkyl refers to a C 1 to C 4 saturated straight, branched, or if appropriate, a cyclic (for example, cyclopropyl) alkyl group, including both substituted and unsubstituted forms. Unless otherwise specifically stated in this application, when alkyl is a suitable moiety, lower alkyl is preferred. Similarly, when alkyl or lower alkyl is a suitable moiety, unsubstituted alkyl or lower alkyl is preferred.
  • alkylamino or arylamino refers to an amino group that has one or two alkyl or aryl substituents, respectively.
  • protected 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.
  • aryl refers to phenyl, biphenyl, or naphthyl, and preferably phenyl.
  • the term includes both substituted and unsubstituted moieties.
  • the aryl group can be substituted with one or more moieties selected from the group consisting of 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
  • alkaryl or alkylaryl refers to an alkyl group with an aryl substituent.
  • aralkyl or arylalkyl refers to an aryl group with an alkyl substituent.
  • halo includes chloro, bromo, iodo, and fluoro.
  • purine or pyrimidine base includes, but is not limited to, adenine, N 6 -alkylpurines, N 6 -acylpurines (wherein acyl is C(O)(alkyl, aryl, alkylaryl, or arylalkyl), N 6 -benzylpurine, N 6 -halopurine, N 6 -vinylpurine, N 6 -acetylenic purine, N 6 -acyl purine, N 6 -hydroxyalkyl purine, N 6 -thioalkyl purine, N 2 -alkylpurines, N 2 -alkyl-6-thiopurines, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6-azapyrimidine, including 6-azacytosine, 2- and/or 4-mercaptopyrmidine, uracil, 5-halouracil, including 5-fluorouracil, C 5 -alkylpyrimidines, C 5 -
  • Purine bases include, but are not limited to, guanine, adenine, hypoxanthine, 2,6-diaminopurine, 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, dimethylhexylsilyl, t-butyldimethylsilyl and t-butyldiphenylsilyl, trityl, alkyl groups, and acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl.
  • the purine or pyrimidine base can optionally substituted such that it forms a viable prodrug, which can be cleaved in vivo.
  • appropriate substituents include acyl moiety, an amine or cyclopropyl (e.g., 2-amino, 2,6-diamino or cyclopropyl guanosine).
  • acyl refers to a carboxylic acid ester in which the non-carbonyl moiety of the ester group is selected from straight, branched, or cyclic alkyl or lower alkyl, alkoxyalkyl including methoxymethyl, aralkyl including benzyl, aryloxyalkyl such as phenoxymethyl, aryl including phenyl optionally substituted with halogen, C 1 to C 4 alkyl or C 1 to C 4 alkoxy, sulfonate esters such as alkyl or aralkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxytrityl, substituted benzyl, trialkylsilyl (e.g.
  • esters dimethyl-t-butylsilyl or diphenylmethylsilyl.
  • Aryl groups in the esters optimally comprise a phenyl group.
  • lower acyl refers to an acyl group in which the non-carbonyl moiety is lower alkyl.
  • the term “substantially free of” or “substantially in the absence of” refers to a nucleoside composition that includes at least 85 or 90% by weight, preferably 95% to 98% by weight, and even more preferably 99% to 100% by weight, of the designated enantiomer of that nucleoside. In a preferred embodiment, in the methods and compounds of this invention, the compounds are substantially free of enantiomers.
  • isolated refers to a nucleoside composition that includes at least 85 or 90% by weight, preferably 95% to 98% by weight, and even more preferably 99% to 100% by weight, of the nucleoside, the remainder comprising other chemical species or enantiomers.
  • both R′′ can be carbon, both R′′ can be nitrogen, or one R′′ can be carbon and the other R′′ nitrogen.
  • host refers to an unicellular or multicellular organism in which the virus can replicate, including cell lines and animals, and preferably a human. Alternatively, the host can be carrying a part of the flavivirus or pestivirus genome, whose replication or function can be altered by the compounds of the present invention.
  • host specifically refers to infected cells, cells transfected with all or part of the flavivirus or pestivirus genome and animals, in particular, primates (including chimpanzees) and humans.
  • the host is a human patient.
  • Veterinary applications in certain indications, however, are clearly anticipated by the present invention (such as chimpanzees).
  • pharmaceutically acceptable salt or prodrug is used throughout the specification to describe any pharmaceutically acceptable form (such as an ester, phosphate ester, salt of an ester or a related group) of a nucleoside compound which, upon administration to a patient, provides the nucleoside compound.
  • Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable inorganic or organic bases and acids. Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium and magnesium, among numerous other acids well known in the pharmaceutical art.
  • Pharmaceutically acceptable prodrugs refer to a compound that is metabolized, for example hydrolyzed or oxidized, in the host to form the compound of the present invention.
  • prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound.
  • Prodrugs include compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, dephosphorylated to produce the active compound.
  • the compounds of this invention possess antiviral activity against flavivirus or pestivirus, or are metabolized to a compound that exhibits such activity.
  • pharmaceutically acceptable salts are organic acid addition salts formed with acids, which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, ⁇ -ketoglutarate, and ⁇ -glycerophosphate.
  • Suitable inorganic salts may also be formed, including, sulfate, nitrate, bicarbonate, and carbonate salts.
  • salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine
  • a suitable acid affording a physiologically acceptable anion.
  • Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
  • nucleosides described herein can be administered as a nucleotide prodrug to increase the activity, bioavailability, stability or otherwise alter the properties of the nucleoside.
  • a number of nucleotide prodrug ligands are known.
  • alkylation, acylation or other lipophilic modification of the mono, di or triphosphate of the nucleoside will increase the stability of the nucleotide.
  • substituent groups that can replace one or more hydrogens on the phosphate moiety are alkyl, aryl, steroids, carbohydrates, including sugars, 1,2-diacylglycerol and alcohols. Many are described in R. Jones and N. Bischofberger, Antiviral Research, 27 (1995) 1-17. Any of these can be used in combination with the disclosed nucleosides to achieve a desired effect.
  • the active nucleoside can also be provided as a 5′-phosphoether lipid or a 5′-ether lipid, as disclosed in the following references, which are incorporated by reference herein: Kucera, L. S., N. Iyer, E. Leake, A. Raben, Modest E. K., D. L. W., and C. Piantadosi, “Novel membrane-interactive ether lipid analogs that inhibit infectious HIV-1 production and induce defective virus formation,” AIDS Res. Hum. Retro Viruses, 1990, 6, 491-501; Piantadosi, C., J. Marasco C. J., S. L. Morris-Natschke, K. L. Meyer, F. Gumus, J. R. Surles, K. S.
  • Nonlimiting examples of U.S. patents that disclose suitable lipophilic substituents that can be covalently incorporated into the nucleoside, preferably at the 5′-OH position of the nucleoside or lipophilic preparations include U.S. Pat. No. 5,149,794 (Sep. 22, 1992, Yatvin et al.); U.S. Pat. No. 5,194,654 (Mar. 16, 1993, Hostetler et al., U.S. Pat. No. 5,223,263 (Jun. 29, 1993, Hostetler et al.); U.S. Pat. No. 5,256,641 (Oct. 26, 1993, Yatvin et al.); U.S. Pat. No.
  • Drug resistance most typically occurs by mutation of a gene that encodes for an enzyme used in viral replication.
  • the efficacy of a drug against flavivirus or pestivirus infection can be prolonged, augmented, or restored by administering the compound in combination or alternation with a second, and perhaps third, antiviral compound that induces a different mutation from that caused by the principle drug.
  • the pharmacokinetics, biodistribution or other parameter of the drug can be altered by such combination or alternation therapy.
  • combination therapy is typically preferred over alternation therapy because it induces multiple simultaneous stresses on the virus.
  • antiviral agents that can be used in combination or alternation with the compounds disclosed herein include:
  • Inhibitors of serine proteases particularly hepatitis C virus NS3 protease, PCT WO 98/17679), including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an electrophile such as a boronic acid or phosphonate.
  • Llinas-Brunet et al Hepatitis C inhibitor peptide analogues, PCT WO 99/07734.
  • Non-substrate-based inhibitors such as 2,4,6-trihydroxy-3-nitro-benzamide derivatives(Sudo K. et al., Biochemical and Biophysical Research Communications, 238:643-647, 1997; Sudo K. et al. Antiviral Chemistry and Chemotherapy 9:186, 1998), including RD3-4082 and RD3-4078, the former substituted on the amide with a 14 carbon chain and the latter processing a para-phenoxyphenyl group;
  • Helicase inhibitors (Diana G. D. et al., Compounds, compositions and methods for treatment of hepatitis C, U.S. Pat. No. 5,633,358; Diana G. D. et al., Piperidine derivatives, pharmaceutical compositions thereof and their use in the treatment of hepatitis C, PCT WO 97/36554);
  • 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 22:707-717, 1995), or nucleotides 326-348 comprising the 3′ end of the NCR and nucleotides 371-388 located in the core coding region of the IICV RNA (Alt M. et al., Archives of Virology 142:589-599, 1997; Galderisi U. et al., Journal of Cellular Physiology 181:251-257, 1999);
  • Inhibitors of IRES-dependent translation (Ikeda N et al., Agent for the prevention and treatment of hepatitis C, Japanese Patent Publication JP-08268890; Kai Y. et al. Prevention and treatment of viral diseases, Japanese Patent Publication JP-10101591);
  • miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S. Pat. No. 6,034,134 to Gold et al.), alkyl lipids (U.S. Pat. No. 5,922,757 to Chojkier et al.), vitamin E and other antioxidants (U.S. Pat. No. 5,922,757 to Chojkier et al.), squalene, amantadine, bile acids (U.S. Pat. No. 5,846,964 to Ozeki et al.), N-(phosphonoacetyl)-L-aspartic acid, (U.S. Pat. No.
  • Host including humans, infected with flavivirus or pestivirus, or a gene fragment thereof can be treated by administering to the patient an effective amount of the active compound or a pharmaceutically acceptable prodrug or salt thereof in the presence of a pharmaceutically acceptable carrier or diluent.
  • the active materials can be administered by any appropriate route, for example, orally, parenterally, intravenously, intradermally, subcutaneously, or topically, in liquid or solid form.
  • a preferred dose of the compound for flavivirus or pestivirus infection will be in the range from about 1 to 50 mg/kg, preferably 1 to 20 mg/kg, of body weight per day, more generally 0.1 to about 100 mg per kilogram body weight of the recipient per day.
  • the effective dosage range of the pharmaceutically acceptable salts and prodrugs can be calculated based on the weight of the parent nucleoside to be delivered. If the salt or prodrug exhibits activity in itself, the effective dosage can be estimated as above using the weight of the salt or prodrug, or by other means known to those skilled in the art.
  • the compound is conveniently administered in unit any suitable dosage form, including but not limited to one containing 7 to 3000 mg, preferably 70 to 1400 mg of active ingredient per unit dosage form.
  • a oral dosage of 50-1000 mg is usually convenient.
  • the active ingredient should be administered to achieve peak plasma concentrations of the active compound of from about 0.2 to 70 ⁇ M, preferably about 1.0 to 10 ⁇ M. This may be achieved, for example, by the intravenous injection of a 0.1 to 5% solution of the active ingredient, optionally in saline, or administered as a bolus of the active ingredient.
  • the concentration of active compound in the drug composition 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 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, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.
  • the active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
  • Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets.
  • the active compound can be incorporated with excipients and used in the form of tablets, troches or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compound can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like.
  • a syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • the compound or a pharmaceutically acceptable prodrug or salts thereof can also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antibiotics, antifungals, anti-inflammatories, or other antivirals, including other nucleoside compounds.
  • Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • preferred carriers are physiological saline or phosphate buffered saline (PBS).
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation.
  • Liposomal suspensions are also preferred as pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811 (which is incorporated herein by reference in its entirety). For example, liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container.
  • appropriate lipid(s) such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol
  • aqueous solution of the active compound or its monophosphate, diphosphate, and/or triphosphate derivatives is then introduced into the container.
  • the container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension.
  • nucleosides of the present invention can be synthesized by any means known in the art.
  • the synthesis of the present nucleosides can be achieved by either alkylating the appropriately modified sugar, followed by glycosylation or glycosylation followed by alkylation of the nucleoside.
  • the following non-limiting embodiments illustrate some general methodology to obtain the nucleosides of the present invention.
  • BASE is a purine or pyrimidine base as defined herein;
  • R 7 and R 9 are independently hydrogen, OR 2 , hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO 2 , NH 2 , —NH(lower alkyl), —NH(acyl), —N(lower alkyl) 2 , —N(acyl) 2 ;
  • R 8 and R 10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine; alternatively, R 7 and R 9 , R 7 and R 10 , R 8 and R
  • the key starting material for this process is an appropriately substituted lactone.
  • the lactone can be purchased or can be prepared by any known means including standard epimerization, substitution and cyclization techniques.
  • the lactone can be optionally protected with a suitable protecting group, preferably with an acyl or silyl group, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the protected lactone can then be coupled with a suitable coupling agent, such as an organometallic carbon nucleophile, such as a Grignard reagent, an organolithium, lithium dialkylcopper or R 6 —SiMe 3 in TBAF with the appropriate non-protic solvent at a suitable temperature, to give the 1′-alkylated sugar.
  • a suitable coupling agent such as an organometallic carbon nucleophile, such as a Grignard reagent, an organolithium, lithium dialkylcopper or R 6 —SiMe 3 in TBAF with the appropriate non-protic solvent at a suitable temperature, to give the 1′-alkylated sugar.
  • the optionally activated sugar can then be coupled to the BASE by methods well known to those skilled in the art, as taught by Townsend Chemistry of Nucleosides and Nucleotides , Plenum Press, 1994.
  • an acylated sugar can be coupled to a silylated base with a lewis acid, such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperature.
  • nucleoside can be deprotected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the 1′-C-branched ribonucleoside is desired.
  • the synthesis of a ribonucleoside is shown in Scheme 1.
  • deoxyribo-nucleoside is desired.
  • the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent.
  • the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • the key starting material for this process is an appropriately substituted hexose.
  • the hexose can be purchased or can be prepared by any known means including standard epimerization (e.g. via alkaline treatment), substitution and coupling techniques.
  • the hexose can be selectively protected to give the appropriate hexa-furanose, as taught by Townsend Chemistry of Nucleosides and Nucleotides , Plenum Press, 1994.
  • the 1′-hydroxyl can be optionally activated to a suitable leaving group such as an acyl group or a halogen via acylation or halogenation, respectively.
  • the optionally activated sugar can then be coupled to the BASE by methods well known to those skilled in the art, as taught by Townsend Chemistry of Nucleosides and Nucleotides , Plenum Press, 1994.
  • an acylated sugar can be coupled to a silylated base with a lewis acid, such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperature.
  • a halo-sugar can be coupled to a silylated base with the presence of trimethylsilyltriflate.
  • the 1′-CH 2 —OH if protected, can be selectively deprotected by methods well known in the art.
  • the resultant primary hydroxyl can be functionalized to yield various C-branched nucleosides.
  • the primary hydroxyl can be reduced to give the methyl, using a suitable reducing agent.
  • the hydroxyl can be activated prior to reduction to facilitate the reaction; i.e. via the Barton reduction.
  • the primary hydroxyl can be oxidized to the aldehyde, then coupled with a carbon nucleophile, such as a Grignard reagent, an organolithium, lithium dialkylcopper or R 6 —SiMe 3 in TBAF with the appropriate non-protic solvent at a suitable temperature.
  • a carbon nucleophile such as a Grignard reagent, an organolithium, lithium dialkylcopper or R 6 —SiMe 3 in TBAF with the appropriate non-protic solvent at a suitable temperature.
  • the 1′-C-branched ribonucleoside is desired.
  • the synthesis of a ribonucleoside is shown in Scheme 2.
  • deoxyribo-nucleoside is desired.
  • the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent.
  • the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • L-enantiomers corresponding to the compounds of the invention can be prepared following the same general methods (1 or 2), beginning with the corresponding L-sugar or nucleoside L-enantiomer as starting material.
  • BASE is a purine or pyrimidine base as defined herein;
  • R 7 and R 9 are independently hydrogen, OR 2 , hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO 2 , NH 2 , —NH(lower alkyl), —NH(acyl), —N(lower alkyl) 2 , —N(acyl) 2 ;
  • R 10 is alkyl (including lower alkyl), chlorine, bromine or iodine; alternatively, R 7 and R 9 , or R 7 and R 10 can come together to form a pi bond;
  • the key starting material for this process is an appropriately substituted sugar with a 2′-OH and 2′-H, with the appropriate leaving group (LG), for example an acyl group or a halogen.
  • the sugar can be purchased or can be prepared by any known means including standard epimerization, substitution, oxidation and reduction techniques.
  • the substituted sugar can then be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperature to yield the 2′-modified sugar.
  • Possible oxidizing agents are Jones reagent (a mixture of chromic acid and sulfuric acid), Collins's reagent (dipyridine Cr(VI) oxide, Corey's reagent (pyridinium chlorochromate), pyridinium dichromate, acid dichromate, potassium permanganate, MnO 2 , ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 O 2 -ammonium molybdate, NaBrO 2 —CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (aluminum t-butoxide with another ketone) and N-bromosuccinimide.
  • Jones reagent a mixture of chromic acid and sulfuric acid
  • Collins's reagent dipyridine Cr(VI) oxide
  • an organometallic carbon nucleophile such as a Grignard reagent, an organolithium, lithium dialkylcopper or R 6 —SiMe 3 in TBAF with the ketone with the appropriate non-protic solvent at a suitable temperature, yields the 2′-alkylated sugar.
  • the alkylated sugar can be optionally protected with a suitable protecting group, preferably with an acyl or silyl group, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the optionally protected sugar can then be coupled to the BASE by methods well known to those skilled in the art, as taught by Townsend Chemistry of Nucleosides and Nucleotides , Plenum Press, 1994.
  • an acylated sugar can be coupled to a silylated base with a lewis acid, such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperature.
  • a halo-sugar can be coupled to a silylated base with the presence of trimethylsilyltriflate.
  • nucleoside can be deprotected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the 2′-C-branched ribonucleoside is desired.
  • the synthesis of a ribonucleoside is shown in Scheme 3.
  • deoxyribo-nucleoside is desired.
  • the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent.
  • the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • the key starting material for this process is an appropriately substituted nucleoside with a 2′-OH and 2′-H.
  • the nucleoside can be purchased or can be prepared by any known means including standard coupling techniques.
  • the nucleoside can be optionally protected with suitable protecting groups, preferably with acyl or silyl groups, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the appropriately protected nucleoside can then be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperature to yield the 2′-modified sugar.
  • Possible oxidizing agents are Jones reagent (a mixture of chromic acid and sulfuric acid), Collins's reagent (dipyridine Cr(VI) oxide, Corey's reagent (pyridinium chlorochromate), pyridinium dichromate, acid dichromate, potassium permanganate, MnO 2 , ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 O 2 -ammonium molybdate, NaBrO 2 —CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (aluminum t-butoxide with another ketone) and N-bromosuccinimide.
  • nucleoside can be deprotected by methods well known to those skilled in the art, as taught by GreeneGreene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the 2′-C-branched ribonucleoside is desired.
  • the synthesis of a ribonucleoside is shown in Scheme 4.
  • deoxyribo-nucleoside is desired.
  • the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent.
  • the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • the L-enantiomers are desired. Therefore, the L-enantiomers can be corresponding to the compounds of the invention can be prepared following the same foregoing general methods, beginning with the corresponding L-sugar or nucleoside L-enantiomer as starting material.
  • BASE is a purine or pyrimidine base as defined herein;
  • R 7 and R 9 are independently hydrogen, OR 2 , hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO 2 , NH 2 , —NH(lower alkyl), —NH(acyl), —N(lower alkyl) 2 , —N(acyl) 2 ;
  • R 8 is H, alkyl (including lower alkyl), chlorine, bromine or iodine; alternatively, R 7 and R 9 , or R 8 and R 9 can come together to form a pi bond
  • the key starting material for this process is an appropriately substituted sugar with a 3′-OH and 3′-H, with the appropriate leaving group (LG), for example an acyl group or a halogen.
  • LG leaving group
  • the sugar can be purchased or can be prepared by any known means including standard epimerization, substitution, oxidation and reduction techniques.
  • the substituted sugar can then be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperature to yield the 3′-modified sugar.
  • Possible oxidizing agents are Jones reagent (a mixture of chromic acid and sulfuric acid), Collins's reagent (dipyridine Cr(VI) oxide, Corey's reagent (pyridinium chlorochromate), pyridinium dichromate, acid dichromate, potassium permanganate, MnO 2 , ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 O 2 -ammonium molybdate, NaBrO 2 —CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (aluminum t-butoxide with another ketone) and N-bromosuccinimide.
  • Jones reagent a mixture of chromic acid and sulfuric acid
  • Collins's reagent dipyridine Cr(VI) oxide
  • an organometallic carbon nucleophile such as a Grignard reagent, an organolithium, lithium dialkylcopper or R 6 —SiMe 3 in TBAF with the ketone with the appropriate non-protic solvent at a suitable temperature, yields the 3′-C-branched sugar.
  • the 3′-C-branched sugar can be optionally protected with a suitable protecting group, preferably with an acyl or silyl group, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the optionally protected sugar can then be coupled to the BASE by methods well known to those skilled in the art, as taught by Townsend Chemistry of Nucleosides and Nucleotides , Plenum Press, 1994.
  • an acylated sugar can be coupled to a silylated base with a Lewis acid, such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperature.
  • a halo-sugar can be coupled to a silylated base with the presence of trimethylsilyltriflate.
  • nucleoside can be deprotected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the 3′-C-branched ribonucleoside is desired.
  • the synthesis of a ribonucleoside is shown in Scheme 5.
  • deoxyribo-nucleoside is desired.
  • the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent.
  • the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • the key starting material for this process is an appropriately substituted nucleoside with a 3′-OH and 3′-H.
  • the nucleoside can be purchased or can be prepared by any known means including standard coupling techniques.
  • the nucleoside can be optionally protected with suitable protecting groups, preferably with acyl or silyl groups, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the appropriately protected nucleoside can then be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperature to yield the 2′-modified sugar.
  • Possible oxidizing agents are Jones reagent (a mixture of chromic acid and sulfuric acid), Collins's reagent (dipyridine Cr(VI) oxide, Corey's reagent (pyridinium chlorochromate), pyridinium dichromate, acid dichromate, potassium permanganate, MnO 2 , ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 O 2 -ammonium molybdate, NaBrO 2 —CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (aluminum t-butoxide with another ketone) and N-bromosuccinimide.
  • nucleoside can be deprotected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the 3′-C-branched ribonucleoside is desired.
  • the synthesis of a ribonucleoside is shown in Scheme 6.
  • deoxyribo-nucleoside is desired.
  • the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent.
  • the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • the L-enantiomers are desired. Therefore, the L-enantiomers can be corresponding to the compounds of the invention can be prepared following the same foregoing general methods, beginning with the corresponding L-sugar or nucleoside L-enantiomer as starting material.
  • Example 1 Preparation of 1′-C-methylriboadenine via 6-amino-9-(1-deoxy- ⁇ -D-psicofuranosyl)purine
  • nucleosides of Formula I are prepared.
  • nucleosides of Formula IV are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula VII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • R 1 R 2 R 3 R 6 Base H H H CH 3 O 2,4-O- Diacetyluracil H H H CH 3 O Hypoxanthine H H H CH 3 O 2,4-O- Diacetylthymine H H H CH 3 O Thymine H H H CH 3 O Cytosine H H H CH 3 O 4-(N-mono- acetyl)cytosine H H H CH 3 O 4-(N,N- diacetyl)cytosine H H H CH 3 O Uracil H H H CH 3 O 5-Fluorouracil H H CH 3 S 2,4-O- Diacetyluraci H H H CH 3 S Hypoxanthine H H H CH 3 S 2,4-O- Diacetylthymine H H H CH 3 S Thymine H H H CH 3 S Cytosine H H H CH 3 S 4-(N-mono- acetyl)cytosine H H H CH 3 S 4-(N,N- diacetyl)cytosine H H H CH 3 S
  • nucleosides of Formula VIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • R 1 R 2 R 6 Base H H CH 3 O 2,4-O-Diacetyluracil H H CH 3 O Hypoxanthine H H CH 3 O 2,4-O-Diacetylthymine H H CH 3 O Thymine H H CH 3 O Cytosine H H CH 3 O 4-(N-mono-acetyl)cytosine H H CH 3 O 4-(N,N-diacetyl)cytosine H H CH 3 O Uracil H H CH 3 O 5-Fluorouracil H H CH 3 S 2,4-O-Diacetyluracil H H CH 3 S Hypoxanthine H H CH 3 S 2,4-O-Diacetylthymine H H CH 3 S Thymine H H CH 3 S Cytosine H H CH 3 S 4-(N-mono-acetyl)cytosine H H CH 3 S 4-(N,N-diacetyl)cytosine H H CH 3 S Uracil H H CH 3 S 5-Fluorouracil H
  • nucleosides of Formula IX are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • R 1 R 6 Base H CH 3 O 2,4-O-Diacetyluracil H CH 3 O Hypoxanthine H CH 3 O 2,4-O-Diacetylthymine H CH 3 O Thymine H CH 3 O Cytosine H CH 3 O 4-(N-mono-acetyl)cytosine H CH 3 O 4-(N,N-diacetyl)cytosine H CH 3 O Uracil H CH 3 O 5-Fluorouracil H CH 3 S 2,4-O-Diacetyluracil H CH 3 S Hypoxanthine H CH 3 S 2,4-O-Diacetylthymine H CH 3 S Thymine H CH 3 S Cytosine H CH 3 S 4-(N-mono-acetyl)cytosine H CH 3 S 4-(N,N-diacetyl)cytosine H CH 3 S Uracil H CH 3 S 5-Fluorouracil monophosphate CH 3 O 2,4-O-Diacetyluracil monophosphat
  • nucleosides of Formula XVI are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula II are prepared.
  • nucleosides of Formula V are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula X are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • R 1 R 2 R 3 R 6 Base H H H CH 3 O 2,4-O- Diacetyluracil H H H CH 3 O Hypoxanthine H H H CH 3 O 2,4-O- Diacetylthymine H H H CH 3 O Thymine H H H CH 3 O Cytosine H H H CH 3 O 4-(N-mono- acetyl)cytosine H H H CH 3 O 4-(N,N- diacetyl)cytosine H H H CH 3 O Uracil H H H CH 3 O 5-Fluorouracil H H CH 3 S 2,4-O- Diacetyluraci H H H CH 3 S Hypoxanthine H H H CH 3 S 2,4-O- Diacetylthymine H H H CH 3 S Thymine H H H CH 3 S Cytosine H H H CH 3 S 4-(N-mono- acetyl)cytosine H H H CH 3 S 4-(N,N- diacetyl)cytosine H H H CH 3 S
  • nucleosides of Formula XI are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • R 1 R 6 Base H CH 3 O 2,4-O-Diacetyluracil H CH 3 O Hypoxanthine H CH 3 O 2,4-O-Diacetylthymine H CH 3 O Thymine H CH 3 O Cytosine H CH 3 O 4-(N-mono-acetyl)cytosine H CH 3 O 4-(N,N-diacetyl)cytosine H CH 3 O Uracil H CH 3 O 5-Fluorouracil H CH 3 S 2,4-O-Diacetyluracil H CH 3 S Hypoxanthine H CH 3 S 2,4-O-Diacetylthymine H CH 3 S Thymine H CH 3 S Cytosine H CH 3 S 4-(N-mono-acetyl)cytosine H CH 3 S 4-(N,N-diacetyl)cytosine H CH 3 S Uracil H CH 3 S 5-Fluorouracil monophosphate CH 3 O 2,4-O-Diacetyluracil monophosphat
  • nucleosides of Formula XVII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula III are prepared.
  • nucleosides of Formula VI are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • R 1 R 2 R 3 R 6 Base H H H CH 3 O 2,4-O- Diacetyluracil H H H CH 3 O Hypoxanthine H H H CH 3 O 2,4-O- Diacetylthymine H H H CH 3 O Thymine H H H CH 3 O Cytosine H H H CH 3 O 4-(N-mono- acetyl)cytosine H H H CH 3 O 4-(N,N- diacetyl)cytosine H H H CH 3 O Uracil H H H CH 3 O 5-Fluorouracil H H CH 3 S 2,4-O- Diacetyluraci H H H CH 3 S Hypoxanthine H H H CH 3 S 2,4-O- Diacetylthymine H H H CH 3 S Thymine H H H CH 3 S Cytosine H H H CH 3 S 4-(N-mono- acetyl)cytosine H H H CH 3 S 4-(N,N- diacetyl)cytosine H H H CH 3 S
  • nucleosides of Formula XIV are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • R 1 R 2 R 6 Base H H CH 3 O 2,4-O-Diacetyluracil H H CH 3 O Hypoxanthine H H CH 3 O 2,4-O-Diacetylthymine H H CH 3 O Thymine H H CH 3 O Cytosine H H CH 3 O 4-(N-mono-acetyl)cytosine H H CH 3 O 4-(N,N-diacetyl)cytosine H H CH 3 O Uracil H H CH 3 O 5-Fluorouracil H H CH 3 S 2,4-O-Diacetyluracil H H CH 3 S Hypoxanthine H H CH 3 S 2,4-O-Diacetylthymine H H CH 3 S Thymine H H CH 3 S Cytosine H H CH 3 S 4-(N-mono-acetyl)cytosin H H CH 3 S 4-(N,N-diacetyl)cytosine H H CH 3 S Uracil H H CH 3 S 5-Fluorouracil
  • nucleosides of Formula XV are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • R 1 R 6 Base H CH 3 O 2,4-O-Diacetyluracil H CH 3 O Hypoxanthine H CH 3 O 2,4-O-Diacetylthymine H CH 3 O Thymine H CH 3 O Cytosine H CH 3 O 4-(N-mono-acetyl)cytosine H CH 3 O 4-(N,N-diacetyl)cytosine H CH 3 O Uracil H CH 3 O 5-Fluorouracil H CH 3 S 2,4-O-Diacetyluracil H CH 3 S Hypoxanthine H CH 3 S 2,4-O-Diacetylthymine H CH 3 S Thymine H CH 3 S Cytosine H CH 3 S 4-(N-mono-acetyl)cytosine H CH 3 S 4-(N,N-diacetyl)cytosine H CH 3 S Uracil H CH 3 S 5-Fluorouracil monophosphate CH 3 O 2,4-O-Diacetyluracil monophosphat
  • nucleosides of Formula XVIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Compounds can exhibit anti-flavivirus or pestivirus activity by inhibiting flavivirus or pestivirus polymerase, by inhibiting other enzymes needed in the replication cycle, or by other pathways.
  • test compounds were dissolved in DMSO at an initial concentration of 200 and then were serially diluted in culture medium.
  • BHK-21 baby hamster kidney (ATCC CCL-10) and Bos Taurus (BT) (ATCC CRL 1390) cells were grown at 37° C. in a humidified CO 2 (5%) atmosphere.
  • FBS fetal bovine serum
  • BT cells were passaged in Dulbecco's modified Eagle's medium with 4 mM L-glutamine and 10% horse serum (HS, Gibco), adjusted to contain 1.5 g/L sodium bicarbonate, 4.5 g/L glucose and 1.0 mM sodium pyruvate.
  • the vaccine strain 17D (YFV-17D) (Stamaril®, Pasteur Merieux) and Bovine Viral Diarrhea virus (BVDV) (ATCC VR-534) were used to infect BHK and BT cells, respectively, in 75 cm 2 bottles. After a 3 day incubation period at 37° C., extensive cytopathic effect was observed.
  • HepG2 cells were obtained from the American Type Culture Collection (Rockville, Md.), and were grown in 225 cm 2 tissue culture flasks in minimal essential medium supplemented with non-essential amino acids, 1% penicillin-streptomycin. The medium was renewed every three days, and the cells were subcultured once a week.
  • confluent HepG2 cells were seeded at a density of 2.5 ⁇ 10 6 cells per well in a 6-well plate and exposed to 10 ⁇ M of [ 3 H] labeled active compound (500 dpm/pmol) for the specified time periods.
  • the cells were maintained at 37° C. under a 5% CO 2 atmosphere.
  • the cells were washed three times with ice-cold phosphate-buffered saline (PBS). Intracellular active compound and its respective metabolites were extracted by incubating the cell pellet overnight at ⁇ 20° C.
  • PBS ice-cold phosphate-buffered saline
  • the cynomolgus monkey was surgically implanted with a chronic venous catheter and subcutaneous venous access port (VAP) to facilitate blood collection and underwent a physical examination including hematology and serum chemistry evaluations and the body weight was recorded.
  • VAP chronic venous catheter and subcutaneous venous access port
  • Each monkey (six total), received approximately 250 uCi of 3 H activity with each dose of active compound, namely ⁇ -D-2′-CH 3 -riboG at a dose level of 10 mg/kg at a dose concentration of 5 mg/mL, either via an intravenous bolus (3 monkeys, IV), or via oral gavage (3 monkeys, PO).
  • Each dosing syringe was weighed before dosing to gravimetrically determine the quantity of formulation administered.
  • Urine samples were collected via pan catch at the designated intervals (approximately 18-0 hours pre-dose, 0-4, 4-8 and 8-12 hours post-dosage) and processed. Blood samples were collected as well (pre-dose, 0.25, 0.5, 1, 2, 3, 6, 8, 12 and 24 hours post-dosage) via the chronic venous catheter and VAP or from a peripheral vessel if the chronic venous catheter procedure should not be possible.
  • C max maximum concentration
  • T max time when the maximum concentration was achieved
  • AUC area under the curve
  • T 1/2 half life of the dosage concentration
  • CL clearance
  • V ss steady state volume and distribution
  • F bioavailability
  • Human bone marrow cells were collected from normal healthy volunteers and the mononuclear population was separated by Ficoll-Hypaque gradient centrifugation as described previously by Sommadossi J-P, Carlisle R. “Toxicity of 3′-azido-3′-deoxythymidine and 9-(1,3-dihydroxy-2-propoxymethyl)guanine for normal human hematopoietic progenitor cells in vitro” Antimicrobial Agents and Chemotherapy 1987; 31:452-454; and Sommadossi J-P, Schinazi R F, Chu C K, Xie M-Y.
  • HepG2 cells were cultured in 12-well plates as described above and exposed to various concentrations of drugs as taught by Pan-Zhou X-R, Cui L, Zhou X-J, Sommadossi J-P, Darley-Usmer V M. “Differential effects of antiretroviral nucleoside analogs on mitochondrial function in HepG2 cells” Antimicrob Agents Chemother 2000; 44:496-503. Lactic acid levels in the culture medium after 4 day drug exposure was measured using a Boehringer lactic acid assay kit. Lactic acid levels were normalized by cell number as measured by hemocytometer count. The preliminary results from this assay are tabulated in Table 5.
  • the assay was performed essentially as described by Baginski, S. G.; Pevear, D. C.; Seipel, M.; Sun, S. C. C.; Benetatos, C. A.; Chunduru, S. K.; Rice, C. M. and M. S. Collett “Mechanism of action of a pestivirus antiviral compound” PNAS USA 2000, 97(14), 7981-7986.
  • MDBK cells ATCC were seeded onto 96-well culture plates (4,000 cells per well) 24 hours before use.
  • test compounds were added to both infected and uninfected cells in a final concentration of 0.5% DMSO in growth medium. Each dilution was tested in quadruplicate. Cell densities and virus inocula were adjusted to ensure continuous cell growth throughout the experiment and to achieve more than 90% virus-induced cell destruction in the untreated controls after four days post-infection. After four days, plates were fixed with 50% TCA and stained with sulforhodamine B. The optical density of the wells was read in a microplate reader at 550 nm.
  • FIGS. 4 and 5 provide a graphical illustration of the methodology used to arrive at the 50% effective concentration (EC 50 ) values for ⁇ -D-2′-CH 3 -riboG and ribavirin.
  • FIG. 6 compares the results of the CPA for ⁇ -D-2′-CH 3 -riboG, ⁇ -D-2′-CH 3 -riboC, riboU, ⁇ -D-2′-CH 3 -riboA and ribavirin
  • FIG. 7 is a graphical illustration of the results from the Plaque Reduction Assay.
  • FIG. 8 is an image of BVDV plaque formation in the presence of increasing concentrations of ⁇ -D-2′-CH 3 -riboU.
  • MDBK cells were seeded onto 24-well plates (2 ⁇ 105 cells per well) 24 hours before infection with BVDV (NADL strain) at a multiplicity of infection (MOI) of 0.1 PFU per cell.
  • MOI multiplicity of infection
  • Serial dilutions of test compounds were added to cells in a final concentration of 0.5% DMSO in growth medium. Each dilution was tested in triplicate. After three days, cell cultures (cell monolayers and supernatants) were lysed by three freeze-thaw cycles, and virus yield was quantified by plaque assay.
  • MDBK cells were seeded onto 6-well plates (5 ⁇ 105 cells per well) 24 h before use.
  • FIG. 9 is a graphical illustration of the results from the Yield Reduction Assay.
  • FIG. 8 is an image of BVDV yield reduction in the presence of increasing concentrations of ⁇ -D-2′-CH 3 -riboC.
  • Table 10 summarizes the cytoxicity of two compounds of this invention, ⁇ -D-1′-CH 3 -riboA and ⁇ -D-2′-CH 3 -riboA, in comparison to RBV (“ribavirin”), in various cell systems.
  • Table 11 summarizes the antiviral activity of ⁇ -D-1′-CH 3 -riboA and ⁇ -D-2′-CH 3 -riboA against several viruses within the flavivirus and pestivirus genuses.
  • Table 12 summarizes the antiviral activity and toxicity of ⁇ -D-2′-methyl-riboG, ⁇ -D-2′-methyl-riboC and ⁇ -D-2′-methyl-riboU, against a couple of viruses within the flavivirus and pestivirus genuses.
  • Table 13 summarizes the anti-viral activity of several compounds of this invention against BVDV in three different assays.

Abstract

A method and composition for treating a host infected with flavivirus or pestivirus comprising administering an effective flavivirus or pestivirus treatment amount of a described 1′, 2′ or 3′-modified nucleoside or a pharmaceutically acceptable salt or prodrug thereof, is provided.

Description

  • This application claims priority to U.S. provisional application No. 60/207,674, filed on May 26, 2000 and U.S. provisional application No. 60/283,276, filed on Apr. 11, 2001.
  • FIELD OF THE INVENTION
  • This invention is in the area of pharmaceutical chemistry, and in particular, is a compound, method and composition for the treatment of flaviviruses and pestiviruses.
  • BACKGROUND OF THE INVENTION
  • Pestiviruses and flaviviruses belong to the Flaviviridae family of viruses along with hepatitis C virus. The pestivirus genus includes bovine viral diarrhea virus (BVDV), classical swine fever virus (CSFV, also called hog cholera virus) and border disease virus (BDV) of sheep (Moennig, V. et al. Adv. Vir. Res. 1992, 41, 53-98). Pestivirus infections of domesticated livestock (cattle, pigs and sheep) cause significant economic losses worldwide. BVDV causes mucosal disease in cattle and is of significant economic importance to the livestock industry (Meyers, G. and Thiel, H.-J., Advances in Virus Research, 1996, 47, 53-118; Moennig V., et al, Adv. Vir. Res. 1992, 41, 53-98).
  • Human pestiviruses have not been as extensively characterized as the animal pestiviruses. However, serological surveys indicate considerable pestivirus exposure in humans. Pestivirus infections in man have been implicated in several diseases including congenital brain injury, infantile gastroenteritis and chronic diarrhea in human immunodeficiency virus (HIV) positive patients. M. Giangaspero et al., Arch. Virol. Suppl., 1993, 7, 53-62; M. Giangaspero et al., Int. J. Std. Aids, 1993, 4 (5): 300-302.
  • The flavivirus genus includes more than 68 members separated into groups on the basis of serological relatedness (Calisher et al., J. Gen. Virol, 1993, 70, 37-43). Clinical symptoms vary and include fever, encephalitis and hemorrhagic fever. Fields Virology, Editors: Fields, B. N., Knipe, D. M., and Howley, P. M., Lippincott-Raven Publishers, Philadelphia, Pa., 1996, Chapter 31, 931-959. Flaviviruses of global concern that are associated with human disease include the dengue hemorrhagic fever viruses (DHF), yellow fever virus, shock syndrome and Japanese encephalitis virus. Halstead, S. B., Rev. Infect. Dis., 1984, 6, 251-264; Halstead, S. B., Science, 239:476-481, 1988; Monath, T. P., New Eng. J. Med., 1988, 319, 641-643.
  • Examples of antiviral agents that have been identified as active against the flavivirus or pestiviruses include:
      • (1) interferon and ribavirin (Battaglia, A. M. et al., Ann. Pharmacother, 2000, 34, 487-494); Berenguer, M. et al. Antivir. Ther., 1998, 3 (Suppl. 3), 125-136);
      • (2) 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. DE 19914474; Tung et al. Inhibitors of serine proteases, particularly hepatitis C virus NS3 protease, PCT WO 98/17679), including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an electrophile such as a boronic acid or phosphonate (Llinas-Brunet et al, Hepatitis C inhibitor peptide analogues, PCT WO 99/07734).
      • (3) Non-substrate-based inhibitors such as 2,4,6-trihydroxy-3-nitro-benzamide derivatives (Sudo K. et al., Biochemical and Biophysical Research Communications, 1997, 238, 643-647; Sudo K. et al. Antiviral Chemistry and Chemotherapy, 1998, 9, 186), including RD3-4082 and RD3-4078, the former substituted on the amide with a 14 carbon chain and the latter processing a para-phenoxyphenyl group;
      • (4) Thiazolidine derivatives which show relevant inhibition in a reverse-phase HPLC assay with an NS3/4A fusion protein and NS5A/5B substrate (Sudo K. et al., Antiviral Research, 1996, 32, 9-18), especially compound RD-1-6250, possessing a fused cinnamoyl moiety substituted with a long alkyl chain, RD4 6205 and RD4 6193;
      • (5) Thiazolidines and benzanilides identified in Kakiuchi N. et al. J. EBS Letters 421, 217-220; Takeshita N. et al. Analytical Biochemistry, 1997, 247, 242-246;
      • (6) A phenan-threnequinone possessing activity against protease in a SDS-PAGE and autoradiography assay isolated from the fermentation culture broth of Streptomyces sp., Sch 68631 (Chu M. et al., Tetrahedron Letters, 1996, 37, 7229-7232), and Sch 351633, isolated from the fungus Penicillium griscofuluum, which demonstrates activity in a scintillation proximity assay (Chu M. et al., Bioorganic and Medicinal Chemistry Letters 9, 1949-1952);
      • (7) Selective NS3 inhibitors based on the macromolecule elgin c, isolated from leech (Qasim M. A. et al., Biochemistry, 1997, 36, 1598-1607);
      • (8) Helicase inhibitors (Diana G. D. et al., Compounds, compositions and methods for treatment of hepatitis C, U.S. Pat. No. 5,633,358; Diana G. D. et al., Piperidine derivatives, pharmaceutical compositions thereof and their use in the treatment of hepatitis C, PCT WO 97/36554);
      • (9) Polymerase inhibitors such as nucleotide analogues, gliotoxin (Ferrari R. et al. Journal of Virology, 1999, 73, 1649-1654), and the natural product cerulenin (Lohmann V. et al., Virology, 1998, 249, 108-118);
      • (10) 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 IICV RNA (Alt M. et al., Archives of Virology, 1997, 142, 589-599; Galderisi U. et al., Journal of Cellular Physiology, 1999, 181, 251-257);
      • (11) 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);
      • (12) Nuclease-resistant ribozymes (Maccjak, D. J. et al., Hepatology 1999, 30, abstract 995); and
      • (13) Other miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S. Pat. No. 6,034,134 to Gold et al.), alkyl lipids (U.S. Pat. No. 5,922,757 to Chojkier et al), vitamin E and other antioxidants (U.S. Pat. No. 5,922,757 to Chojkier et al.), squalene, amantadine, bile acids (U.S. Pat. No. 5,846,964 to Ozeki et al.), N-(phosphonoacetyl)-L-aspartic acid, (U.S. Pat. No. 5,830,905 to Diana et benzenedicarboxamides (U.S. Pat. No. 5,633,388 to Diana et al.), polyadenylic acid derivatives (U.S. Pat. No. 5,496,546 to Wang et al.), 2′,3′-dideoxyinosine (U.S. Pat. No. 5,026,687 to Yarchoan et al.), and benzimidazoles (U.S. Pat. No. 5,891,874 to Colacino et al.).
  • In view of the severity of diseases associated with pestiviruses and flaviviruses, and their pervasiveness in animal and man, it is an object of the present invention to provide a compound, method and composition for the treatment of a host infected with flavivirus or pestivirus.
  • SUMMARY OF THE INVENTION
  • Compounds, methods and compositions for the treatment of a host infected with a flavivirus or pestivirus infection are described that includes an effective treatment amount of a β-D- or β-L-nucleoside of the Formulas (I)-(XVIII), or a pharmaceutically acceptable salt or prodrug thereof.
  • In a first principal embodiment, a compound of Formula I, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00001
  • wherein:
    R1, R2 and R3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 and X2 are independently selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a second principal embodiment, a compound of Formula II, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00002
  • wherein:
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 and X2 are independently selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a third principal embodiment, a compound of Formula III, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00003
  • wherein:
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 and X2 are independently selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a fourth principal embodiment, a compound of Formula IV, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00004
  • wherein:
    R1, R2 and R3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 is selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a fifth principal embodiment, a compound of Formula V, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00005
  • wherein:
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 is selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a sixth principal embodiment, a compound of Formula VI, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00006
  • wherein:
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 is selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a seventh principal embodiment, a compound selected from Formulas VII, VIII and IX, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00007
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, 2-Br-ethyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), CF3, chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2; and
    X is O, S, SO2 or CH2.
  • In a eighth principal embodiment, a compound of Formulas X, XI and XII, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00008
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R7 is hydrogen, OR3, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2; and
    X is O, S, SO2 or CH2.
  • In a ninth principal embodiment a compound selected from Formulas XIII, XIV and XV, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00009
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2; and
    X is O, S, SO2, or CH2.
  • In a tenth principal embodiment the invention provides a compound of Formula XVI, or a pharmaceutically acceptable salt or prodrug thereof:
  • Figure US20180235993A1-20180823-C00010
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1 and R2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 or R2 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R7 and R9 are independently hydrogen, OR2, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine;
    alternatively, R7 and R9, R7 and R10, R8 and R9, or R8 and R10 can come together to form a pi bond; and
    X is O, S, SO2 or CH2.
  • In a eleventh principal embodiment the invention provides a compound of Formula XVII, or a pharmaceutically acceptable salt or prodrug thereof:
  • Figure US20180235993A1-20180823-C00011
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1 and R2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 or R2 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R7 and R9 are independently hydrogen, OR2, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R10 is H, alkyl (including lower alkyl), chlorine, bromine or iodine;
    alternatively, R7 and R9, or R7 and R10 can come together to form a pi bond; and
    X is O, S, SO2 or CH2.
  • In an twelfth principal embodiment, the invention provides a compound of Formula XVIII, or a pharmaceutically acceptable salt or prodrug thereof:
  • Figure US20180235993A1-20180823-C00012
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1 and R2 independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl);
  • sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 or R2 is independently H or phosphate;
  • R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(lower-alkyl)amino;
    R8 is H, alkyl (including lower alkyl), chlorine, bromine or iodine;
    alternatively, R7 and R9, or R8 and R9 can come together to form a pi bond;
    X is O, S, SO2 or CH2.
  • The β-D- and β-L-nucleosides of this invention may inhibit flavivirus or pestivirus polymerase activity. These nucleosides can be assessed for their ability to inhibit flavivirus or pestivirus polymerase activity in vitro according to standard screening methods.
  • In one embodiment the efficacy of the anti-flavivirus or pestivirus compound is measured according to the concentration of compound necessary to reduce the plaque number of the virus in vitro, according to methods set forth more particularly herein, by 50% (i.e. the compound's EC50). In preferred embodiments the compound exhibits an EC50 of less than 15 or preferably, less than 10 micromolar in vitro.
  • In another embodiment, the active compound can be administered in combination or alternation with another anti-flavivirus or pestivirus agent. In combination therapy, effective dosages of two or more agents are administered together, whereas during alternation therapy an effective dosage of each agent is administered serially. The dosages 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.
  • HCV is a member of the Flaviviridae family; however, now, HCV has been placed in a new monotypic genus, hepacivirus. Therefore, in one embodiment, the flavivirus or pestivirus is not HCV.
  • Nonlimiting examples of antiviral agents that can be used in combination with the compounds disclosed herein include:
  • (1) an interferon and/or ribavirin (Battaglia, A. M. et al., Ann. Pharmacother. 34:487-494, 2000); Berenguer, M. et al. Antivir. Ther. 3(Suppl. 3):125-136, 1998);
  • (2) Substrate-based NS3 protease inhibitors (Attwood et al., Antiviral peptide derivatives, PCT WO 98/22496, 1998; Attwood et al., Antiviral Chemistry and Chemotherapy 10.259-273, 1999; Attwood et al., Preparation and use of amino acid derivatives as anti-viral agents, German Patent Publication DE 19914474; Tung et al. Inhibitors of serine proteases, particularly hepatitis C virus NS3 protease, PCT WO 98/17679), including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an electrophile such as a boronic acid or phosphonate. Llinas-Brunet et al, Hepatitis C inhibitor peptide analogues, PCT WO 99/07734.
  • (3) Non-substrate-based inhibitors such as 2,4,6-trihydroxy-3-nitro-benzamide derivatives(Sudo K. et al., Biochemical and Biophysical Research Communications, 238:643-647, 1997; Sudo K. et al. Antiviral Chemistry and Chemotherapy 9:186, 1998), including RD3-4082 and RD3-4078, the former substituted on the amide with a 14 carbon chain and the latter processing a para-phenoxyphenyl group;
  • (4) Thiazolidine derivatives which show relevant inhibition in a reverse-phase HPLC assay with an NS3/4A fusion protein and NS5A/5B substrate (Sudo K. et al., Antiviral Research 32:9-18, 1996), especially compound RD-1-6250, possessing a fused cinnamoyl moiety substituted with a long alkyl chain, RD4 6205 and RD4 6193;
  • (5) Thiazolidines and benzanilides identified in Kakiuchi N. et al. J. EBS Letters 421:217-220; Takeshita N. et al. Analytical Biochemistry 247:242-246, 1997;
  • (6) A phenan-threnequinone possessing activity against protease in a SDS-PAGE and autoradiography assay isolated from the fermentation culture broth of Streptomyces sp., Sch 68631 (Chu M. et al., Tetrahedron Letters 37:7229-7232, 1996), and Sch 351633, isolated from the fungus Penicillium griscofuluum, which demonstrates activity in a scintillation proximity assay (Chu M. et al., Bioorganic and Medicinal Chemistry Letters 9:1949-1952);
  • (7) Selective NS3 inhibitors based on the macromolecule elgin c, isolated from leech (Qasim M. A. et al., Biochemistry 36:1598-1607, 1997);
  • (8) Helicase inhibitors (Diana G. D. et al., Compounds, compositions and methods for treatment of hepatitis C, U.S. Pat. No. 5,633,358; Diana G. D. et al., Piperidine derivatives, pharmaceutical compositions thereof and their use in the treatment of hepatitis C, PCT WO 97/36554);
  • (9) Polymerase inhibitors such as nucleotide analogues, gliotoxin (Ferrari R. et al. Journal of Virology 73:1649-1654, 1999), and the natural product cerulenin (Lohmann V. et al., Virology 249:108-118, 1998);
  • (10) Antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5′ non-coding region (NCR) of the virus (Alt M. et al., Hepatology 22:707-717, 1995), or nucleotides 326-348 comprising the 3′ end of the NCR and nucleotides 371-388 located in the core coding region of the IICV RNA (Alt M. et al., Archives of Virology 142:589-599, 1997; Galderisi U. et al., Journal of Cellular Physiology 181:251-257, 1999);
  • (11) Inhibitors of IRES-dependent translation (Ikeda N et al., Agent for the prevention and treatment of hepatitis C, Japanese Patent Publication JP-08268890; Kai Y. et al. Prevention and treatment of viral diseases, Japanese Patent Publication JP-10101591);
  • (12) Nuclease-resistant ribozymes. (Maccjak D. J. et al., Hepatology 30 abstract 995, 1999); and
  • (13) Other miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S. Pat. No. 6,034,134 to Gold et al.), alkyl lipids (U.S. Pat. No. 5,922,757 to Chojkier et al.), vitamin E and other antioxidants (U.S. Pat. No. 5,922,757 to Chojkier et al.), squalene, amantadine, bile acids (U.S. Pat. No. 5,846,964 to Ozeki et al.), N-(phosphonoacetyl)-L-aspartic acid, (U.S. Pat. No. 5,830,905 to Diana et al.), benzenedicarboxamides (U.S. Pat. No. 5,633,388 to Diana et al.), polyadenylic acid derivatives (U.S. Pat. No. 5,496,546 to Wang et al.), 2′,3′-dideoxyinosine (U.S. Pat. No. 5,026,687 to Yarchoan et al.), and benzimidazoles (U.S. Pat. No. 5,891,874 to Colacino et al.).
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 provides the structure of various non-limiting examples of nucleosides of the present invention, as well as other known nucleosides, FIAU and Ribavirin, which are used as comparative examples in the text.
  • FIG. 2 is a line graph of the pharmacokinetics (plasma concentrations) of β-D-2′-CH3-riboG administered to Cynomolgus Monkeys over time after administration.
  • FIGS. 3a and 3b are line graphs of the pharmacokinetics (plasma concentrations) of β-D-2′-CH3-riboG administered to Cynomolgus Monkeys either intravenously (3a) or orally (3b) over time after administration.
  • FIG. 4 depicts line graphs of the results of the cell protection assay of β-D-2′-CH3-riboG against BVDV.
  • FIG. 5 depicts line graphs of the results of the cell protection assay of ribavirin against BVDV.
  • FIG. 6 are line graphs of the cell protection assay of β-D-2′-CH3-riboG, β-D-2′-CH3-riboC, β-D-2′-CH3-riboU, β-D-2′-CH3-riboA and ribavirin.
  • FIG. 7 are line graphs of the results of the plaque reduction assay for β-D-2′-CH3-riboU, β-D-2′-CH3-riboC and β-D-2′-CH3-riboG.
  • FIG. 8 is an illustration of plaque reduction based on increasing concentrations of β-D-2′-CH3-riboU.
  • FIG. 9 is a line graph of the results of the yield reduction assay for β-D-2′-CH3-riboG, depicting a 4 log reduction at 9 μM.
  • FIG. 10 is an illustration of the yield reduction based on increasing concentrations of β-D-2′-CH3-riboC.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention as disclosed herein is a compound, method and composition for the treatment of pestiviruses and flaviviruses in humans and other host animals, that includes the administration of an effective flavivirus or pestivirus treatment amount of an β-D- or β-L-nucleoside as described herein or a pharmaceutically acceptable salt or prodrug thereof, optionally in a pharmaceutically acceptable carrier. The compounds of this invention either possess antiviral (i.e., anti-flavivirus or pestivirus) activity, or are metabolized to a compound that exhibits such activity.
  • In summary, the present invention includes the following features:
      • (a) β-D- and β-L-nucleosides, as described herein, and pharmaceutically acceptable salts and prodrugs thereof;
      • (b) β-D- and β-L-nucleosides as described herein, and pharmaceutically acceptable salts and prodrugs thereof for use in the treatment or prophylaxis of a flavivirus or pestivirus infection, especially in individuals diagnosed as having a flavivirus or pestivirus infection or being at risk for becoming infected by flavivirus or pestivirus;
      • (c) use of these β-D- and β-L-nucleosides, and pharmaceutically acceptable salts and prodrugs thereof in the manufacture of a medicament for treatment of a flavivirus or pestivirus infection;
      • (d) pharmaceutical formulations comprising the β-D- and β-L-nucleosides or pharmaceutically acceptable salts or prodrugs thereof together with a pharmaceutically acceptable carrier or diluent;
      • (e) β-D- and β-L-nucleosides as described herein substantially in the absence of enantiomers of the described nucleoside, or substantially isolated from other chemical entities;
      • (f) processes for the preparation of β-D- and β-L-nucleosides, as described in more detail below; and
      • (g) processes for the preparation of β-D- and β-L-nucleosides substantially in the absence of enantiomers of the described nucleoside, or substantially isolated from other chemical entities.
  • Flaviviruses included within the scope of this invention are discussed generally in Fields Virology, Editors: Fields, B. N., Knipe, D. M., and Howley, P. M., Lippincott-Raven Publishers, Philadelphia, Pa., Chapter 31, 1996. Specific flaviviruses include, without limitation: Absettarov, Alfuy, Apoi, Aroa, Bagaza, Banzi, Bouboui, Bussuquara, Cacipacore, Carey Island, 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, Karshi, Kedougou, Kokobera, Koutango, Kumlinge, Kunjin, Kyasanur Forest disease, Langat, Louping ill, Meaban, Modoc, Montana myotis leukoencephalitis, Murray valley encephalitis, Naranjal, Negishi, Ntaya, Omsk hemorrhagic fever, Phnom-Penh bat, Powassan, Rio Bravo, Rocio, Royal Farm, Russian spring-summer encephalitis, Saboya, St. Louis encephalitis, Sal Vieja, San Perlita, Saumarez Reef, Sepik, Sokuluk, Spondweni, Stratford, Tembusu, Tyuleniy, Uganda S, Usutu, Wesselsbron, West Nile, Yaounde, Yellow fever, and Zika.
  • Pestiviruses included within the scope of this invention are discussed generally in Fields Virology, Editors: Fields, B. N., Knipe, D. M., and Howley, P. M., Lippincott-Raven Publishers, Philadelphia, Pa., Chapter 33, 1996. Specific pestiviruses include, without limitation: bovine viral diarrhea virus (“BVDV”), classical swine fever virus (“CSFV,” also called hog cholera virus), and border disease virus (“BDV”).
  • I. Active Compound, and Physiologically Acceptable Salts and Prodrugs Thereof
  • In a first principal embodiment, a compound of Formula I, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00013
  • wherein:
    R1, R2 and R3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 and X2 are independently selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a preferred subembodiment, a compound of Formula I, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • R1, R2 and R3 are independently H or phosphate (preferably H);
  • X1 is H;
  • X2 is H or NH2; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, NH2 or OH.
  • In a second principal embodiment, a compound of Formula II, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00014
  • wherein:
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 and X2 are independently selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a preferred subembodiment, a compound of Formula II, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • R1, R2 and R3 are independently H or phosphate (preferably H);
  • X1 is H;
  • X2 is H or NH2; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, NH2 or OH.
  • In a third principal embodiment, a compound of Formula III, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00015
  • wherein:
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 and X2 are independently selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a preferred subembodiment, a compound of Formula III, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • R1, R2 and R3 are independently H or phosphate (preferably H);
  • X1 is H;
  • X2 is H or NH2; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, NH2 or OH.
  • In a fourth principal embodiment, a compound of Formula IV, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00016
  • wherein:
    R1, R2 and R3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 is selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a preferred subembodiment, a compound of Formula IV, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • R1, R2 and R3 are independently H or phosphate (preferably H);
    X1 is H or CH3; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, NH2 or OH.
  • In a fifth principal embodiment, a compound of Formula V, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00017
  • wherein:
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 is selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a preferred subembodiment, a compound of Formula V, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • R1, R2 and R3 are independently H or phosphate (preferably H);
    X1 is H or CH3; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, NH2 or OH.
  • In a sixth principal embodiment, a compound of Formula VI, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00018
  • wherein:
  • R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate; and
  • Y is hydrogen, bromo, chloro, fluoro, iodo, OR4, NR4R5 or SR4;
    X1 is selected from the group consisting of H, straight chained, branched or cyclic alkyl, CO-alkyl, CO-aryl, CO-alkoxyalkyl, chloro, bromo, fluoro, iodo, OR4, NR4NR5 or SR5; and
    R4 and R5 are independently hydrogen, acyl (including lower acyl), or alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl).
  • In a preferred subembodiment, a compound of Formula VI, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • R1, R2 and R3 are independently H or phosphate (preferably H);
    X1 is H or CH3; and
    Y is hydrogen, bromo, chloro, fluoro, iodo, NH2 or OH.
  • In a seventh principal embodiment, a compound selected from Formulas VII, VIII and IX, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00019
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, 2-Br-ethyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), CF3, chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2; and
    X is O, S, SO2, or CH2.
  • In a first preferred subembodiment, a compound of Formula VII, VIII or IX, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently hydrogen or phosphate;
    R6 is alkyl; and
    X is O, S, SO2 or CH2.
  • In a second preferred subembodiment, a compound of Formula VII, VIII or IX, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are hydrogens;
    R6 is alkyl; and
    X is O, S, SO2 or CH2.
  • In a third preferred subembodiment, a compound of Formula VII, VIII or IX, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently hydrogen or phosphate;
    R6 is alkyl; and
  • X is O.
  • In a eighth principal embodiment, a compound of Formula X, XI or XII, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00020
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R7 is hydrogen, OR3, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(loweralkyl)2, —N(acyl)2; and
    X is O, S, SO2 or CH2.
  • In a first preferred subembodiment, a compound of Formula X, XI or XII, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently hydrogen or phosphate;
    R6 is alkyl; and
    X is O, S, SO2 or CH2.
  • In a second preferred subembodiment, a compound of Formula X, XI or XII, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are hydrogens;
    R6 is alkyl; and
    X is O, S, SO2 or CH2.
  • In a third preferred subembodiment, a compound of Formula X, XI or XII, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently H or phosphate;
    R6 is alkyl; and
  • X is O.
  • In even more preferred subembodiments, a compound of Formula XI, or its pharmaceutically acceptable salt or prodrug, is provided:
  • Figure US20180235993A1-20180823-C00021
  • wherein:
    Base is a purine or pyrimidine base as defined herein; optionally substituted with an amine or cyclopropyl (e.g., 2-amino, 2,6-diamino or cyclopropyl guanosine); and
    R1 and R2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 or R2 is independently H or phosphate.
  • In a ninth principal embodiment a compound selected from Formula XIII, XIV or XV, or a pharmaceutically acceptable salt or prodrug thereof, is provided:
  • Figure US20180235993A1-20180823-C00022
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 or R3 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2; and
    X is O, S, SO2 or CH2.
  • In a first preferred subembodiment, a compound of Formula XIII, XIV or XV, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently hydrogen or phosphate;
    R6 is alkyl; and
    X is O, S, SO2 or CH2.
  • In a second preferred subembodiment, a compound of Formula XIII, XIV or XV, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are hydrogens;
    R6 is alkyl; and
    X is O, S, SO2 or CH2.
  • In a third preferred subembodiment, a compound of Formula XIII, XIV or XV, or a pharmaceutically acceptable salt or prodrug thereof, is provided wherein:
  • Base is a purine or pyrimidine base as defined herein;
    R1, R2 and R3 are independently hydrogen or phosphate;
    R6 is alkyl; and
  • X is O.
  • In a tenth principal embodiment the invention provides a compound of Formula XVI, or a pharmaceutically acceptable salt or prodrug thereof:
  • Figure US20180235993A1-20180823-C00023
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1 and R2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 and R2 are independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R7 and R9 are independently hydrogen, OR2, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine;
    alternatively, R7 and R9, R7 and R10, R8 and R9, or R8 and R10 can come together to form a pi bond; and
    X is O, S, SO2 or CH2.
  • In a first preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2, alkyl, alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(loweralkyl)amino; (5) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O, S, SO2 or CH2.
  • In a second preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl, alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O, S, SO2 or CH2.
  • In a third preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl, alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2, alkyl, alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(loweralkyl)amino; (5) R8 and R10 are H; and (6) X is O, S, SO2 or CH2.
  • In a fourth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl, alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently OR2, alkyl, alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (5) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O.
  • In a fifth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR1; (5) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine; and (6) X is O, S, SO2 or CH2.
  • In a sixth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R′ is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R′ is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (5) R8 and R10 are H; and (6) X is O, S, SO2, or CH2.
  • In a seventh preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R′ is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R′ is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(loweralkyl)amino; (5) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine; and (6) X is O.
  • In a eighth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R8 and R10 are hydrogen; and (6) X is O, S, SO2 or CH2.
  • In a ninth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R′ is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine; and (6) X is O.
  • In a tenth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (5) R8 and R10 are hydrogen; and (6) X is O.
  • In an eleventh preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R8 and R10 are hydrogen; and (6) X is O, S, SO2 or CH2.
  • In a twelfth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2; (5) R8 and R10 are hydrogen; and (6) X is O, S, SO2, or CH2.
  • In a thirteenth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2; (5) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O.
  • In a fourteenth preferred subembodiment, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(loweralkyl)amino; (5) R8 and R10 are hydrogen; and (6) X is O.
  • In even more preferred subembodiments, a compound of Formula XVI, or its pharmaceutically acceptable salt or prodrug, is provided in which:
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is guanine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is cytosine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl;
  • (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is thymine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is uracil; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is phosphate; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is ethyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is propyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is butyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 is hydrogen and R9 is hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is S;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is SO2;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is CH2;
  • In a eleventh principal embodiment the invention provides a compound of Formula XVII, or a pharmaceutically acceptable salt or prodrug thereof:
  • Figure US20180235993A1-20180823-C00024
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1 is H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R7 and R9 are independently hydrogen, OR2, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R10 is H, alkyl (including lower alkyl), chlorine, bromine, or iodine;
    alternatively, R7 and R9, or R7 and R10 can come together to form a pi bond; and
    X is O, S, SO2 or CH2.
  • In a first preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(loweralkyl)-amino; (5) R10 is H; and (6) X is O, S, SO2, or CH2.
  • In a second preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R10 is H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O, S, SO2 or CH2.
  • In a third preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(loweralkyl)-amino; (5) R10 is H, alkyl (including lower alkyl), chlorine, bromine or iodine; and (6) X is O.
  • In a fourth preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R10 is H; and (6) X is O, S, SO2 or CH2.
  • In a fifth preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R10 is H, alkyl (including lower alkyl), chlorine, bromine or iodine; and (6) X is O.
  • In a sixth preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (5) R10 is H; and (6) X is O.
  • In a seventh preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R10 is H; and (6) X is O.
  • In an eighth preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(loweralkyl)-amino; (5) R10 is H, alkyl (including lower alkyl), chlorine, bromine or iodine; and (6) X is O, S, SO2, or CH2.
  • In a ninth preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R10 is H; and (6) X is O, S, SO2, or CH2.
  • In a tenth preferred subembodiment, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2; (5) R10 is H; and (6) X is O, S, SO2, or CH2.
  • In even more preferred subembodiments, a compound of Formula XVII, or its pharmaceutically acceptable salt or prodrug, is provided in which:
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is guanine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is cytosine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is thymine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is uracil; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is phosphate; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is ethyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is propyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is butyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is S;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is SO2; or
      • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R10 is hydrogen; and (6) X is CH2.
  • In an twelfth principal embodiment the invention provides a compound of Formula XVIII, or a pharmaceutically acceptable salt or prodrug thereof:
  • Figure US20180235993A1-20180823-C00025
  • wherein:
    Base is a purine or pyrimidine base as defined herein;
    R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate;
    R6 is hydrogen, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chloro, bromo, fluoro, iodo, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, lower alkylamino, or di(loweralkyl)amino;
    R8 is H, alkyl (including lower alkyl), chlorine, bromine or iodine;
    alternatively, R7 and R9, or R8 and R9 can come together to form a pi bond;
    X is O, S, SO2 or CH2.
  • In a first preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino or di(loweralkyl)amino; (5) R8 is H, alkyl (including lower alkyl), chlorine, bromine or iodine; and (6) X is O, S, SO2 or CH2.
  • In a second preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di-(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R8 is H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O, S, SO2 or CH2.
  • In a third preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(lower-alkyl)amino; (4) R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (5) R8 is H; and (6) X is O, S, SO2 or CH2.
  • In a fourth preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (5) R8 is H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O.
  • In a fifth preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R8 is H; and (6) X is O, S, SO2, or CH2.
  • In a sixth preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R8 is H, alkyl (including lower alkyl), chlorine, bromine, or iodine; and (6) X is O.
  • In a seventh preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (4) R7 and R9 are independently hydrogen, OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO2, amino, loweralkylamino, or di(loweralkyl)amino; (5) R8 is H; and (6) X is O.
  • In an eighth preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO2, amino, loweralkylamino or di(loweralkyl)amino; (4) R7 and R9 are independently OR2; (5) R8 is H; and (6) X is O, S, SO2 or CH2.
  • In a ninth preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2; (5) R8 is H; and (6) X is O, S, SO2, or CH2.
  • In a tenth preferred subembodiment, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which: (1) Base is a purine or pyrimidine base as defined herein; (2) R1 is independently H or phosphate; (3) R6 is alkyl; (4) R7 and R9 are independently OR2; (5) R8 is H; and (6) X is O.
  • In even more preferred subembodiments, a compound of Formula XVIII, or its pharmaceutically acceptable salt or prodrug, is provided in which:
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is O;
  • (1) Base is guanine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is O;
  • (1) Base is cytosine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is O;
  • (1) Base is thymine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is O;
  • (1) Base is uracil; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5)
  • R8 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is phosphate; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is ethyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is propyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is butyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is O;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is S;
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is SO2; or
  • (1) Base is adenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 is hydrogen; and (6) X is CH2.
  • The β-D- and β-L-nucleosides of this invention belong to a class of anti-flavivirus or pestivirus agents that may inhibit flavivirus or pestivirus polymerase activity. Nucleosides can be screened for their ability to inhibit flavivirus or pestivirus polymerase activity in vitro according to screening methods set forth more particularly herein. One can readily determine the spectrum of activity by evaluating the compound in the assays described herein or with another confirmatory assay.
  • In one embodiment the efficacy of the anti-flavivirus or pestivirus compound is measured according to the concentration of compound necessary to reduce the plaque number of the virus in vitro, according to methods set forth more particularly herein, by 50% (i.e. the compound's EC50). In preferred embodiments the compound exhibits an EC50 of less than 15 or 10 micromolar.
  • HCV is a member of the Flaviviridae family; however, now, HCV has been placed in a new monotypic genus, hepacivirus. Therefore, in one embodiment, the flavivirus or pestivirus is not HCV.
  • The active compound can be administered as any salt or prodrug that upon administration to the recipient is capable of providing directly or indirectly the parent compound, or that exhibits activity itself. Nonlimiting examples are the pharmaceutically acceptable salts (alternatively referred to as “physiologically acceptable salts”), and a compound, which has been alkylated or acylated at the 5′-position, or on the purine or pyrimidine base (a type of “pharmaceutically acceptable prodrug”). Further, the modifications can affect the biological activity of the compound, in some cases increasing the activity over the parent compound. This can easily be assessed by preparing the salt or prodrug and testing its antiviral activity according to the methods described herein, or other methods known to those skilled in the art.
  • II. Definitions
  • The term alkyl, as used herein, unless otherwise specified, refers to a saturated straight, branched, or cyclic, primary, secondary, or tertiary hydrocarbon of typically C1 to C10, and specifically includes methyl, trifluoromethyl, ethyl, propyl, isopropyl, cyclopropyl, butyl, isobutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 3-methylpentyl, 2,2-dimethylbutyl, and 2,3-dimethylbutyl. The term includes both substituted and unsubstituted alkyl groups. Moieties with which the alkyl group can be substituted are selected from the group consisting of 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, hereby incorporated by reference.
  • The term lower alkyl, as used herein, and unless otherwise specified, refers to a C1 to C4 saturated straight, branched, or if appropriate, a cyclic (for example, cyclopropyl) alkyl group, including both substituted and unsubstituted forms. Unless otherwise specifically stated in this application, when alkyl is a suitable moiety, lower alkyl is preferred. Similarly, when alkyl or lower alkyl is a suitable moiety, unsubstituted alkyl or lower alkyl is preferred.
  • The term alkylamino or arylamino refers to an amino group that has one or two alkyl or aryl substituents, respectively.
  • The term “protected” as used herein and unless otherwise defined refers to a group that is added to an oxygen, nitrogen, or phosphorus atom to prevent its further reaction or for other purposes. A wide variety of oxygen and nitrogen protecting groups are known to those skilled in the art of organic synthesis.
  • The term aryl, as used herein, and unless otherwise specified, refers to phenyl, biphenyl, or naphthyl, and preferably phenyl. The term includes both substituted and unsubstituted moieties. The aryl group can be substituted with one or more moieties selected from the group consisting of 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.
  • The term alkaryl or alkylaryl refers to an alkyl group with an aryl substituent. The term aralkyl or arylalkyl refers to an aryl group with an alkyl substituent.
  • The term halo, as used herein, includes chloro, bromo, iodo, and fluoro.
  • The term purine or pyrimidine base includes, but is not limited to, adenine, N6-alkylpurines, N6-acylpurines (wherein acyl is C(O)(alkyl, aryl, alkylaryl, or arylalkyl), N6-benzylpurine, N6-halopurine, N6-vinylpurine, N6-acetylenic purine, N6-acyl purine, N6-hydroxyalkyl purine, N6-thioalkyl purine, N2-alkylpurines, N2-alkyl-6-thiopurines, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6-azapyrimidine, including 6-azacytosine, 2- and/or 4-mercaptopyrmidine, uracil, 5-halouracil, including 5-fluorouracil, C5-alkylpyrimidines, C5-benzylpyrimidines, C5-halopyrimidines, C5-vinylpyrimidine, C5-acetylenic pyrimidine, C5-acyl pyrimidine, C5-hydroxyalkyl purine, C5-amidopyrimidine, C5-cyanopyrimidine, C5-nitropyrimidine, C5-aminopyrimidine, N2-alkylpurines, N2-alkyl-6-thiopurines, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and pyrazolo-pyrimidinyl. Purine bases include, but are not limited to, guanine, adenine, hypoxanthine, 2,6-diaminopurine, 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, dimethylhexylsilyl, t-butyldimethylsilyl and t-butyldiphenylsilyl, trityl, alkyl groups, and acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl. Alternatively, the purine or pyrimidine base can optionally substituted such that it forms a viable prodrug, which can be cleaved in vivo. Examples of appropriate substituents include acyl moiety, an amine or cyclopropyl (e.g., 2-amino, 2,6-diamino or cyclopropyl guanosine).
  • The term acyl refers to a carboxylic acid ester in which the non-carbonyl moiety of the ester group is selected from straight, branched, or cyclic alkyl or lower alkyl, alkoxyalkyl including methoxymethyl, aralkyl including benzyl, aryloxyalkyl such as phenoxymethyl, aryl including phenyl optionally substituted with halogen, C1 to C4 alkyl or C1 to C4 alkoxy, sulfonate esters such as alkyl or aralkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxytrityl, substituted benzyl, trialkylsilyl (e.g. dimethyl-t-butylsilyl) or diphenylmethylsilyl. Aryl groups in the esters optimally comprise a phenyl group. The term “lower acyl” refers to an acyl group in which the non-carbonyl moiety is lower alkyl.
  • As used herein, the term “substantially free of” or “substantially in the absence of” refers to a nucleoside composition that includes at least 85 or 90% by weight, preferably 95% to 98% by weight, and even more preferably 99% to 100% by weight, of the designated enantiomer of that nucleoside. In a preferred embodiment, in the methods and compounds of this invention, the compounds are substantially free of enantiomers.
  • Similarly, the term “isolated” refers to a nucleoside composition that includes at least 85 or 90% by weight, preferably 95% to 98% by weight, and even more preferably 99% to 100% by weight, of the nucleoside, the remainder comprising other chemical species or enantiomers.
  • The term “independently” is used herein to indicate that the variable, which is independently applied, varies independently from application to application. Thus, in a compound such as R″XYR″, wherein R″ is “independently carbon or nitrogen,” both R″ can be carbon, both R″ can be nitrogen, or one R″ can be carbon and the other R″ nitrogen.
  • The term host, as used herein, refers to an unicellular or multicellular organism in which the virus can replicate, including cell lines and animals, and preferably a human. Alternatively, the host can be carrying a part of the flavivirus or pestivirus genome, whose replication or function can be altered by the compounds of the present invention. The term host specifically refers to infected cells, cells transfected with all or part of the flavivirus or pestivirus 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 term “pharmaceutically acceptable salt or prodrug” is used throughout the specification to describe any pharmaceutically acceptable form (such as an ester, phosphate ester, salt of an ester or a related group) of a nucleoside compound which, upon administration to a patient, provides the nucleoside compound. Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable inorganic or organic bases and acids. Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium and magnesium, among numerous other acids well known in the pharmaceutical art. Pharmaceutically acceptable prodrugs refer to a compound that is metabolized, for example hydrolyzed or oxidized, in the host to form the compound of the present invention. Typical examples of prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound. Prodrugs include compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, dephosphorylated to produce the active compound. The compounds of this invention possess antiviral activity against flavivirus or pestivirus, or are metabolized to a compound that exhibits such activity.
  • III. Nucleotide Salt or Prodrug Formulations
  • In cases where compounds are sufficiently basic or acidic to form stable nontoxic acid or base salts, administration of the compound as a pharmaceutically acceptable salt may be appropriate. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids, which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, α-ketoglutarate, and α-glycerophosphate. Suitable inorganic salts may also be formed, including, sulfate, nitrate, bicarbonate, and carbonate salts.
  • Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion. Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
  • Any of the nucleosides described herein can be administered as a nucleotide prodrug to increase the activity, bioavailability, stability or otherwise alter the properties of the nucleoside. A number of nucleotide prodrug ligands are known. In general, alkylation, acylation or other lipophilic modification of the mono, di or triphosphate of the nucleoside will increase the stability of the nucleotide. Examples of substituent groups that can replace one or more hydrogens on the phosphate moiety are alkyl, aryl, steroids, carbohydrates, including sugars, 1,2-diacylglycerol and alcohols. Many are described in R. Jones and N. Bischofberger, Antiviral Research, 27 (1995) 1-17. Any of these can be used in combination with the disclosed nucleosides to achieve a desired effect.
  • The active nucleoside can also be provided as a 5′-phosphoether lipid or a 5′-ether lipid, as disclosed in the following references, which are incorporated by reference herein: Kucera, L. S., N. Iyer, E. Leake, A. Raben, Modest E. K., D. L. W., and C. Piantadosi, “Novel membrane-interactive ether lipid analogs that inhibit infectious HIV-1 production and induce defective virus formation,” AIDS Res. Hum. Retro Viruses, 1990, 6, 491-501; Piantadosi, C., J. Marasco C. J., S. L. Morris-Natschke, K. L. Meyer, F. Gumus, J. R. Surles, K. S. Ishaq, L. S. Kucera, N. Iyer, C. A. Wallen, S. Piantadosi, and E. J. Modest, “Synthesis and evaluation of novel ether lipid nucleoside conjugates for anti-HIV activity,” J. Med. Chem., 1991, 34, 1408-1414; Hosteller, K. Y., D. D. Richman, D. A. Carson, L. M. Stuhmiller, G. M. T. van Wijk, and H. van den Bosch, “Greatly enhanced inhibition of human immunodeficiency virus type 1 replication in CEM and HT4-6C cells by 3′-deoxythymidine diphosphate dimyristoylglycerol, a lipid prodrug of 3,-deoxythymidine,” Antimicrob. Agents Chemother., 1992, 36, 2025-2029; Hosetler, K. Y., L. M. Stuhmiller, H. B. Lenting, H. van den Bosch, and D. D. Richman, “Synthesis and antiretroviral activity of phospholipid analogs of azidothymidine and other antiviral nucleosides.” J Biol. Chem., 1990, 265, 61127.
  • Nonlimiting examples of U.S. patents that disclose suitable lipophilic substituents that can be covalently incorporated into the nucleoside, preferably at the 5′-OH position of the nucleoside or lipophilic preparations, include U.S. Pat. No. 5,149,794 (Sep. 22, 1992, Yatvin et al.); U.S. Pat. No. 5,194,654 (Mar. 16, 1993, Hostetler et al., U.S. Pat. No. 5,223,263 (Jun. 29, 1993, Hostetler et al.); U.S. Pat. No. 5,256,641 (Oct. 26, 1993, Yatvin et al.); U.S. Pat. No. 5,411,947 (May 2, 1995, Hostetler et al.); U.S. Pat. No. 5,463,092 (Oct. 31, 1995, Hostetler et al.); U.S. Pat. No. 5,543,389 (Aug. 6, 1996, Yatvin et al.); U.S. Pat. No. 5,543,390 (Aug. 6, 1996, Yatvin et al.); U.S. Pat. No. 5,543,391 (Aug. 6, 1996, Yatvin et al.); and U.S. Pat. No. 5,554,728 (Sep. 10, 1996; Basava et al.), all of which are incorporated herein by reference. Foreign patent applications that disclose lipophilic substituents that can be attached to the nucleosides of the present invention, or lipophilic preparations, include WO 89/02733, WO 90/00555, WO 91/16920, WO 91/18914, WO 93/00910, WO 94/26273, WO 96/15132, EP 0 350 287, EP 93917054.4, and WO 91/19721.
  • IV. Combination and Alternation Therapy
  • It has been recognized that drug-resistant variants of viruses can emerge after prolonged treatment with an antiviral agent. Drug resistance most typically occurs by mutation of a gene that encodes for an enzyme used in viral replication. The efficacy of a drug against flavivirus or pestivirus infection can be prolonged, augmented, or restored by administering the compound in combination or alternation with a second, and perhaps third, antiviral compound that induces a different mutation from that caused by the principle drug. Alternatively, the pharmacokinetics, biodistribution or other parameter of the drug can be altered by such combination or alternation therapy. In general, combination therapy is typically preferred over alternation therapy because it induces multiple simultaneous stresses on the virus.
  • Nonlimiting examples of antiviral agents that can be used in combination or alternation with the compounds disclosed herein include:
  • (1) an interferon and/or ribavirin (Battaglia, A. M. et al., Ann. Pharmacother. 34:487-494, 2000); Berenguer, M. et al. Antivir. Ther. 3(Suppl. 3):125-136, 1998);
  • (2) Substrate-based NS3 protease inhibitors (Attwood et al., Antiviral peptide derivatives, PCT WO 98/22496, 1998; Attwood et al., Antiviral Chemistry and Chemotherapy 10.259-273, 1999; Attwood et al., Preparation and use of amino acid derivatives as anti-viral agents, German Patent Publication DE 19914474; Tung et al. Inhibitors of serine proteases, particularly hepatitis C virus NS3 protease, PCT WO 98/17679), including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an electrophile such as a boronic acid or phosphonate. Llinas-Brunet et al, Hepatitis C inhibitor peptide analogues, PCT WO 99/07734.
  • (3) Non-substrate-based inhibitors such as 2,4,6-trihydroxy-3-nitro-benzamide derivatives(Sudo K. et al., Biochemical and Biophysical Research Communications, 238:643-647, 1997; Sudo K. et al. Antiviral Chemistry and Chemotherapy 9:186, 1998), including RD3-4082 and RD3-4078, the former substituted on the amide with a 14 carbon chain and the latter processing a para-phenoxyphenyl group;
  • (4) Thiazolidine derivatives which show relevant inhibition in a reverse-phase HPLC assay with an NS3/4A fusion protein and NS5A/5B substrate (Sudo K. et al., Antiviral Research 32:9-18, 1996), especially compound RD-1-6250, possessing a fused cinnamoyl moiety substituted with a long alkyl chain, RD4 6205 and RD4 6193;
  • (5) Thiazolidines and benzanilides identified in Kakiuchi N. et al. J. EBS Letters 421:217-220; Takeshita N. et al. Analytical Biochemistry 247:242-246, 1997;
  • (6) A phenan-threnequinone possessing activity against protease in a SDS-PAGE and autoradiography assay isolated from the fermentation culture broth of Streptomyces sp., Sch 68631 (Chu M. et al., Tetrahedron Letters 37:7229-7232, 1996), and Sch 351633, isolated from the fungus Penicillium griscofuluum, which demonstrates activity in a scintillation proximity assay (Chu M. et al., Bioorganic and Medicinal Chemistry Letters 9:1949-1952);
  • (7) Selective NS3 inhibitors based on the macromolecule elgin c, isolated from leech (Qasim M. A. et al., Biochemistry 36:1598-1607, 1997);
  • (8) Helicase inhibitors (Diana G. D. et al., Compounds, compositions and methods for treatment of hepatitis C, U.S. Pat. No. 5,633,358; Diana G. D. et al., Piperidine derivatives, pharmaceutical compositions thereof and their use in the treatment of hepatitis C, PCT WO 97/36554);
  • (9) Polymerase inhibitors such as nucleotide analogues, gliotoxin (Ferrari R. et al. Journal of Virology 73:1649-1654, 1999), and the natural product cerulenin (Lohmann V. et al., Virology 249:108-118, 1998);
  • (10) Antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5′ non-coding region (NCR) of the virus (Alt M. et al., Hepatology 22:707-717, 1995), or nucleotides 326-348 comprising the 3′ end of the NCR and nucleotides 371-388 located in the core coding region of the IICV RNA (Alt M. et al., Archives of Virology 142:589-599, 1997; Galderisi U. et al., Journal of Cellular Physiology 181:251-257, 1999);
  • (11) Inhibitors of IRES-dependent translation (Ikeda N et al., Agent for the prevention and treatment of hepatitis C, Japanese Patent Publication JP-08268890; Kai Y. et al. Prevention and treatment of viral diseases, Japanese Patent Publication JP-10101591);
  • (12) Nuclease-resistant ribozymes. (Maccjak D. J. et al., Hepatology 30 abstract 995, 1999); and
  • (13) Other miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S. Pat. No. 6,034,134 to Gold et al.), alkyl lipids (U.S. Pat. No. 5,922,757 to Chojkier et al.), vitamin E and other antioxidants (U.S. Pat. No. 5,922,757 to Chojkier et al.), squalene, amantadine, bile acids (U.S. Pat. No. 5,846,964 to Ozeki et al.), N-(phosphonoacetyl)-L-aspartic acid, (U.S. Pat. No. 5,830,905 to Diana et al.), benzenedicarboxamides (U.S. Pat. No. 5,633,388 to Diana et al.), polyadenylic acid derivatives (U.S. Pat. No. 5,496,546 to Wang et al.), 2′,3′-dideoxyinosine (U.S. Pat. No. 5,026,687 to Yarchoan et al.), and benzimidazoles (U.S. Pat. No. 5,891,874 to Colacino et al.).
  • V. Pharmaceutical Compositions
  • Host, including humans, infected with flavivirus or pestivirus, or a gene fragment thereof can be treated by administering to the patient an effective amount of the active compound or a pharmaceutically acceptable prodrug or salt thereof in the presence of a pharmaceutically acceptable carrier or diluent. The active materials can be administered by any appropriate route, for example, orally, parenterally, intravenously, intradermally, subcutaneously, or topically, in liquid or solid form.
  • A preferred dose of the compound for flavivirus or pestivirus infection will be in the range from about 1 to 50 mg/kg, preferably 1 to 20 mg/kg, of body weight per day, more generally 0.1 to about 100 mg per kilogram body weight of the recipient per day. The effective dosage range of the pharmaceutically acceptable salts and prodrugs can be calculated based on the weight of the parent nucleoside to be delivered. If the salt or prodrug exhibits activity in itself, the effective dosage can be estimated as above using the weight of the salt or prodrug, or by other means known to those skilled in the art.
  • The compound is conveniently administered in unit any suitable dosage form, including but not limited to one containing 7 to 3000 mg, preferably 70 to 1400 mg of active ingredient per unit dosage form. A oral dosage of 50-1000 mg is usually convenient.
  • Ideally the active ingredient should be administered to achieve peak plasma concentrations of the active compound of from about 0.2 to 70 μM, preferably about 1.0 to 10 μM. This may be achieved, for example, by the intravenous injection of a 0.1 to 5% solution of the active ingredient, optionally in saline, or administered as a bolus of the active ingredient.
  • The concentration of active compound in the drug composition 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 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, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
  • A preferred mode of administration of the active compound is oral. Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or other enteric agents.
  • The compound can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • The compound or a pharmaceutically acceptable prodrug or salts thereof can also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, such as antibiotics, antifungals, anti-inflammatories, or other antivirals, including other nucleoside compounds. Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • If administered intravenously, preferred carriers are physiological saline or phosphate buffered saline (PBS).
  • In a preferred embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) are also preferred as pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811 (which is incorporated herein by reference in its entirety). For example, liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the active compound or its monophosphate, diphosphate, and/or triphosphate derivatives is then introduced into the container. The container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension.
  • VI. Processes for the Preparation of Active Compounds
  • The nucleosides of the present invention can be synthesized by any means known in the art. In particular, the synthesis of the present nucleosides can be achieved by either alkylating the appropriately modified sugar, followed by glycosylation or glycosylation followed by alkylation of the nucleoside. The following non-limiting embodiments illustrate some general methodology to obtain the nucleosides of the present invention.
  • A. General Synthesis of 1′-C-Branched Nucleosides
  • 1′-C-Branched ribonucleosides of the following structure:
  • Figure US20180235993A1-20180823-C00026
  • wherein BASE is a purine or pyrimidine base as defined herein;
    R7 and R9 are independently hydrogen, OR2, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine;
    alternatively, R7 and R9, R7 and R10, R8 and R9, or R8 and R10 can come together to form a pi bond;
    R1 and R2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate;
    R6 is an alkyl, halogeno-alkyl (i.e. CF3), alkenyl, or alkynyl (i.e. allyl); and
    X is O, S, SO2 or CH2
    can be prepared by one of the following general methods.
    1) Modification from the Lactone
  • The key starting material for this process is an appropriately substituted lactone. The lactone can be purchased or can be prepared by any known means including standard epimerization, substitution and cyclization techniques. The lactone can be optionally protected with a suitable protecting group, preferably with an acyl or silyl group, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991. The protected lactone can then be coupled with a suitable coupling agent, such as an organometallic carbon nucleophile, such as a Grignard reagent, an organolithium, lithium dialkylcopper or R6—SiMe3 in TBAF with the appropriate non-protic solvent at a suitable temperature, to give the 1′-alkylated sugar.
  • The optionally activated sugar can then be coupled to the BASE by methods well known to those skilled in the art, as taught by Townsend Chemistry of Nucleosides and Nucleotides, Plenum Press, 1994. For example, an acylated sugar can be coupled to a silylated base with a lewis acid, such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperature.
  • Subsequently, the nucleoside can be deprotected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • In a particular embodiment, the 1′-C-branched ribonucleoside is desired. The synthesis of a ribonucleoside is shown in Scheme 1. Alternatively, deoxyribo-nucleoside is desired. To obtain these nucleosides, the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent. Optionally, the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • Figure US20180235993A1-20180823-C00027
  • 2. Alternative Method for the Preparation of 1′-C-Branched Nucleosides
  • The key starting material for this process is an appropriately substituted hexose. The hexose can be purchased or can be prepared by any known means including standard epimerization (e.g. via alkaline treatment), substitution and coupling techniques. The hexose can be selectively protected to give the appropriate hexa-furanose, as taught by Townsend Chemistry of Nucleosides and Nucleotides, Plenum Press, 1994.
  • The 1′-hydroxyl can be optionally activated to a suitable leaving group such as an acyl group or a halogen via acylation or halogenation, respectively. The optionally activated sugar can then be coupled to the BASE by methods well known to those skilled in the art, as taught by Townsend Chemistry of Nucleosides and Nucleotides, Plenum Press, 1994. For example, an acylated sugar can be coupled to a silylated base with a lewis acid, such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperature. Alternatively, a halo-sugar can be coupled to a silylated base with the presence of trimethylsilyltriflate.
  • The 1′-CH2—OH, if protected, can be selectively deprotected by methods well known in the art. The resultant primary hydroxyl can be functionalized to yield various C-branched nucleosides. For example, the primary hydroxyl can be reduced to give the methyl, using a suitable reducing agent. Alternatively, the hydroxyl can be activated prior to reduction to facilitate the reaction; i.e. via the Barton reduction. In an alternate embodiment, the primary hydroxyl can be oxidized to the aldehyde, then coupled with a carbon nucleophile, such as a Grignard reagent, an organolithium, lithium dialkylcopper or R6—SiMe3 in TBAF with the appropriate non-protic solvent at a suitable temperature.
  • In a particular embodiment, the 1′-C-branched ribonucleoside is desired. The synthesis of a ribonucleoside is shown in Scheme 2. Alternatively, deoxyribo-nucleoside is desired. To obtain these nucleosides, the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent. Optionally, the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • Figure US20180235993A1-20180823-C00028
  • In addition, the L-enantiomers corresponding to the compounds of the invention can be prepared following the same general methods (1 or 2), beginning with the corresponding L-sugar or nucleoside L-enantiomer as starting material.
  • B. General Synthesis of 2′-C-Branched Nucleosides
  • 2′-C-Branched ribonucleosides of the following structure:
  • Figure US20180235993A1-20180823-C00029
  • wherein BASE is a purine or pyrimidine base as defined herein;
    R7 and R9 are independently hydrogen, OR2, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R10 is alkyl (including lower alkyl), chlorine, bromine or iodine;
    alternatively, R7 and R9, or R7 and R10 can come together to form a pi bond;
    R1 and R2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate;
    R6 is an alkyl, halogeno-alkyl (i.e. CF3), alkenyl, or alkynyl (i.e. allyl); and
    X is O, S, SO2 or CH2
    can be prepared by one of the following general methods.
    1. Glycosylation of the Nucleobase with an Appropriately Modified Sugar
  • The key starting material for this process is an appropriately substituted sugar with a 2′-OH and 2′-H, with the appropriate leaving group (LG), for example an acyl group or a halogen. The sugar can be purchased or can be prepared by any known means including standard epimerization, substitution, oxidation and reduction techniques. The substituted sugar can then be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperature to yield the 2′-modified sugar. Possible oxidizing agents are Jones reagent (a mixture of chromic acid and sulfuric acid), Collins's reagent (dipyridine Cr(VI) oxide, Corey's reagent (pyridinium chlorochromate), pyridinium dichromate, acid dichromate, potassium permanganate, MnO2, ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H2O2-ammonium molybdate, NaBrO2—CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (aluminum t-butoxide with another ketone) and N-bromosuccinimide.
  • Then coupling of an organometallic carbon nucleophile, such as a Grignard reagent, an organolithium, lithium dialkylcopper or R6—SiMe3 in TBAF with the ketone with the appropriate non-protic solvent at a suitable temperature, yields the 2′-alkylated sugar. The alkylated sugar can be optionally protected with a suitable protecting group, preferably with an acyl or silyl group, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • The optionally protected sugar can then be coupled to the BASE by methods well known to those skilled in the art, as taught by Townsend Chemistry of Nucleosides and Nucleotides, Plenum Press, 1994. For example, an acylated sugar can be coupled to a silylated base with a lewis acid, such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperature. Alternatively, a halo-sugar can be coupled to a silylated base with the presence of trimethylsilyltriflate.
  • Subsequently, the nucleoside can be deprotected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • In a particular embodiment, the 2′-C-branched ribonucleoside is desired. The synthesis of a ribonucleoside is shown in Scheme 3. Alternatively, deoxyribo-nucleoside is desired. To obtain these nucleosides, the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent. Optionally, the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • Figure US20180235993A1-20180823-C00030
  • 2. Modification of a Pre Formed Nucleoside
  • The key starting material for this process is an appropriately substituted nucleoside with a 2′-OH and 2′-H. The nucleoside can be purchased or can be prepared by any known means including standard coupling techniques. The nucleoside can be optionally protected with suitable protecting groups, preferably with acyl or silyl groups, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • The appropriately protected nucleoside can then be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperature to yield the 2′-modified sugar. Possible oxidizing agents are Jones reagent (a mixture of chromic acid and sulfuric acid), Collins's reagent (dipyridine Cr(VI) oxide, Corey's reagent (pyridinium chlorochromate), pyridinium dichromate, acid dichromate, potassium permanganate, MnO2, ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H2O2-ammonium molybdate, NaBrO2—CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (aluminum t-butoxide with another ketone) and N-bromosuccinimide.
  • Subsequently, the nucleoside can be deprotected by methods well known to those skilled in the art, as taught by GreeneGreene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • In a particular embodiment, the 2′-C-branched ribonucleoside is desired. The synthesis of a ribonucleoside is shown in Scheme 4. Alternatively, deoxyribo-nucleoside is desired. To obtain these nucleosides, the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent. Optionally, the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • Figure US20180235993A1-20180823-C00031
  • In another embodiment of the invention, the L-enantiomers are desired. Therefore, the L-enantiomers can be corresponding to the compounds of the invention can be prepared following the same foregoing general methods, beginning with the corresponding L-sugar or nucleoside L-enantiomer as starting material.
  • C. General Synthesis of 3′-C-Branched Nucleosides
  • 3′-C-Branched ribonucleosides of the following structure:
  • Figure US20180235993A1-20180823-C00032
  • wherein BASE is a purine or pyrimidine base as defined herein;
    R7 and R9 are independently hydrogen, OR2, hydroxy, alkyl (including lower alkyl), azido, cyano, alkenyl, alkynyl, Br-vinyl, —C(O)O(alkyl), —C(O)O(lower alkyl), —O(acyl), —O(lower acyl), —O(alkyl), —O(lower alkyl), —O(alkenyl), chlorine, bromine, iodine, NO2, NH2, —NH(lower alkyl), —NH(acyl), —N(lower alkyl)2, —N(acyl)2;
    R8 is H, alkyl (including lower alkyl), chlorine, bromine or iodine;
    alternatively, R7 and R9, or R8 and R9 can come together to form a pi bond;
    R1 and R2 are independently H; phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester including alkyl or arylalkyl sulfonyl including methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as described in the definition of aryl given herein; a lipid, including a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is independently H or phosphate;
    R6 is an alkyl, halogeno-alkyl (i.e. CF3), alkenyl, or alkynyl (i.e. allyl); and
    X is O, S, SO2 or CH2
    can be prepared by one of the following general methods.
    1 Glycosylation of the Nucleobase with an Appropriately Modified Sugar
  • The key starting material for this process is an appropriately substituted sugar with a 3′-OH and 3′-H, with the appropriate leaving group (LG), for example an acyl group or a halogen. The sugar can be purchased or can be prepared by any known means including standard epimerization, substitution, oxidation and reduction techniques. The substituted sugar can then be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperature to yield the 3′-modified sugar. Possible oxidizing agents are Jones reagent (a mixture of chromic acid and sulfuric acid), Collins's reagent (dipyridine Cr(VI) oxide, Corey's reagent (pyridinium chlorochromate), pyridinium dichromate, acid dichromate, potassium permanganate, MnO2, ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H2O2-ammonium molybdate, NaBrO2—CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (aluminum t-butoxide with another ketone) and N-bromosuccinimide.
  • Then coupling of an organometallic carbon nucleophile, such as a Grignard reagent, an organolithium, lithium dialkylcopper or R6—SiMe3 in TBAF with the ketone with the appropriate non-protic solvent at a suitable temperature, yields the 3′-C-branched sugar. The 3′-C-branched sugar can be optionally protected with a suitable protecting group, preferably with an acyl or silyl group, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • The optionally protected sugar can then be coupled to the BASE by methods well known to those skilled in the art, as taught by Townsend Chemistry of Nucleosides and Nucleotides, Plenum Press, 1994. For example, an acylated sugar can be coupled to a silylated base with a Lewis acid, such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperature. Alternatively, a halo-sugar can be coupled to a silylated base with the presence of trimethylsilyltriflate.
  • Subsequently, the nucleoside can be deprotected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • In a particular embodiment, the 3′-C-branched ribonucleoside is desired. The synthesis of a ribonucleoside is shown in Scheme 5. Alternatively, deoxyribo-nucleoside is desired. To obtain these nucleosides, the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent. Optionally, the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • Figure US20180235993A1-20180823-C00033
  • 2. Modification of a Pre Formed Nucleoside
  • The key starting material for this process is an appropriately substituted nucleoside with a 3′-OH and 3′-H. The nucleoside can be purchased or can be prepared by any known means including standard coupling techniques. The nucleoside can be optionally protected with suitable protecting groups, preferably with acyl or silyl groups, by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • The appropriately protected nucleoside can then be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperature to yield the 2′-modified sugar. Possible oxidizing agents are Jones reagent (a mixture of chromic acid and sulfuric acid), Collins's reagent (dipyridine Cr(VI) oxide, Corey's reagent (pyridinium chlorochromate), pyridinium dichromate, acid dichromate, potassium permanganate, MnO2, ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H2O2-ammonium molybdate, NaBrO2—CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (aluminum t-butoxide with another ketone) and N-bromosuccinimide.
  • Subsequently, the nucleoside can be deprotected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • In a particular embodiment, the 3′-C-branched ribonucleoside is desired. The synthesis of a ribonucleoside is shown in Scheme 6. Alternatively, deoxyribo-nucleoside is desired. To obtain these nucleosides, the formed ribonucleoside can optionally be protected by methods well known to those skilled in the art, as taught by Greene et al. Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, and then the 2′-OH can be reduced with a suitable reducing agent. Optionally, the 2′-hydroxyl can be activated to facilitate reduction; i.e. via the Barton reduction.
  • Figure US20180235993A1-20180823-C00034
  • In another embodiment of the invention, the L-enantiomers are desired. Therefore, the L-enantiomers can be corresponding to the compounds of the invention can be prepared following the same foregoing general methods, beginning with the corresponding L-sugar or nucleoside L-enantiomer as starting material.
  • Examples Example 1: Preparation of 1′-C-methylriboadenine via 6-amino-9-(1-deoxy-β-D-psicofuranosyl)purine
  • The title compound could also be prepared according to a published procedure (J. Farkas, and F. Sorm, “Nucleic acid components and their analogues. XCIV. Synthesis of 6-amino-9-(1-deoxy-β-D-psicofuranosyl)purine” Collect. Czech. Chem. Commun. 1967, 32, 2663-2667; J. Farkas”, Collect. Czech. Chem. Commun. 1966, 31, 1535) (Scheme 7).
  • Figure US20180235993A1-20180823-C00035
  • In a similar manner, but using the appropriate sugar and pyrimidine or purine bases, the following nucleosides of Formula I are prepared.
  • Figure US20180235993A1-20180823-C00036
  • wherein:
  • R1 R2 R3 X1 X2 Y
    H H H H H H
    H H H H H NH2
    H H H H H NH-cyclopropyl
    H H H H H NH-methyl
    H H H H H NH-ethyl
    H H H H H NH-acetyl
    H H H H H OH
    H H H H H OMe
    H H H H H OEt
    H H H H H O-cyclopropyl
    H H H H H O-acetyl
    H H H H H SH
    H H H H H SMe
    H H H H H SEt
    H H H H H S-cyclopropyl
    H H H H H F
    H H H H H Cl
    H H H H H Br
    H H H H H I
    monophosphate H H H H NH2
    monophosphate H H H H NH-acetyl
    monophosphate H H H H NH-cyclopropyl
    monophosphate H H H H NH-methyl
    monophosphate H H H H NH-ethyl
    monophosphate H H H H OH
    monophosphate H H H H O-acetyl
    monophosphate H H H H OMe
    monophosphate H H H H OEt
    monophosphate H H H H O-cyclopropyl
    monophosphate H H H H SH
    monophosphate H H H H SMe
    monophosphate H H H H SEt
    monophosphate H H H H S-cyclopropyl
    monophosphate H H H H F
    monophosphate H H H H Cl
    monophosphate H H H H Br
    monophosphate H H H H I
    diphosphate H H H H NH2
    diphosphate H H H H NH-acetyl
    diphosphate H H H H NH-cyclopropyl
    diphosphate H H H H NH-methyl
    diphosphate H H H H NH-ethyl
    diphosphate H H H H OH
    diphosphate H H H H O-acetyl
    diphosphate H H H H OMe
    diphosphate H H H H OEt
    diphosphate H H H H O-cyclopropyl
    diphosphate H H H H SH
    diphosphate H H H H SMe
    diphosphate H H H H SEt
    diphosphate H H H H S-cyclopropyl
    diphosphate H H H H F
    diphosphate H H H H Cl
    diphosphate H H H H Br
    diphosphate H H H H I
    triphosphate H H H H NH2
    triphosphate H H H H NH-acetyl
    triphosphate H H H H NH-cyclopropyl
    triphosphate H H H H NH-methyl
    triphosphate H H H H NH-ethyl
    triphosphate H H H H OH
    triphosphate H H H H OMe
    triphosphate H H H H OEt
    triphosphate H H H H O-cyclopropyl
    triphosphate H H H H O-acetyl
    triphosphate H H H H SH
    triphosphate H H H H SMe
    triphosphate H H H H SEt
    triphosphate H H H H S-cyclopropyl
    triphosphate H H H H F
    triphosphate H H H H Cl
    triphosphate H H H H Br
    triphosphate H H H H I
    monophosphate monophosphate monophosphate H H NH2
    monophosphate monophosphate monophosphate H H NH-cyclopropyl
    monophosphate monophosphate monophosphate H H OH
    monophosphate monophosphate monophosphate H H F
    monophosphate monophosphate monophosphate H H Cl
    diphosphate diphosphate diphosphate H H NH2
    diphosphate diphosphate diphosphate H H NH-cyclopropyl
    diphosphate diphosphate diphosphate H H OH
    diphosphate diphosphate diphosphate H H F
    diphosphate diphosphate diphosphate H H Cl
    triphosphate triphosphate triphosphate H H NH2
    triphosphate triphosphate triphosphate H H NH-cyclopropyl
    triphosphate triphosphate triphosphate H H OH
    triphosphate triphosphate triphosphate H H F
    triphosphate triphosphate triphosphate H H Cl
    H H H F H NH2
    H H H F H NH-cyclopropyl
    H H H F H OH
    H H H F H F
    H H H F H Cl
    H H H Cl H NH2
    H H H Cl H NH-cyclopropyl
    H H H Cl H OH
    H H H Cl H F
    H H H Cl H Cl
    H H H Br H NH2
    H H H Br H NH-cyclopropyl
    H H H Br H OH
    H H H Br H F
    H H H Br H Cl
    H H H NH2 H NH2
    H H H NH2 H NH-cyclopropyl
    H H H NH2 H OH
    H H H NH2 H F
    H H H NH2 H Cl
    H H H SH H NH2
    H H H SH H NH-cyclopropyl
    H H H SH H OH
    H H H SH H F
    H H H SH H Cl
    acetyl H H H H NH2
    acetyl H H H H NH-cyclopropyl
    acetyl H H H H OH
    acetyl H H H H F
    acetyl H H H H Cl
    acetyl H H F H NH2
    acetyl H H F H NH-cyclopropyl
    acetyl H H F H OH
    acetyl H H F H F
    acetyl H H F H Cl
    H acetyl acetyl H H NH2
    H acetyl acetyl H H NH-cyclopropyl
    H acetyl acetyl H H OH
    H acetyl acetyl H H F
    H acetyl acetyl H H Cl
    acetyl acetyl acetyl H H NH2
    acetyl acetyl acetyl H H NH-cyclopropyl
    acetyl acetyl acetyl H H OH
    acetyl acetyl acetyl H H F
    acetyl acetyl acetyl H H Cl
    monophosphate acetyl acetyl H H NH2
    monophosphate acetyl acetyl H H NH-cyclopropyl
    monophosphate acetyl acetyl H H OH
    monophosphate acetyl acetyl H H F
    monophosphate acetyl acetyl H H Cl
    diphosphate acetyl acetyl H H NH2
    diphosphate acetyl acetyl H H NH-cyclopropyl
    diphosphate acetyl acetyl H H OH
    diphosphate acetyl acetyl H H F
    diphosphate acetyl acetyl H H Cl
    triphosphate acetyl acetyl H H NH2
    triphosphate acetyl acetyl H H NH-cyclopropyl
    triphosphate acetyl acetyl H H OH
    triphosphate acetyl acetyl H H F
    triphosphate acetyl acetyl H H Cl
    H H H H NH2 H
    H H H H NH2 NH2
    H H H H NH2 NH-cyclopropyl
    H H H H NH2 NH-methyl
    H H H H NH2 NH-ethyl
    H H H H NH2 NH-acetyl
    H H H H NH2 OH
    H H H H NH2 OMe
    H H H H NH2 OEt
    H H H H NH2 O-cyclopropyl
    H H H H NH2 O-acetyl
    H H H H NH2 SH
    H H H H NH2 SMe
    H H H H NH2 SEt
    H H H H NH2 S-cyclopropyl
    H H H H NH2 F
    H H H H NH2 Cl
    H H H H NH2 Br
    H H H H NH2 I
    monophosphate H H H NH2 NH2
    monophosphate H H H NH2 NH-acetyl
    monophosphate H H H NH2 NH-cyclopropyl
    monophosphate H H H NH2 NH-methyl
    monophosphate H H H NH2 NH-ethyl
    monophosphate H H H NH2 OH
    monophosphate H H H NH2 O-acetyl
    monophosphate H H H NH2 OMe
    monophosphate H H H NH2 OEt
    monophosphate H H H NH2 O-cyclopropyl
    monophosphate H H H NH2 SH
    monophosphate H H H NH2 SMe
    monophosphate H H H NH2 SEt
    monophosphate H H H NH2 S-cyclopropyl
    monophosphate H H H NH2 F
    monophosphate H H H NH2 Cl
    monophosphate H H H NH2 Br
    monophosphate H H H NH2 I
    diphosphate H H H NH2 NH2
    diphosphate H H H NH2 NH-acetyl
    diphosphate H H H NH2 NH-cyclopropyl
    diphosphate H H H NH2 NH-methyl
    diphosphate H H H NH2 NH-ethyl
    diphosphate H H H NH2 OH
    diphosphate H H H NH2 O-acetyl
    diphosphate H H H NH2 OMe
    diphosphate H H H NH2 OEt
    diphosphate H H H NH2 O-cyclopropyl
    diphosphate H H H NH2 SH
    diphosphate H H H NH2 SMe
    diphosphate H H H NH2 SEt
    diphosphate H H H NH2 S-cyclopropyl
    diphosphate H H H NH2 F
    diphosphate H H H NH2 Cl
    diphosphate H H H NH2 Br
    diphosphate H H H NH2 I
    triphosphate H H H NH2 NH2
    triphosphate H H H NH2 NH-acetyl
    triphosphate H H H NH2 NH-cyclopropyl
    triphosphate H H H NH2 NH-methyl
    triphosphate H H H NH2 NH-ethyl
    triphosphate H H H NH2 OH
    triphosphate H H H NH2 OMe
    triphosphate H H H NH2 OEt
    triphosphate H H H NH2 O-cyclopropyl
    triphosphate H H H NH2 O-acetyl
    triphosphate H H H NH2 SH
    triphosphate H H H NH2 SMe
    triphosphate H H H NH2 SEt
    triphosphate H H H NH2 S-cyclopropyl
    triphosphate H H H NH2 F
    triphosphate H H H NH2 Cl
    triphosphate H H H NH2 Br
    triphosphate H H H NH2 I
    monophosphate monophosphate monophosphate H NH2 NH2
    monophosphate monophosphate monophosphate H NH2 NH-cyclopropyl
    monophosphate monophosphate monophosphate H NH2 OH
    monophosphate monophosphate monophosphate H NH2 F
    monophosphate monophosphate monophosphate H NH2 Cl
    diphosphate diphosphate diphosphate H NH2 NH2
    diphosphate diphosphate diphosphate H NH2 NH-cyclopropyl
    diphosphate diphosphate diphosphate H NH2 OH
    diphosphate diphosphate diphosphate H NH2 F
    diphosphate diphosphate diphosphate H NH2 Cl
    triphosphate triphosphate triphosphate H NH2 NH2
    triphosphate triphosphate triphosphate H NH2 NH-cyclopropyl
    triphosphate triphosphate triphosphate H NH2 OH
    triphosphate triphosphate triphosphate H NH2 F
    triphosphate triphosphate triphosphate H NH2 Cl
    H H H F NH2 NH2
    H H H F NH2 NH-cyclopropyl
    H H H F NH2 OH
    H H H F NH2 F
    H H H F NH2 Cl
    H H H Cl NH2 NH2
    H H H Cl NH2 NH-cyclopropyl
    H H H Cl NH2 OH
    H H H Cl NH2 F
    H H H Cl NH2 Cl
    H H H Br NH2 NH2
    H H H Br NH2 NH-cyclopropyl
    H H H Br NH2 OH
    H H H Br NH2 F
    H H H Br NH2 Cl
    H H H NH2 NH2 NH2
    H H H NH2 NH2 NH-cyclopropyl
    H H H NH2 NH2 OH
    H H H NH2 NH2 F
    H H H NH2 NH2 Cl
    H H H SH NH2 NH2
    H H H SH NH2 NH-cyclopropyl
    H H H SH NH2 OH
    H H H SH NH2 F
    H H H SH NH2 Cl
    acetyl H H H NH2 NH2
    acetyl H H H NH2 NH-cyclopropyl
    acetyl H H H NH2 OH
    acetyl H H H NH2 F
    acetyl H H H NH2 Cl
    acetyl H H F NH2 NH2
    acetyl H H F NH2 NH-cyclopropyl
    acetyl H H F NH2 OH
    acetyl H H F NH2 F
    acetyl H H F NH2 Cl
    H acetyl acetyl H NH2 NH2
    H acetyl acetyl H NH2 NH-cyclopropyl
    H acetyl acetyl H NH2 OH
    H acetyl acetyl H NH2 F
    H acetyl acetyl H NH2 Cl
    acetyl acetyl acetyl H NH2 NH2
    acetyl acetyl acetyl H NH2 NH-cyclopropyl
    acetyl acetyl acetyl H NH2 OH
    acetyl acetyl acetyl H NH2 F
    acetyl acetyl acetyl H NH2 Cl
    monophosphate acetyl acetyl H NH2 NH2
    monophosphate acetyl acetyl H NH2 NH-cyclopropyl
    monophosphate acetyl acetyl H NH2 OH
    monophosphate acetyl acetyl H NH2 F
    monophosphate acetyl acetyl H NH2 Cl
    diphosphate acetyl acetyl H NH2 NH2
    diphosphate acetyl acetyl H NH2 NH-cyclopropyl
    diphosphate acetyl acetyl H NH2 OH
    diphosphate acetyl acetyl H NH2 F
    diphosphate acetyl acetyl H NH2 Cl
    triphosphate acetyl acetyl H NH2 NH2
    triphosphate acetyl acetyl H NH2 NH-cyclopropyl
    triphosphate acetyl acetyl H NH2 OH
    triphosphate acetyl acetyl H NH2 F
    triphosphate acetyl acetyl H NH2 Cl
    H H H H Cl H
    H H H H Cl H
    H H H H Cl NH2
    H H H H Cl NH-cyclopropyl
    H H H H Cl NH-methyl
    H H H H Cl NH-ethyl
    H H H H Cl NH-acetyl
    H H H H Cl OH
    H H H H Cl OMe
    H H H H Cl OEt
    H H H H Cl O-cyclopropyl
    H H H H Cl O-acetyl
    H H H H Cl SH
    H H H H Cl SMe
    H H H H Cl SEt
    H H H H Cl S-cyclopropyl
    monophosphate H H H Cl NH2
    monophosphate H H H Cl NH-acetyl
    monophosphate H H H Cl NH-cyclopropyl
    monophosphate H H H Cl NH-methyl
    monophosphate H H H Cl NH-ethyl
    monophosphate H H H Cl OH
    monophosphate H H H Cl O-acetyl
    monophosphate H H H Cl OMe
    monophosphate H H H Cl OEt
    monophosphate H H H Cl O-cyclopropyl
    monophosphate H H H Cl SH
    monophosphate H H H Cl SMe
    monophosphate H H H Cl SEt
    monophosphate H H H Cl S-cyclopropyl
    diphosphate H H H Cl NH2
    diphosphate H H H Cl NH-acetyl
    diphosphate H H H Cl NH-cyclopropyl
    diphosphate H H H Cl NH-methyl
    diphosphate H H H Cl NH-ethyl
    diphosphate H H H Cl OH
    diphosphate H H H Cl O-acetyl
    diphosphate H H H Cl OMe
    diphosphate H H H Cl OEt
    diphosphate H H H Cl O-cyclopropyl
    diphosphate H H H Cl SH
    diphosphate H H H Cl SMe
    diphosphate H H H Cl SEt
    diphosphate H H H Cl S-cyclopropyl
    triphosphate H H H Cl NH2
    triphosphate H H H Cl NH-acetyl
    triphosphate H H H Cl NH-cyclopropyl
    triphosphate H H H Cl NH-methyl
    triphosphate H H H Cl NH-ethyl
    triphosphate H H H Cl OH
    triphosphate H H H Cl OMe
    triphosphate H H H Cl OEt
    triphosphate H H H Cl O-cyclopropyl
    triphosphate H H H Cl O-acetyl
    triphosphate H H H Cl SH
    triphosphate H H H Cl SMe
    triphosphate H H H Cl SEt
    triphosphate H H H Cl S-cyclopropyl
    monophosphate monophosphate monophosphate H Cl NH2
    monophosphate monophosphate monophosphate H Cl NH-cyclopropyl
    monophosphate monophosphate monophosphate H Cl OH
    diphosphate diphosphate diphosphate H Cl NH2
    diphosphate diphosphate diphosphate H Cl NH-cyclopropyl
    diphosphate diphosphate diphosphate H Cl OH
    triphosphate triphosphate triphosphate H Cl NH2
    triphosphate triphosphate triphosphate H Cl NH-cyclopropyl
    triphosphate triphosphate triphosphate H Cl OH
    H H H F Cl NH2
    H H H F Cl NH-cyclopropyl
    H H H F Cl OH
    H H H Cl Cl NH2
    H H H Cl Cl NH-cyclopropyl
    H H H Cl Cl OH
    H H H Br Cl NH2
    H H H Br Cl NH-cyclopropyl
    H H H Br Cl OH
    H H H NH2 Cl NH2
    H H H NH2 Cl NH-cyclopropyl
    H H H NH2 Cl OH
    H H H SH Cl NH2
    H H H SH Cl NH-cyclopropyl
    H H H SH Cl OH
    acetyl H H H Cl NH2
    acetyl H H H Cl NH-cyclopropyl
    acetyl H H H Cl OH
    acetyl H H F Cl NH2
    acetyl H H F Cl NH-cyclopropyl
    acetyl H H F Cl OH
    H acetyl acetyl H Cl NH2
    H acetyl acetyl H Cl NH-cyclopropyl
    H acetyl acetyl H Cl OH
    acetyl acetyl acetyl H Cl NH2
    acetyl acetyl acetyl H Cl NH-cyclopropyl
    acetyl acetyl acetyl H Cl OH
    monophosphate acetyl acetyl H Cl NH2
    monophosphate acetyl acetyl H Cl NH-cyclopropyl
    monophosphate acetyl acetyl H Cl OH
    diphosphate acetyl acetyl H Cl NH2
    diphosphate acetyl acetyl H Cl NH-cyclopropyl
    diphosphate acetyl acetyl H Cl OH
    triphosphate acetyl acetyl H Cl NH2
    triphosphate acetyl acetyl H Cl NH-cyclopropyl
    triphosphate acetyl acetyl H Cl OH
    H H H H Cl NH2
    H H H H Cl NH-cyclopropyl
    H H H H Cl OH
    H H H H Br NH2
    H H H H Br NH-cyclopropyl
    H H H H Br OH
  • Alternatively, the following nucleosides of Formula IV are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00037
  • wherein:
  • R1 R2 R3 X1 Y
    H H H H H
    H H H H NH2
    H H H H NH-cyclopropyl
    H H H H NH-methyl
    H H H H NH-ethyl
    H H H H NH-acetyl
    H H H H OH
    H H H H OMe
    H H H H OEt
    H H H H O-cyclopropyl
    H H H H O-acetyl
    H H H H SH
    H H H H SMe
    H H H H SEt
    H H H H S-cyclopropyl
    monophosphate H H H NH2
    monophosphate H H H NH-acetyl
    monophosphate H H H NH-cyclopropyl
    monophosphate H H H NH-methyl
    monophosphate H H H NH-ethyl
    monophosphate H H H OH
    monophosphate H H H O-acetyl
    monophosphate H H H OMe
    monophosphate H H H OEt
    monophosphate H H H O-cyclopropyl
    monophosphate H H H SH
    monophosphate H H H SMe
    monophosphate H H H SEt
    monophosphate H H H S-cyclopropyl
    diphosphate H H H NH2
    diphosphate H H H NH-acetyl
    diphosphate H H H NH-cyclopropyl
    diphosphate H H H NH-methyl
    diphosphate H H H NH-ethyl
    diphosphate H H H OH
    diphosphate H H H O-acetyl
    diphosphate H H H OMe
    diphosphate H H H OEt
    diphosphate H H H O-cyclopropyl
    diphosphate H H H SH
    diphosphate H H H SMe
    diphosphate H H H SEt
    diphosphate H H H S-cyclopropyl
    triphosphate H H H NH2
    triphosphate H H H NH-acetyl
    triphosphate H H H NH-cyclopropyl
    triphosphate H H H NH-methyl
    triphosphate H H H NH-ethyl
    triphosphate H H H OH
    triphosphate H H H OMe
    triphosphate H H H OEt
    triphosphate H H H O-cyclopropyl
    triphosphate H H H O-acetyl
    triphosphate H H H SH
    triphosphate H H H SMe
    triphosphate H H H SEt
    triphosphate H H H S-cyclopropyl
    monophosphate monophosphate monophosphate H NH2
    monophosphate monophosphate monophosphate H NH-cyclopropyl
    monophosphate monophosphate monophosphate H OH
    diphosphate diphosphate diphosphate H NH2
    diphosphate diphosphate diphosphate H NH-cyclopropyl
    diphosphate diphosphate diphosphate H OH
    triphosphate triphosphate triphosphate H NH2
    triphosphate triphosphate triphosphate H NH-cyclopropyl
    triphosphate triphosphate triphosphate H OH
    H H H F NH2
    H H H F NH-cyclopropyl
    H H H F OH
    H H H Cl NH2
    H H H Cl NH-cyclopropyl
    H H H Cl OH
    H H H Br NH2
    H H H Br NH-cyclopropyl
    H H H Br OH
    H H H NH2 NHa
    H H H NH2 NH-cyclopropyl
    H H H NH2 OH
    H H H SH NH2
    H H H SH NH-cyclopropyl
    H H H SH OH
    acetyl H H H NH2
    acetyl H H H NH-cyclopropyl
    acetyl H H H OH
    acetyl H H F NH2
    acetyl H H F NH-cyclopropyl
    acetyl H H F OH
    H acetyl acetyl H NH2
    H acetyl acetyl H NH-cyclopropyl
    H acetyl acetyl H OH
    acetyl acetyl acetyl H NH2
    acetyl acetyl acetyl H NH-cyclopropyl
    acetyl acetyl acetyl H OH
    monophosphate acetyl acetyl H NH2
    monophosphate acetyl acetyl H NH-cyclopropyl
    monophosphate acetyl acetyl H OH
    diphosphate acetyl acetyl H NH2
    diphosphate acetyl acetyl H NH-cyclopropyl
    diphosphate acetyl acetyl H OH
    triphosphate acetyl acetyl H NH2
    triphosphate acetyl acetyl H NH-cyclopropyl
    triphosphate acetyl acetyl H OH
  • Alternatively, the following nucleosides of Formula VII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00038
  • wherein:
  • R1 R2 R3 R6 X Base
    H H H CH3 O 2,4-O-
    Diacetyluracil
    H H H CH3 O Hypoxanthine
    H H H CH3 O 2,4-O-
    Diacetylthymine
    H H H CH3 O Thymine
    H H H CH3 O Cytosine
    H H H CH3 O 4-(N-mono-
    acetyl)cytosine
    H H H CH3 O 4-(N,N-
    diacetyl)cytosine
    H H H CH3 O Uracil
    H H H CH3 O 5-Fluorouracil
    H H H CH3 S 2,4-O-
    Diacetyluraci
    H H H CH3 S Hypoxanthine
    H H H CH3 S 2,4-O-
    Diacetylthymine
    H H H CH3 S Thymine
    H H H CH3 S Cytosine
    H H H CH3 S 4-(N-mono-
    acetyl)cytosine
    H H H CH3 S 4-(N,N-
    diacetyl)cytosine
    H H H CH3 S Uracil
    H H H CH3 S 5-Fluorouracil
    monophosphate H H CH3 O 2,4-O-
    Diacetyluracil
    monophosphate H H CH3 O Hypoxanthine
    monophosphate H H CH3 O 2,4-O-
    Diacetylthym
    monophosphate H H CH3 O Thymine
    monophosphate H H CH3 O Cytosine
    monophosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    monophosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    monophosphate H H CH3 O Uracil
    monophosphate H H CH3 O 5-Fluorouracil
    monophosphate H H CH3 S 2,4-O-
    Diacetyluracil
    monophosphate H H CH3 S Hypoxanthine
    monophosphate H H CH3 S 2,4-O-
    Diacetylthym
    monophosphate H H CH3 S Thymine
    monophosphate H H CH3 S Cytosine
    monophosphate H H CH3 S 4-(N-mono-
    acetyl)cytosine
    monophosphate H H CH3 S 4-(N,N-
    diacetyl)cytosine
    monophosphate H H CH3 S Uracil
    monophosphate H H CH3 S 5-Fluorouracil
    diphosphate H H CH3 O 2,4-O-
    Diacetyluracil
    diphosphate H H CH3 O Hypoxanthine
    diphosphate H H CH3 O 2,4-O-
    Diacetylthymine
    diphosphate H H CH3 O Thymine
    diphosphate H H CH3 O Cytosine
    diphosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    diphosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    diphosphate H H CH3 O Uracil
    diphosphate H H CH3 O 5-Fluorouracil
    diphosphate H H CH3 S 2,4-O-
    Diacetyluracil
    diphosphate H H CH3 S Hypoxanthine
    diphosphate H H CH3 S 2,4-O-
    Diacetylthym
    diphosphate H H CH3 S Thymine
    diphosphate H H CH3 S Cytosine
    triphosphate H H CH3 O 2,4-O-
    Diacetyluracil
    triphosphate H H CH3 O Hypoxanthine
    triphosphate H H CH3 O 2,4-O-
    Diacetylthymine
    triphosphate H H CH3 O Thymine
    triphosphate H H CH3 O Cytosine
    triphosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    triphosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    triphosphate H H CH3 O Uracil
    triphosphate H H CH3 O 5-Fluorouracil
    triphosphate H H CH3 S 2,4-O-
    Diacetyluracil
    triphosphate H H CH3 S Hypoxanthine
    triphosphate H H CH3 S 2,4-O-
    Diacetylthymine
    triphosphate H H CH3 S Thymine
    triphosphate H H CH3 S Cytosine
    monophosphate monophosphate monophosphate CF3 O 2,4-O-
    Diacetyluracil
    monophosphate monophosphate monophosphate CF3 O Hypoxanthine
    monophosphate monophosphate monophosphate CF3 O 2,4-O-
    Diacetylthymine
    monophosphate monophosphate monophosphate CF3 O Thymine
    monophosphate monophosphate monophosphate CF3 O Cytosine
    monophosphate monophosphate monophosphate CF3 O 4-(N-mono-
    acetyl)cytosine
    monophosphate monophosphate monophosphate CF3 O 4-(N,N-
    diacetyl)cytosine
    monophosphate monophosphate monophosphate CF3 O Uracil
    monophosphate monophosphate monophosphate CF3 O 5-Fluorouracil
    monophosphate monophosphate monophosphate CF3 S 2,4-O-
    Diacetyluracil
    monophosphate monophosphate monophosphate CF3 S Hypoxanthine
    monophosphate monophosphate monophosphate CF3 S 2,4-O-
    Diacetylthymine
    monophosphate monophosphate monophosphate CF3 S Thymine
    monophosphate monophosphate monophosphate CF3 S Cytosine
    monophosphate monophosphate monophosphate CF3 S 4-(N-mono-
    acetyl)cytosine
    monophosphate monophosphate monophosphate CF3 S 4-(N,N-
    diacetyl)cytosine
    monophosphate monophosphate monophosphate CF3 S Uracil
    monophosphate monophosphate monophosphate CF3 S 5-Fluorouracil
    acetyl acetyl acetyl CF3 O 4-(N,N-
    diacetyl)cytosine
    acetyl acetyl acetyl CF3 S 4-(N,N-
    diacetyl)cytosine
    acetyl acetyl acetyl 2-bromo-vinyl O 4-(N,N-
    diacetyl)cytosine
    acetyl acetyl acetyl 2-bromo-vinyl S 4-(N,N-
    diacetyl)cytosine
    H H H CH3 O 2-(N,N-diacetyl)-
    guanine
    H H H CH3 O 6-O-acetyl
    guanine
    H H H CH3 O 8-fluoroguanine
    H H H CH3 O guanine
    H H H CH3 O 6-(N,N-diacetyl)-
    adenine
    H H H CH3 O 2-fluoroadenine
    H H H CH3 O 8-fluoroadenine
    H H H CH3 O 2,8-difluoro-
    adenine
    H H H CH3 O adenine
    H H H CH3 S 2-(N,N-diacetyl)-
    guanine
    H H H CH3 S 6-O-acetyl
    guanine
    H H H CH3 S 8-fluoroguanine
    H H H CH3 S guanine
    H H H CH3 S 6-(N,N-diacetyl)-
    adenine
    H H H CH3 S 2-fluoroadenine
    H H H CH3 S 8-fluoroadenine
    H H H CH3 S 2,8-difluoro-
    adenine
    H H H CH3 S adenine
    monophosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    monophosphate H H CH3 O 6-O-acetyl
    guanine
    monophosphate H H CH3 O 8-fluoroguanine
    monophosphate H H CH3 O guanine
    monophosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    monophosphate H H CH3 O 2-fluoroadenine
    monophosphate H H CH3 O 8-fluoroadenine
    monophosphate H H CH3 O 2,8-difluoro-
    adenine
    monophosphate H H CH3 O adenine
    monophosphate H H CH3 S 2-(N,N-diacetyl)-
    guanine
    monophosphate H H CH3 S 6-O-acetyl
    guanine
    monophosphate H H CH3 S 8-fluoroguanine
    monophosphate H H CH3 S guanine
    monophosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    monophosphate H H CH3 S 2-fluoroadenine
    monophosphate H H CH3 S 8-fluoroadenine
    monophosphate H H CH3 S 2,8-difluoro-
    adenine
    monophosphate H H CH3 S adenine
    diphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    diphosphate H H CH3 O 6-O-acetyl
    guanine
    diphosphate H H CH3 O 8-fluoroguanine
    diphosphate H H CH3 O guanine
    diphosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    diphosphate H H CH3 O 2-fluoroadenine
    diphosphate H H CH3 O 8-fluoroadenine
    diphosphate H H CH3 O 2,8-difluoro-
    adenine
    diphosphate H H CH3 O adenine
    diphosphate H H CH3 S 2-(N,N-diacetyl)-
    guanine
    diphosphate H H CH3 S 6-O-acetyl
    guanine
    diphosphate H H CH3 S 8-fluoroguanine
    diphosphate H H CH3 S guanine
    diphosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    diphosphate H H CH3 S 2-fluoroadenine
    diphosphate H H CH3 S 8-fluoroadenine
    diphosphate H H CH3 S 2,8-difluoro-
    adenine
    diphosphate H H CH3 S adenine
    triphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    triphosphate H H CH3 O 6-O-acetyl
    guanine
    triphosphate H H CH3 O 8-fluoroguanine
    triphosphate H H CH3 O guanine
    triphosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    triphosphate H H CH3 O 2-fluoroadenine
    triphosphate H H CH3 O 8-fluoroadenine
    triphosphate H H CH3 O 2,8-difluoro-
    adenine
    triphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    triphosphate H H CH3 S 6-O-acetyl
    guanine
    triphosphate H H CH3 S 8-fluoroguanine
    triphosphate H H CH3 S guanine
    triphosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    triphosphate H H CH3 S 2-fluoroadenine
    triphosphate H H CH3 S 8-fluoroadenine
    triphosphate H H CH3 S 2,8-difluoro-
    adenine
    triphosphate H H CH3 S adenine
    monophosphate monophosphate monophosphate CF3 O 2-(N,N-diacetyl)-
    guanine
    monophosphate monophosphate monophosphate CF3 O 6-O-acetyl
    guanine
    monophosphate monophosphate monophosphate CF3 O 8-fluoroguanine
    monophosphate monophosphate monophosphate CF3 O guanine
    monophosphate monophosphate monophosphate CF3 O 6-(N,N-diacetyl)-
    adenine
    monophosphate monophosphate monophosphate CF3 O 2-fluoroadenine
    monophosphate monophosphate monophosphate CF3 O 8-fluoroadenine
    monophosphate monophosphate monophosphate CF3 O 2,8-difluoro-
    adenine
    monophosphate monophosphate monophosphate CF3 O adenine
    monophosphate monophosphate monophosphate CF3 S 2-(N,N-diacetyl)-
    guanine
    monophosphate monophosphate monophosphate CF3 S 6-O-acetyl
    guanine
    monophosphate monophosphate monophosphate CF3 S 8-fluoroguanine
    monophosphate monophosphate monophosphate CF3 S guanine
    monophosphate monophosphate monophosphate CF3 S 6-(N,N-diacetyl)-
    adenine
    monophosphate monophosphate monophosphate CF3 S 2-fluoroadenine
    monophosphate monophosphate monophosphate CF3 S 8-fluoroadenine
    monophosphate monophosphate monophosphate CF3 S 2,8-difluoro-
    adenine
    monophosphate monophosphate monophosphate CF3 S adenine
    acetyl acetyl acetyl CF3 O guanine
    acetyl acetyl acetyl CF3 S guanine
    acetyl acetyl acetyl 2-bromo-vinyl O guanine
    acetyl acetyl acetyl 2-bromo-vinyl S guanine
  • Alternatively, the following nucleosides of Formula VIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00039
  • wherein
  • R1 R2 R6 X Base
    H H CH3 O 2,4-O-Diacetyluracil
    H H CH3 O Hypoxanthine
    H H CH3 O 2,4-O-Diacetylthymine
    H H CH3 O Thymine
    H H CH3 O Cytosine
    H H CH3 O 4-(N-mono-acetyl)cytosine
    H H CH3 O 4-(N,N-diacetyl)cytosine
    H H CH3 O Uracil
    H H CH3 O 5-Fluorouracil
    H H CH3 S 2,4-O-Diacetyluracil
    H H CH3 S Hypoxanthine
    H H CH3 S 2,4-O-Diacetylthymine
    H H CH3 S Thymine
    H H CH3 S Cytosine
    H H CH3 S 4-(N-mono-acetyl)cytosine
    H H CH3 S 4-(N,N-diacetyl)cytosine
    H H CH3 S Uracil
    H H CH3 S 5-Fluorouracil
    monophosphate H CH3 O 2,4-O-Diacetyluracil
    monophosphate H CH3 O Hypoxanthine
    monophosphate H CH3 O 2,4-O-Diacetylthymine
    monophosphate H CH3 O Thymine
    monophosphate H CH3 O Cytosine
    monophosphate H CH3 O 4-(N-mono-acetyl)cytosine
    monophosphate H CH3 O 4-(N,N-diacetyl)cytosine
    monophosphate H CH3 O Uracil
    monophosphate H CH3 O 5-Fluorouracil
    monophosphate H CH3 S 2,4-O-Diacetyluracil
    monophosphate H CH3 S Hypoxanthine
    monophosphate H CH3 S 2,4-O-Diacetylthymine
    monophosphate H CH3 S Thymine
    monophosphate H CH3 S Cytosine
    monophosphate H CH3 S 4-(N-mono-acetyl)cytosine
    monophosphate H CH3 S 4-(N,N-diacetyl)cytosine
    monophosphate H CH3 S Uracil
    monophosphate H CH3 S 5-Fluorouracil
    diphosphate H CH3 O 2,4-O-Diacetyluracil
    diphosphate H CH3 O Hypoxanthine
    diphosphate H CH3 O 2,4-O-Diacetylthymine
    diphosphate H CH3 O Thymine
    diphosphate H CH3 O Cytosine
    diphosphate H CH3 O 4-(N-mono-acetyl)cytosine
    diphosphate H CH3 O 4-(N,N-diacetyl)cytosine
    diphosphate H CH3 O Uracil
    diphosphate H CH3 O 5-Fluorouracil
    diphosphate H CH3 S 2,4-O-Diacetyluracil
    diphosphate H CH3 S Hypoxanthine
    diphosphate H CH3 S 2,4-O-Diacetylthymine
    diphosphate H CH3 S Thymine
    diphosphate H CH3 S Cytosine
    diphosphate H CH3 S 4-(N-mono-acetyl)cytosine
    diphosphate H CH3 S 4-(N,N-diacetyl)cytosine
    diphosphate H CH3 S Uracil
    diphosphate H CH3 S 5-Fluorouracil
    triphosphate H CH3 O 2,4-O-Diacetyluracil
    triphosphate H CH3 O Hypoxanthine
    triphosphate H CH3 O 2,4-O-diacethylthymine
    triphosphate H CH3 O Thymine
    triphosphate H CH3 O Cytosine
    triphosphate H CH3 O 4-(N-mono-acetyl)cytosine
    triphosphate H CH3 O 4-(N,N-diacetyl)cytosine
    triphosphate H CH3 O Uracil
    triphosphate H CH3 O 5-Fluorouracil
    triphosphate H CH3 S 2,4-O-Diacetyluracil
    triphosphate H CH3 S Hypoxanthine
    triphosphate H CH3 S 2,4-O-Diacetylthymine
    triphosphate H CH3 S Thymine
    triphosphate H CH3 S Cytosine
    triphosphate H CH3 S 4-(N-mono-acetyl)cytosine
    triphosphate H CH3 S 4-(N,N-diacetyl)cytosine
    triphosphate H CH3 S Uracil
    triphosphate H CH3 S 5-Fluorouracil
    monophosphate monophosphate CF3 O 2,4-O-Diacetyluracil
    monophosphate monophosphate CF3 O Hypoxanthine
    monophosphate monophosphate CF3 O 2,4-O-Diacetylthymine
    monophosphate monophosphate CF3 O Thymine
    monophosphate monophosphate CF3 O Cytosine
    monophosphate monophosphate CF3 O 4-(N-mono-acetyl)cytosine
    monophosphate monophosphate CF3 O 4-(N,N-diacetyl)cytosine
    monophosphate monophosphate CF3 O Uracil
    monophosphate monophosphate CF3 O 5-Fluorouracil
    monophosphate monophosphate CF3 S 2,4-O-Diacetyluracil
    monophosphate monophosphate CF3 S Hypoxanthine
    monophosphate monophosphate CF3 S 2,4-O-Diacetylthymine
    monophosphate monophosphate CF3 S Thymine
    monophosphate monophosphate CF3 S Cytosine
    monophosphate monophosphate CF3 S 4-(N-mono-acetyl)cytosine
    monophosphate monophosphate CF3 S 4-(N,N-diacetyl)cytosine
    monophosphate monophosphate CF3 S Uracil
    monophosphate monophosphate CF3 S 5-Fluorouracil
    acetyl acetyl CF3 O 4-(N,N-diacetyl)cytosine
    acetyl acetyl CF3 S 4-(N,N-diacetyl)cytosine
    acetyl acetyl 2-bromo-vinyl O 4-(N,N-diacetyl)cytosine
    acetyl acetyl 2-bromo-vinyl S 4-(N,N-diacetyl)cytosine
    H H CH3 O 2-(N,N-diacetyl)-guanine
    H H CH3 O 6-O-acetyl guanine
    H H CH3 O 8-fluoroguanine
    H H CH3 O guanine
    H H CH3 O 6-(N,N-diacetyl)-adenine
    H H CH3 O 2-fluoroadenine
    H H CH3 O 8-fluoroadenine
    H H CH3 O 2,8-difluoro-adenine
    H H CH3 O adenine
    H H CH3 S 2-(N,N-diacetyl)-guanine
    H H CH3 S 6-O-acetyl guanine
    H H CH3 S 8-fluoroguanine
    H H CH3 S guanine
    H H CH3 S 6-(N,N-diacetyl)-adenine
    H H CH3 S 2-fluoroadenine
    H H CH3 S 8-fluoroadenine
    H H CH3 S 2,8-difluoro-adenine
    H H CH3 S adenine
    monophosphate H CH3 O 2-(N,N-diacetyl)-guanine
    monophosphate H CH3 O 6-O-acetyl guanine
    monophosphate H CH3 O 8-fluoroguanine
    monophosphate H CH3 O guanine
    monophosphate H CH3 O 6-(N,N-diacetyl)-adenine
    monophosphate H CH3 O 2-fluoroadenine
    monophosphate H CH3 O 8-fluoroadenine
    monophosphate H CH3 O 2,8-difluoro-adenine
    monophosphate H CH3 O adenine
    monophosphate H CH3 S 2-(N,N-diacetyl)-guanine
    monophosphate H CH3 S 6-O-acetyl guanine
    monophosphate H CH3 S 8-fluoroguanine
    monophosphate H CH3 S guanine
    monophosphate H CH3 S 6-(N,N-diacetyl)-adenine
    monophosphate H CH3 S 2-fluoroadenine
    monophosphate H CH3 S 8-fluoroadenine
    monophosphate H CH3 S 2,8-difluoro-adenine
    monophosphate H CH3 S adenine
    diphosphate H CH3 O 2-(N,N-diacetyl)-guanine
    diphosphate H CH3 O 6-O-acetyl guanine
    diphosphate H CH3 O 8-fluoroguanine
    diphosphate H CH3 O guanine
    diphosphate H CH3 O 6-(N,N-diacetyl)-adenine
    diphosphate H CH3 O 2-fluoroadenine
    diphosphate H CH3 O 8-fluoroadenine
    diphosphate H CH3 O 2,8-difluoro-adenine
    diphosphate H CH3 O adenine
    diphosphate H CH3 S 2-(N,N-diacetyl)-guanine
    diphosphate H CH3 S 6-O-acetyl guanine
    diphosphate H CH3 S 8-fluoroguanine
    diphosphate H CH3 S guanine
    diphosphate H CH3 S 6-(N,N-diacetyl)-adenine
    diphosphate H CH3 S 2-fluoroadenine
    diphosphate H CH3 S 8-fluoroadenine
    diphosphate H CH3 S 2,8-difluoro-adenine
    diphosphate H CH3 S adenine
    triphosphate H CH3 O 2-(N,N-diacetyl)-guanine
    triphosphate H CH3 O 6-O-acetyl guanine
    triphosphate H CH3 O 8-fluoroguanine
    triphosphate H CH3 O guanine
    triphosphate H CH3 O 6-(N,N-diacetyl)-adenine
    triphosphate H CH3 O 2-fluoroadenine
    triphosphate H CH3 O 8-fluoroadenine
    triphosphate H CH3 O 2,8-difluoro-adenine
    triphosphate H CH3 O adenine
    triphosphate H CH3 S 2-(N,N-diacetyl)-guanine
    triphosphate H CH3 S 6-O-acetyl guanine
    triphosphate H CH3 S 8-fluoroguanine
    triphosphate H CH3 S guanine
    triphosphate H CH3 S 6-(N,N-diacetyl)-adenine
    triphosphate H CH3 S 2-fluoroadenine
    triphosphate H CH3 S 8-fluoroadenine
    triphosphate H CH3 S 2,8-difluoro-adenine
    triphosphate H CH3 S adenine
    monophosphate monophosphate CF3 O 2-(N,N-diacetyl)-guanine
    monophosphate monophosphate CF3 O 6-O-acetyl guanine
    monophosphate monophosphate CF3 O 8-fluoroguanine
    monophosphate monophosphate CF3 O guanine
    monophosphate monophosphate CF3 O 6-(N,N-diacetyl)-adenine
    monophosphate monophosphate CF3 O 2-fluoroadenine
    monophosphate monophosphate CF3 O 8-fluoroadenine
    monophosphate monophosphate CF3 O 2,8-difluoro-adenine
    monophosphate monophosphate CF3 O adenine
    monophosphate monophosphate CF3 S 2-(N,N-diacetyl)-guanine
    monophosphate monophosphate CF3 S 6-O-acetyl guanine
    monophosphate monophosphate CF3 S 8-fluoroguanine
    monophosphate monophosphate CF3 S guanine
    monophosphate monophosphate CF3 S 6-(N,N-diacetyl)-adenine
    monophosphate monophosphate CF3 S 2-fluoroadenine
    monophosphate monophosphate CF3 S 8-fluoroadenine
    monophosphate monophosphate CF3 S 2,8-difluoro-adenine
    monophosphate monophosphate CF3 S adenine
    acetyl acetyl CF3 O guanine
    acetyl acetyl CF3 S guanine
    acetyl acetyl 2-bromo-vinyl O guanine
    acetyl acetyl 2-bromo-vinyl S guanine
  • Alternatively, the following nucleosides of Formula IX are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00040
  • wherein:
  • R1 R6 X Base
    H CH3 O 2,4-O-Diacetyluracil
    H CH3 O Hypoxanthine
    H CH3 O 2,4-O-Diacetylthymine
    H CH3 O Thymine
    H CH3 O Cytosine
    H CH3 O 4-(N-mono-acetyl)cytosine
    H CH3 O 4-(N,N-diacetyl)cytosine
    H CH3 O Uracil
    H CH3 O 5-Fluorouracil
    H CH3 S 2,4-O-Diacetyluracil
    H CH3 S Hypoxanthine
    H CH3 S 2,4-O-Diacetylthymine
    H CH3 S Thymine
    H CH3 S Cytosine
    H CH3 S 4-(N-mono-acetyl)cytosine
    H CH3 S 4-(N,N-diacetyl)cytosine
    H CH3 S Uracil
    H CH3 S 5-Fluorouracil
    monophosphate CH3 O 2,4-O-Diacetyluracil
    monophosphate CH3 O Hypoxanthine
    monophosphate CH3 O 2,4-O-Diacetylthymine
    monophosphate CH3 O Thymine
    monophosphate CH3 O Cytosine
    monophosphate CH3 O 4-(N-mono-acetyl)cytosine
    monophosphate CH3 O 4-(N,N-diacetyl)cytosine
    monophosphate CH3 O Uracil
    monophosphate CH3 O 5-Fluorouracil
    monophosphate CH3 S 2,4-O-Diacetyluracil
    monophosphate CH3 S Hypoxanthine
    monophosphate CH3 S 2,4-O-Diacetylthymine
    monophosphate CH3 S Thymine
    monophosphate CH3 S Cytosine
    monophosphate CH3 S 4-(N-mono-acetyl)cytosine
    monophosphate CH3 S 4-(N,N-diacetyl)cytos
    monophosphate CH3 S Uracil
    monophosphate CH3 S 5-Fluorouracil
    diphosphate CH3 O 2,4-O-Diacetyluracil
    diphosphate CH3 O Hypoxanthine
    diphosphate CH3 O 2,4-O-Diacetylthymine
    diphosphate CH3 O Thymine
    diphosphate CH3 O Cytosine
    diphosphate CH3 O 4-(N-mono-acetyl)cytosine
    diphosphate CH3 O 4-(N,N-diacetyl)cytosine
    diphosphate CH3 O Uracil
    diphosphate CH3 O 5-Fluorouracil
    diphosphate CH3 S 2,4-O-Diacetyluracil
    diphosphate CH3 S Hypoxanthine
    diphosphate CH3 S 2,4-O-Diacetylthymine
    diphosphate CH3 S Thymine
    diphosphate CH3 S Cytosine
    triphosphate CH3 O 2,4-O-Diacetyluracil
    triphosphate CH3 O Hypoxanthine
    triphosphate CH3 O 2,4-O-Diacetylthymine
    triphosphate CH3 O Thymine
    triphosphate CH3 O Cytosine
    triphosphate CH3 O 4-(N-mono-acetyl)cytosine
    triphosphate CH3 O 4-(N,N-diacetyl)cytosine
    triphosphate CH3 O Uracil
    triphosphate CH3 O 5-Fluorouracil
    triphosphate CH3 S 2,4-O-Diacetyluracil
    triphosphate CH3 S Hypoxanthine
    triphospahate CH3 S 2,4-O-Diacetylthymine
    triphospahate CH3 S Thymine
    triphospahate CH3 S Cytosine
    monophosphate CF3 O 2,4-O-Diacetyluracil
    monophosphate CF3 O Hypoxanthine
    monophosphate CF3 O 2,4-O-Diacetylthymine
    monophosphate CF3 O Thymine
    monophosphate CF3 O Cytosine
    monophosphate CF3 O 4-(N-mono-acetyl)cytosine
    monophosphate CF3 O 4-(N,N-diacetyl)cytos
    monophosphate CF3 O Uracil
    monophosphate CF3 O 5-Fluorouracil
    monophosphate CF3 S 2,4-O-Diacetyluracil
    monophosphate CF3 S Hypoxanthine
    monophosphate CF3 S 2,4-O-Diacetylthymine
    monophosphate CF3 S Thymine
    monophosphate CF3 S Cytosine
    monophosphate CF3 S 4-(N-mono-acetyl)cytosine
    monophosphate CF3 S 4-(N,N-diacetyl)cytosine
    monophosphate CF3 S Uracil
    monophosphate CF3 S 5-Fluorouracil
    acetyl CF3 O 4-(N,N-diacetyl)cytosine
    acetyl CF3 S 4-(N,N-diacetyl)cytosine
    acetyl 2-bromo-vinyl O 4-(N,N-diacetyl)cytosine
    acetyl 2-bromo-vinyl S 4-(N,N-diacetyl)cytosine
  • Alternatively, the following nucleosides of Formula XVI are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00041
  • wherein:
  • R1 R6 R7 R8 X Base R10 R9
    H CH3 H H O 2,4-O-Diacetyluracil OH Me
    H CH3 H H O Hypoxanthine OH Me
    H CH3 H H O 2,4-O-Diacetylthymine OH Me
    H CH3 H H O Thymine OH Me
    H CH3 H H O Cytosine OH Me
    H CH3 H H O 4-(N-mono-acetyl)cytosine OH Me
    H CH3 H H O 4-(N,N-diacetyl)cytosine OH Me
    H CH3 H H O Uracil OH Me
    H CH3 H H O 5-Fluorouracil OH Me
    H CH3 H H S 2,4-O-Diacetyluracil OH Me
    H CH3 H H S Hypoxanthine OH Me
    H CH3 H H S 2,4-O-Diacetylthymine OH Me
    H CH3 H H S Thymine OH Me
    H CH3 H H S Cytosine OH Me
    H CH3 H H S 4-(N-mono-acetyl)cytosine OH Me
    H CH3 H H S 4-(N,N-diacetyl)cytosine OH Me
    H CH3 H H S Uracil OH Me
    H CH3 H H S 5-Fluorouracil OH Me
    monophosphate CH3 H H O 2,4-O-Diacetyluracil OH Me
    monophosphate CH3 H H O Hypoxanthine OH Me
    monophosphate CH3 H H O 2,4-O-Diacetylthymine OH Me
    monophosphate CH3 H H O Thymine OH Me
    monophosphate CH3 H H O Cytosine OH Me
    monophosphate CH3 H H O 4-(N-mono-acetyl)cytosine OH Me
    monophosphate CH3 H H O 4-(N,N-diacetyl)cytosine OH Me
    monophosphate CH3 H H O Uracil OH Me
    monophosphate CH3 H H O 5-Fluorouracil OH Me
    monophosphate CH3 H H S 2,4-O-Diacetyluracil OH Me
    monophosphate CH3 H H S Hypoxanthine OH Me
    monophosphate CH3 H H S 2,4-O-Diacetylthymine OH Me
    monophosphate CH3 H H S Thymine OH Me
    monophosphate CH3 H H S Cytosine OH Me
    monophosphate CH3 H H S 4-(N-mono-acetyl)cytosine OH Me
    monophosphate CH3 H H S 4-(N,N-diacetyl)cytosine OH Me
    monophosphate CH3 H H S Uracil OH Me
    monophosphate CH3 H H S 5-Fluorouracil OH Me
    diphosphate CH3 H H O 2,4-O-Diacetyluracil OH Me
    diphosphate CH3 H H O Hypoxanthine OH Me
    diphosphate CH3 H H O 2,4-O-Diacetylthymine OH Me
    diphosphate CH3 H H O Thymine OH Me
    diphosphate CH3 H H O Cytosine OH Me
    diphosphate CH3 H H O 4-(N-mono-acetyl)cytosine OH Me
    diphosphate CH3 H H O 4-(N,N-diacetyl)cytosine OH Me
    diphosphate CH3 H H O Uracil OH Me
    diphosphate CH3 H H O 5-Fluorouracil OH Me
    diphosphate CH3 H H S 2,4-O-Diacetyluracil OH Me
    diphosphate CH3 H H S Hypoxanthine OH Me
    diphosphate CH3 H H S 2,4-O-Diacetylthymine OH Me
    diphosphate CH3 H H S Thymine OH Me
    diphosphate CH3 H H S Cytosine OH Me
    triphosphate CH3 H H O 2,4-O-Diacetyluracil OH Me
    triphosphate CH3 H H O Hypoxanthine OH Me
    triphosphate CH3 H H O 2,4-O-Diacetylthymine OH Me
    triphosphate CH3 H H O Thymine OH Me
    triphosphate CH3 H H O Cytosine OH Me
    triphosphate CH3 H H O 4-(N-mono-acetyl)cytosine OH Me
    triphosphate CH3 H H O 4-(N,N-diacetyl)cytosine OH Me
    triphosphate CH3 H H O Uracil OH Me
    triphosphate CH3 H H O 5-Fluorouracil OH Me
    triphosphate CH3 H H S 2,4-O-Diacetyluracil OH Me
    triphosphate CH3 H H S Hypoxanthine OH Me
    triphosphate CH3 H H S 2,4-O-Diacetylthymine OH Me
    triphosphate CH3 H H S Thymine OH Me
    triphosphate CH3 H H S Cytosine OH Me
    monophosphate CF3 H H O 2,4-O-Diacetyluracil OH Me
    monophosphate CF3 H H O Hypoxanthine OH Me
    monophosphate CF3 H H O 2,4-O-Diacetylthymine OH Me
    monophosphate CF3 H H O Thymine OH Me
    monophosphate CF3 H H O Cytosine OH Me
    monophosphate CF3 H H O 4-(N-mono-acetyl)cytosine OH Me
    monophosphate CF3 H H O 4-(N,N-diacetyl)cytosine OH Me
    monophosphate CF3 H H O Uracil OH Me
    monophosphate CF3 H H O 5-Fluorouracil OH Me
    monophosphate CF3 H H S 2,4-O-Diacetyluracil OH Me
    monophosphate CF3 H H S Hypoxanthine OH Me
    monophosphate CF3 H H S 2,4-O-Diacetylthymine OH Me
    monophosphate CF3 H H S Thymine OH Me
    monophosphate CF3 H H S Cytosine OH Me
    monophosphate CF3 H H S 4-(N-mono-acetyl)cytosine OH Me
    monophosphate CF3 H H S 4-(N,N-diacetyl)cytosine OH Me
    monophosphate CF3 H H S Uracil OH Me
    monophosphate CF3 H H S 5-Fluorouracil OH Me
    acetyl CH3 H H O 4-(N,N-diacetyl)cytosine H Br
    acetyl CH3 H H S 4-(N,N-diacetyl)cytosine H Br
    acetyl CH3 OH H O 4-(N,N-diacetyl)cytosine H Br
    acetyl CH3 OH H S 4-(N,N-diacetyl)cytosine H Br
  • Example 2: Preparation of 2′-C-methylriboadenine
  • The title compound was prepared according to a published procedure (R. E. Harry-O'kuru, J. M. Smith, and M. S. Wolfe, “A short, flexible route toward 2′-C-branched ribonucleosides”, J. Org. Chem. 1997, 62, 1754-1759) (Scheme 8).
  • Figure US20180235993A1-20180823-C00042
      • (a) Dess-Martin periodinane; (b) MeMgBr/TiCl4; (c) BzCl, DMAP, Et3N; (d) bis(trimethylsilyl)acetamide, N6-benzoyl adenine, TMSOTf; (e) NH3/MeOH
  • In a similar manner, but using the appropriate sugar and pyrimidine or purine bases, the following nucleosides of Formula II are prepared.
  • Figure US20180235993A1-20180823-C00043
  • wherein:
  • R1 R2 R3 X1 X2 Y
    H H H H H H
    H H H H H NH2
    H H H H H NH-cyclopropyl
    H H H H H NH-methyl
    H H H H H NH-ethyl
    H H H H H NH-acetyl
    H H H H H OH
    H H H H H OMe
    H H H H H OEt
    H H H H H O-cyclopropyl
    H H H H H O-acetyl
    H H H H H SH
    H H H H H SMe
    H H H H H SEt
    H H H H H S-cyclopropyl
    H H H H H F
    H H H H H Cl
    H H H H H Br
    H H H H H I
    monophosphate H H H H NH2
    monophosphate H H H H NH-acetyl
    monophosphate H H H H NH-cyclopropyl
    monophosphate H H H H NH-methyl
    monophosphate H H H H NH-ethyl
    monophosphate H H H H OH
    monophosphate H H H H O-acetyl
    monophosphate H H H H OMe
    monophosphate H H H H OEt
    monophosphate H H H H O-cyclopropyl
    monophosphate H H H H SH
    monophosphate H H H H SMe
    monophosphate H H H H SEt
    monophosphate H H H H S-cyclopropyl
    monophosphate H H H H F
    monophosphate H H H H Cl
    monophosphate H H H H Br
    monophosphate H H H H I
    diphosphate H H H H NH2
    diphosphate H H H H NH-acetyl
    diphosphate H H H H NH-cyclopropyl
    diphosphate H H H H NH-methyl
    diphosphate H H H H NH-ethyl
    diphosphate H H H H OH
    diphosphate H H H H O-acetyl
    diphosphate H H H H OMe
    diphosphate H H H H OEt
    diphosphate H H H H O-cyclopropyl
    diphosphate H H H H SH
    diphosphate H H H H SMe
    diphosphate H H H H SEt
    diphosphate H H H H S-cyclopropyl
    diphosphate H H H H F
    diphosphate H H H H Cl
    diphosphate H H H H Br
    diphosphate H H H H I
    triphosphate H H H H NH2
    triphosphate H H H H NH-acetyl
    triphosphate H H H H NH-cyclopropyl
    triphosphate H H H H NH-methyl
    triphosphate H H H H NH-ethyl
    triphosphate H H H H OH
    triphosphate H H H H OMe
    triphosphate H H H H OEt
    triphosphate H H H H O-cyclopropyl
    triphosphate H H H H O-acetyl
    triphosphate H H H H SH
    triphosphate H H H H SMe
    triphosphate H H H H SEt
    triphosphate H H H H S-cyclopropyl
    triphosphate H H H H F
    triphosphate H H H H Cl
    triphosphate H H H H Br
    triphosphate H H H H I
    monophosphate monophosphate monophosphate H H NH2
    monophosphate monophosphate monophosphate H H NH-cyclopropyl
    monophosphate monophosphate monophosphate H H OH
    monophosphate monophosphate monophosphate H H F
    monophosphate monophosphate monophosphate H H Cl
    diphosphate diphosphate diphosphate H H NH2
    diphosphate diphosphate diphosphate H H NH-cyclopropyl
    diphosphate diphosphate diphosphate H H OH
    diphosphate diphosphate diphosphate H H F
    diphosphate diphosphate diphosphate H H Cl
    triphosphate triphosphate triphosphate H H NH2
    triphosphate triphosphate triphosphate H H NH-cyclopropyl
    triphosphate triphosphate triphosphate H H OH
    triphosphate triphosphate triphosphate H H F
    triphosphate triphosphate triphosphate H H Cl
    H H H F H NH2
    H H H F H NH-cyclopropyl
    H H H F H OH
    H H H F H F
    H H H F H Cl
    H H H Cl H NH2
    H H H Cl H NH-cyclopropyl
    H H H Cl H OH
    H H H Cl H F
    H H H Cl H Cl
    H H H Br H NH2
    H H H Br H NH-cyclopropyl
    H H H Br H OH
    H H H Br H F
    H H H Br H Cl
    H H H NH2 H NH2
    H H H NH2 H NH-cyclopropyl
    H H H NH2 H OH
    H H H NH2 H F
    H H H NH2 H Cl
    H H H SH H NH2
    H H H SH H NH-cyclopropyl
    H H H SH H OH
    H H H SH H F
    H H H SH H Cl
    acetyl H H H H NH2
    acetyl H H H H NH-cyclopropyl
    acetyl H H H H OH
    acetyl H H H H F
    acetyl H H H H Cl
    acetyl H H F H NH2
    acetyl H H F H NH-cyclopropyl
    acetyl H H F H OH
    acetyl H H F H F
    acetyl H H F H Cl
    H acetyl acetyl H H NH2
    H acetyl acetyl H H NH-cyclopropyl
    H acetyl acetyl H H OH
    H acetyl acetyl H H F
    H acetyl acetyl H H Cl
    acetyl acetyl acetyl H H NH2
    acetyl acetyl acetyl H H NH-cyclopropyl
    acetyl acetyl acetyl H H OH
    acetyl acetyl acetyl H H F
    acetyl acetyl acetyl H H Cl
    monophosphate acetyl acetyl H H NH2
    monophosphate acetyl acetyl H H NH-cyclopropyl
    monophosphate acetyl acetyl H H OH
    monophosphate acetyl acetyl H H F
    monophosphate acetyl acetyl H H Cl
    diphosphate acetyl acetyl H H NH2
    diphosphate acetyl acetyl H H NH-cyclopropyl
    diphosphate acetyl acetyl H H OH
    diphosphate acetyl acetyl H H F
    diphosphate acetyl acetyl H H Cl
    triphosphate acetyl acetyl H H NH2
    triphosphate acetyl acetyl H H NH-cyclopropyl
    triphosphate acetyl acetyl H H OH
    triphosphate acetyl acetyl H H F
    triphosphate acetyl acetyl H H Cl
    H H H H NH2 H
    H H H H NH2 NH2
    H H H H NH2 NH-cyclopropyl
    H H H H NH2 NH-methyl
    H H H H NH2 NH-ethyl
    H H H H NH2 NH-acetyl
    H H H H NH2 OH
    H H H H NH2 OMe
    H H H H NH2 OEt
    H H H H NH2 O-cyclopropyl
    H H H H NH2 O-acetyl
    H H H H NH2 SH
    H H H H NH2 SMe
    H H H H NH2 SEt
    H H H H NH2 S-cyclopropyl
    H H H H NH2 F
    H H H H NH2 Cl
    H H H H NH2 Br
    H H H H NH2 I
    monophosphate H H H NH2 NH2
    monophosphate H H H NH2 NH-acetyl
    monophosphate H H H NH2 NH-cyclopropyl
    monophosphate H H H NH2 NH-methyl
    monophosphate H H H NH2 NH-ethyl
    monophosphate H H H NH2 OH
    monophosphate H H H NH2 O-acetyl
    monophosphate H H H NH2 OMe
    monophosphate H H H NH2 OEt
    monophosphate H H H NH2 O-cyclopropyl
    monophosphate H H H NH2 SH
    monophosphate H H H NH2 SMe
    monophosphate H H H NH2 SEt
    monophosphate H H H NH2 S-cyclopropyl
    monophosphate H H H NH2 F
    monophosphate H H H NH2 Cl
    monophosphate H H H NH2 Br
    monophosphate H H H NH2 I
    diphosphate H H H NH2 NH2
    diphosphate H H H NH2 NH-acetyl
    diphosphate H H H NH2 NH-cyclopropyl
    diphosphate H H H NH2 NH-methyl
    diphosphate H H H NH2 NH-ethyl
    diphosphate H H H NH2 OH
    diphosphate H H H NH2 O-acetyl
    diphosphate H H H NH2 OMe
    diphosphate H H H NH2 OEt
    diphosphate H H H NH2 O-cyclopropyl
    diphosphate H H H NH2 SH
    diphosphate H H H NH2 SMe
    diphosphate H H H NH2 SEt
    diphosphate H H H NH2 S-cyclopropyl
    diphosphate H H H NH2 F
    diphosphate H H H NH2 Cl
    diphosphate H H H NH2 Br
    diphosphate H H H NH2 I
    triphosphate H H H NH2 NH2
    triphosphate H H H NH2 NH-acetyl
    triphosphate H H H NH2 NH-cyclopropyl
    triphosphate H H H NH2 NH-methyl
    triphosphate H H H NH2 NH-ethyl
    triphosphate H H H NH2 OH
    triphosphate H H H NH2 OMe
    triphosphate H H H NH2 OEt
    triphosphate H H H NH2 O-cyclopropyl
    triphosphate H H H NH2 O-acetyl
    triphosphate H H H NH2 SH
    triphosphate H H H NH2 SMe
    triphosphate H H H NH2 SEt
    triphosphate H H H NH2 S-cyclopropyl
    triphosphate H H H NH2 F
    triphosphate H H H NH2 Cl
    triphosphate H H H NH2 Br
    triphosphate H H H NH2 I
    monophosphate monophosphate monophosphate H NH2 NH2
    monophosphate monophosphate monophosphate H NH2 NH-cyclopropyl
    monophosphate monophosphate monophosphate H NH2 OH
    monophosphate monophosphate monophosphate H NH2 F
    monophosphate monophosphate monophosphate H NH2 Cl
    diphosphate diphosphate diphosphate H NH2 NH2
    diphosphate diphosphate diphosphate H NH2 NH-cyclopropyl
    diphosphate diphosphate diphosphate H NH2 OH
    diphosphate diphosphate diphosphate H NH2 F
    diphosphate diphosphate diphosphate H NH2 Cl
    triphosphate triphosphate triphosphate H NH2 NH2
    triphosphate triphosphate triphosphate H NH2 NH-cyclopropyl
    triphosphate triphosphate triphosphate H NH2 OH
    triphosphate triphosphate triphosphate H NH2 F
    triphosphate triphosphate triphosphate H NH2 Cl
    H H H F NH2 NH2
    H H H F NH2 NH-cyclopropyl
    H H H F NH2 OH
    H H H F NH2 F
    H H H F NH2 Cl
    H H H Cl NH2 NH2
    H H H Cl NH2 NH-cyclopropyl
    H H H Cl NH2 OH
    H H H Cl NH2 F
    H H H Cl NH2 Cl
    H H H Br NH2 NH2
    H H H Br NH2 NH-cyclopropyl
    H H H Br NH2 OH
    H H H Br NH2 F
    H H H Br NH2 Cl
    H H H NH2 NH2 NH2
    H H H NH2 NH2 NH-cyclopropyl
    H H H NH2 NH2 OH
    H H H NH2 NH2 F
    H H H NH2 NH2 Cl
    H H H SH NH2 NH2
    H H H SH NH2 NH-cyclopropyl
    H H H SH NH2 OH
    H H H SH NH2 F
    H H H SH NH2 Cl
    acetyl H H H NH2 NH2
    acetyl H H H NH2 NH-cyclopropyl
    acetyl H H H NH2 OH
    acetyl H H H NH2 F
    acetyl H H H NH2 Cl
    acetyl H H F NH2 NH2
    acetyl H H F NH2 NH-cyclopropyl
    acetyl H H F NH2 OH
    acetyl H H F NH2 F
    acetyl H H F NH2 Cl
    H acetyl acetyl H NH2 NH2
    H acetyl acetyl H NH2 NH-cyclopropyl
    H acetyl acetyl H NH2 OH
    H acetyl acetyl H NH2 F
    H acetyl acetyl H NH2 Cl
    acetyl acetyl acetyl H NH2 NH2
    acetyl acetyl acetyl H NH2 NH-cyclopropyl
    acetyl acetyl acetyl H NH2 OH
    acetyl acetyl acetyl H NH2 F
    acetyl acetyl acetyl H NH2 Cl
    monophosphate acetyl acetyl H NH2 NH2
    monophosphate acetyl acetyl H NH2 NH-cyclopropyl
    monophosphate acetyl acetyl H NH2 OH
    monophosphate acetyl acetyl H NH2 F
    monophosphate acetyl acetyl H NH2 Cl
    diphosphate acetyl acetyl H NH2 NH2
    diphosphate acetyl acetyl H NH2 NH-cyclopropyl
    diphosphate acetyl acetyl H NH2 OH
    diphosphate acetyl acetyl H NH2 F
    diphosphate acetyl acetyl H NH2 Cl
    triphosphate acetyl acetyl H NH2 NH2
    triphosphate acetyl acetyl H NH2 NH-cyclopropyl
    triphosphate acetyl acetyl H NH2 OH
    triphosphate acetyl acetyl H NH2 F
    triphosphate acetyl acetyl H NH2 Cl
    H H H H Cl H
    H H H H Cl H
    H H H H Cl NH2
    H H H H Cl NH-cyclopropyl
    H H H H Cl NH-methyl
    H H H H Cl NH-ethyl
    H H H H Cl NH-acetyl
    H H H H Cl OH
    H H H H Cl OMe
    H H H H Cl OEt
    H H H H Cl O-cyclopropyl
    H H H H Cl O-acetyl
    H H H H Cl SH
    H H H H Cl SMe
    H H H H Cl SEt
    H H H H Cl S-cyclopropyl
    monophosphate H H H Cl NH2
    monophosphate H H H Cl NH-acetyl
    monophosphate H H H Cl NH-cyclopropyl
    monophosphate H H H Cl NH-methyl
    monophosphate H H H Cl NH-ethyl
    monophosphate H H H Cl OH
    monophosphate H H H Cl O-acetyl
    monophosphate H H H Cl OMe
    monophosphate H H H Cl OEt
    monophosphate H H H Cl O-cyclopropyl
    monophosphate H H H Cl SH
    monophosphate H H H Cl SMe
    monophosphate H H H Cl SEt
    monophosphate H H H Cl S-cyclopropyl
    diphosphate H H H Cl NH2
    diphosphate H H H Cl NH-acetyl
    diphosphate H H H Cl NH-cyclopropyl
    diphosphate H H H Cl NH-methyl
    diphosphate H H H Cl NH-ethyl
    diphosphate H H H Cl OH
    diphosphate H H H Cl O-acetyl
    diphosphate H H H Cl OMe
    diphosphate H H H Cl OEt
    diphosphate H H H Cl O-cyclopropyl
    diphosphate H H H Cl SH
    diphosphate H H H Cl SMe
    diphosphate H H H Cl SEt
    diphosphate H H H Cl S-cyclopropyl
    triphosphate H H H Cl NH2
    triphosphate H H H Cl NH-acetyl
    triphosphate H H H Cl NH-cyclopropyl
    triphosphate H H H Cl NH-methyl
    triphosphate H H H Cl NH-ethyl
    triphosphate H H H Cl OH
    triphosphate H H H Cl OMe
    triphosphate H H H Cl OEt
    triphosphate H H H Cl O-cyclopropyl
    triphosphate H H H Cl O-acetyl
    triphosphate H H H Cl SH
    triphosphate H H H Cl SMe
    triphosphate H H H Cl SEt
    triphosphate H H H Cl S-cyclopropyl
    monophosphate monophosphate monophosphate H Cl NH2
    monophosphate monophosphate monophosphate H Cl NH-cyclopropyl
    monophosphate monophosphate monophosphate H Cl OH
    diphosphate diphosphate diphosphate H Cl NH2
    diphosphate diphosphate diphosphate H Cl NH-cyclopropyl
    diphosphate diphosphate diphosphate H Cl OH
    triphosphate triphosphate triphosphate H Cl NH2
    triphosphate triphosphate triphosphate H Cl NH-cyclopropyl
    triphosphate triphosphate triphosphate H Cl OH
    H H H F Cl NH2
    H H H F Cl NH-cyclopropyl
    H H H F Cl OH
    H H H Cl Cl NH2
    H H H Cl Cl NH-cyclopropyl
    H H H Cl Cl OH
    H H H Br Cl NH2
    H H H Br Cl NH-cyclopropyl
    H H H Br Cl OH
    H H H NH2 Cl NH2
    H H H NH2 Cl NH-cyclopropyl
    H H H NH2 Cl OH
    H H H SH Cl NH2
    H H H SH Cl NH-cyclopropyl
    H H H SH Cl OH
    acetyl H H H Cl NH2
    acetyl H H H Cl NH-cyclopropyl
    acetyl H H H Cl OH
    acetyl H H F Cl NH2
    acetyl H H F Cl NH-cyclopropyl
    acetyl H H F Cl OH
    H acetyl acetyl H Cl NH2
    H acetyl acetyl H Cl NH-cyclopropyl
    H acetyl acetyl H Cl OH
    acetyl acetyl acetyl H Cl NH2
    acetyl acetyl acetyl H Cl NH-cyclopropyl
    acetyl acetyl acetyl H Cl OH
    monophosphate acetyl acetyl H Cl NH2
    monophosphate acetyl acetyl H Cl NH-cyclopropyl
    monophosphate acetyl acetyl H Cl OH
    diphosphate acetyl acetyl H Cl NH2
    diphosphate acetyl acetyl H Cl NH-cyclopropyl
    diphosphate acetyl acetyl H Cl OH
    triphosphate acetyl acetyl H Cl NH2
    triphosphate acetyl acetyl H Cl NH-cyclopropyl
    triphosphate acetyl acetyl H Cl OH
    H H H H Cl NH2
    H H H H Cl NH-cyclopropyl
    H H H H Cl OH
    H H H H Br NH2
    H H H H Br NH-cyclopropyl
    H H H H Br OH
  • Alternatively, the following nucleosides of Formula V are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00044
  • wherein:
  • R1 R2 R3 X1 Y
    H H H H H
    H H H H NH2
    H H H H NH-
    cyclopropyl
    H H H H NH-methyl
    H H H H NH-ethyl
    H H H H NH-acetyl
    H H H H OH
    H H H H OMe
    H H H H OEt
    H H H H O-cyclopropyl
    H H H H O-acetyl
    H H H H SH
    H H H H SMe
    H H H H SEt
    H H H H S-cyclopropyl
    monophosphate H H H NH2
    monophosphate H H H NH-acetyl
    monophosphate H H H NH-
    cyclopropyl
    monophosphate H H H NH-methyl
    monophosphate H H H NH-ethyl
    monophosphate H H H OH
    monophosphate H H H O-acetyl
    monophosphate H H H OMe
    monophosphate H H H OEt
    monophosphate H H H O-cyclopropyl
    monophosphate H H H SH
    monophosphate H H H SMe
    monophosphate H H H SEt
    monophosphate H H H S-cyclopropyl
    diphosphate H H H NH2
    diphosphate H H H NH-acetyl
    diphosphate H H H NH-
    cyclopropyl
    diphosphate H H H NH-methyl
    diphosphate H H H NH-ethyl
    diphosphate H H H OH
    diphosphate H H H O-acetyl
    diphosphate H H H OMe
    diphosphate H H H OEt
    diphosphate H H H O-cyclopropyl
    diphosphate H H H SH
    diphosphate H H H SMe
    diphosphate H H H SEt
    diphosphate H H H S-cyclopropyl
    triphosphate H H H NH2
    triphosphate H H H NH-acetyl
    triphosphate H H H NH-
    cyclopropyl
    triphosphate H H H NH-methyl
    triphosphate H H H NH-ethyl
    triphosphate H H H OH
    triphosphate H H H OMe
    triphosphate H H H OEt
    triphosphate H H H O-cyclopropyl
    triphosphate H H H O-acetyl
    triphosphate H H H SH
    triphosphate H H H SMe
    triphosphate H H H SEt
    triphosphate H H H S-cyclopropyl
    monophosphate monophosphate monophosphate H NH2
    monophosphate monophosphate monophosphate H NH-
    cyclopropyl
    monophosphate monophosphate monophosphate H OH
    diphosphate diphosphate diphosphate H NH2
    diphosphate diphosphate diphosphate H NH-
    cyclopropyl
    diphosphate diphosphate diphosphate H OH
    triphosphate triphosphate triphosphate H NH2
    triphosphate triphosphate triphosphate H NH-
    cyclopropyl
    triphosphate triphosphate triphosphate H OH
    H H H F NH2
    H H H F NH-
    cyclopropyl
    H H H F OH
    H H H Cl NH2
    H H H Cl NH-
    cyclopropyl
    H H H Cl OH
    H H H Br NH2
    H H H Br NH-
    cyclopropyl
    H H H Br OH
    H H H NH2 NH2
    H H H NH2 NH-
    cyclopropyl
    H H H NH2 OH
    H H H SH NH2
    H H H SH NH-
    cyclopropyl
    H H H SH OH
    acetyl H H H NH2
    acetyl H H H NH-
    cyclopropyl
    acetyl H H H OH
    acetyl H H F NH2
    acetyl H H F NH-
    cyclopropyl
    acetyl H H F OH
    H acetyl acetyl H NH2
    H acetyl acetyl H NH-
    cyclopropyl
    H acetyl acetyl H OH
    acetyl acetyl acetyl H NH2
    acetyl acetyl acetyl H NH-
    cyclopropyl
    acetyl acetyl acetyl H OH
    monophosphate acetyl acetyl H NH2
    monophosphate acetyl acetyl H NH-
    cyclopropyl
    monophosphate acetyl acetyl H OH
    diphosphate acetyl acetyl H NH2
    diphosphate acetyl acetyl H NH-
    cyclopropyl
    diphosphate acetyl acetyl H OH
    triphosphate acetyl acetyl H NH2
    triphosphate acetyl acetyl H NH-
    cyclopropyl
    triphosphate acetyl acetyl H OH
  • Alternatively, the following nucleosides of Formula X are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00045
  • wherein:
  • R1 R2 R3 R6 X Base
    H H H CH3 O 2,4-O-
    Diacetyluracil
    H H H CH3 O Hypoxanthine
    H H H CH3 O 2,4-O-
    Diacetylthymine
    H H H CH3 O Thymine
    H H H CH3 O Cytosine
    H H H CH3 O 4-(N-mono-
    acetyl)cytosine
    H H H CH3 O 4-(N,N-
    diacetyl)cytosine
    H H H CH3 O Uracil
    H H H CH3 O 5-Fluorouracil
    H H H CH3 S 2,4-O-
    Diacetyluraci
    H H H CH3 S Hypoxanthine
    H H H CH3 S 2,4-O-
    Diacetylthymine
    H H H CH3 S Thymine
    H H H CH3 S Cytosine
    H H H CH3 S 4-(N-mono-
    acetyl)cytosine
    H H H CH3 S 4-(N,N-
    diacetyl)cytosine
    H H H CH3 S Uracil
    H H H CH3 S 5-Fluorouracil
    monophosphate H H CH3 O 2,4-O-
    Diacetyluracil
    monophosphate H H CH3 O Hypoxanthine
    monophosphate H H CH3 O 2,4-O-
    Diacetylthym
    monophosphate H H CH3 O Thymine
    monophosphate H H CH3 O Cytosine
    monophosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    monophosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    monophosphate H H CH3 O Uracil
    monophosphate H H CH3 O 5-Fluorouracil
    monophosphate H H CH3 S 2,4-O-
    Diacetyluracil
    monophosphate H H CH3 S Hypoxanthine
    monophosphate H H CH3 S 2,4-O-
    Diacetylthym
    monophosphate H H CH3 S Thymine
    monophosphate H H CH3 S Cytosine
    monophosphate H H CH3 S 4-(N-mono-
    acetyl)cytosine
    monophosphate H H CH3 S 4-(N,N-
    diacetyl)cytosine
    monophosphate H H CH3 S Uracil
    monophosphate H H CH3 S 5-Fluorouracil
    diphosphate H H CH3 O 2,4-O-
    Diacetyluracil
    diphosphate H H CH3 O Hypoxanthine
    diphosphate H H CH3 O 2,4-O-
    Diacetylthymine
    diphosphate H H CH3 O Thymine
    diphosphate H H CH3 O Cytosine
    diphosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    diphosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    diphosphate H H CH3 O Uracil
    diphosphate H H CH3 O 5-Fluorouracil
    diphosphate H H CH3 S 2,4-O-
    Diacetyluracil
    diphosphate H H CH3 S Hypoxanthine
    diphosphate H H CH3 S 2,4-O-
    Diacetylthym
    diphosphate H H CH3 S Thymine
    diphosphate H H CH3 S Cytosine
    triphosphate H H CH3 O 2,4-O-
    Diacetyluracil
    triphosphate H H CH3 O Hypoxanthine
    triphosphate H H CH3 O 2,4-O-
    Diacetylthymine
    triphosphate H H CH3 O Thymine
    triphosphate H H CH3 O Cytosine
    triphosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    triphosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    triphosphate H H CH3 O Uracil
    triphosphate H H CH3 O 5-Fluorouracil
    triphosphate H H CH3 S 2,4-O-
    Diacetyluracil
    triphosphate H H CH3 S Hypoxanthine
    triphosphate H H CH3 S 2,4-O-
    Diacetylthymine
    triphosphate H H CH3 S Thymine
    triphosphate H H CH3 S Cytosine
    monophosphate monophosphate monophosphate CF3 O 2,4-O-
    Diacetyluracil
    monophosphate monophosphate monophosphate CF3 O Hypoxanthine
    monophosphate monophosphate monophosphate CF3 O 2,4-O-
    Diacetylthymine
    monophosphate monophosphate monophosphate CF3 O Thymine
    monophosphate monophosphate monophosphate CF3 O Cytosine
    monophosphate monophosphate monophosphate CF3 O 4-(N-mono-
    acetyl)cytosine
    monophosphate monophosphate monophosphate CF3 O 4-(N,N-
    diacetyl)cytosine
    monophosphate monophosphate monophosphate CF3 O Uracil
    monophosphate monophosphate monophosphate CF3 O 5-Fluorouracil
    monophosphate monophosphate monophosphate CF3 S 2,4-O-
    Diacetyluracil
    monophosphate monophosphate monophosphate CF3 S Hypoxanthine
    monophosphate monophosphate monophosphate CF3 S 2,4-O-
    Diacetylthymine
    monophosphate monophosphate monophosphate CF3 S Thymine
    monophosphate monophosphate monophosphate CF3 S Cytosine
    monophosphate monophosphate monophosphate CF3 S 4-(N-mono-
    acetyl)cytosine
    monophosphate monophosphate monophosphate CF3 S 4-(N,N-
    diacetyl)cytosine
    monophosphate monophosphate monophosphate CF3 S Uracil
    monophosphate monophosphate monophosphate CF3 S 5-Fluorouracil
    acetyl acetyl acetyl CF3 O 4-(N,N-
    diacetyl)cytosine
    acetyl acetyl acetyl CF3 S 4-(N,N-
    diacetyl)cytosine
    acetyl acetyl acetyl 2-bromo- O 4-(N,N-
    vinyl diacetyl)cytosine
    acetyl acetyl acetyl 2-bromo- S 4-(N,N-
    vinyl diacetyl)cytosine
    H H H CH3 O 2-(N,N-diacetyl)-
    guanine
    H H H CH3 O 6-O-acetyl
    guanine
    H H H CH3 O 8-fluoroguanine
    H H H CH3 O guanine
    H H H CH3 O 6-(N,N-diacetyl)-
    adenine
    H H H CH3 O 2-fluoroadenine
    H H H CH3 O 8-fluoroadenine
    H H H CH3 O 2,8-difluoro-
    adenine
    H H H CH3 O adenine
    H H H CH3 S 2-(N,N-diacetyl)-
    guanine
    H H H CH3 S 6-O-acetyl
    guanine
    H H H CH3 S 8-fluoroguanine
    H H H CH3 S guanine
    H H H CH3 S 6-(N,N-diacetyl)-
    adenine
    H H H CH3 S 2-fluoroadenine
    H H H CH3 S 8-fluoroadenine
    H H H CH3 S 2,8-difluoro-
    adenine
    H H H CH3 S adenine
    monophosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    monophosphate H H CH3 O 6-O-acetyl
    guanine
    monophosphate H H CH3 O 8-fluoroguanine
    monophosphate H H CH3 O guanine
    monophosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    monophosphate H H CH3 O 2-fluoroadenine
    monophosphate H H CH3 O 8-fluoroadenine
    monophosphate H H CH3 O 2,8-difluoro-
    adenine
    monophosphate H H CH3 O adenine
    monophosphate H H CH3 S 2-(N,N-diacetyl)-
    guanine
    monophosphate H H CH3 S 6-O-acetyl
    guanine
    monophosphate H H CH3 S 8-fluoroguanine
    monophosphate H H CH3 S guanine
    monophosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    monophosphate H H CH3 S 2-fluoroadenine
    monophosphate H H CH3 S 8-fluoroadenine
    monophosphate H H CH3 S 2,8-difluoro-
    adenine
    monophosphate H H CH3 S adenine
    diphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    diphosphate H H CH3 O 6-O-acetyl
    guanine
    diphosphate H H CH3 O 8-fluoroguanine
    diphosphate H H CH3 O guanine
    diphosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    diphosphate H H CH3 O 2-fluoroadenine
    diphosphate H H CH3 O 8-fluoroadenine
    diphosphate H H CH3 O 2,8-difluoro-
    adenine
    diphosphate H H CH3 O adenine
    diphosphate H H CH3 S 2-(N,N-diacetyl)-
    guanine
    diphosphate H H CH3 S 6-O-acetyl
    guanine
    diphosphate H H CH3 S 8-fluoroguanine
    diphosphate H H CH3 S guanine
    diphosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    diphosphate H H CH3 S 2-fluoroadenine
    diphosphate H H CH3 S 8-fluoroadenine
    diphosphate H H CH3 S 2,8-difluoro-
    adenine
    diphosphate H H CH3 S adenine
    triphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    triphosphate H H CH3 O 6-O-acetyl
    guanine
    triphosphate H H CH3 O 8-fluoroguanine
    triphosphate H H CH3 O guanine
    triphosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    triphosphate H H CH3 O 2-fluoroadenine
    triphosphate H H CH3 O 8-fluoroadenine
    triphosphate H H CH3 O 2,8-difluoro-
    adenine
    triphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    triphosphate H H CH3 S 6-O-acetyl
    guanine
    triphosphate H H CH3 S 8-fluoroguanine
    triphosphate H H CH3 S guanine
    triphosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    triphosphate H H CH3 S 2-fluoroadenine
    triphosphate H H CH3 S 8-fluoroadenine
    triphosphate H H CH3 S 2,8-difluoro-
    adenine
    triphosphate H H CH3 S adenine
    monophosphate monophosphate monophosphate CF3 O 2-(N,N-diacetyl)-
    guanine
    monophosphate monophosphate monophosphate CF3 O 6-O-acetyl
    guanine
    monophosphate monophosphate monophosphate CF3 O 8-fluoroguanine
    monophosphate monophosphate monophosphate CF3 O guanine
    monophosphate monophosphate monophosphate CF3 O 6-(N,N-diacetyl)-
    adenine
    monophosphate monophosphate monophosphate CF3 O 2-fluoroadenine
    monophosphate monophosphate monophosphate CF3 O 8-fluoroadenine
    monophosphate monophosphate monophosphate CF3 O 2,8-difluoro-
    adenine
    monophosphate monophosphate monophosphate CF3 O adenine
    monophosphate monophosphate monophosphate CF3 S 2-(N,N-diacetyl)-
    guanine
    monophosphate monophosphate monophosphate CF3 S 6-O-acetyl
    guanine
    monophosphate monophosphate monophosphate CF3 S 8-fluoroguanine
    monophosphate monophosphate monophosphate CF3 S guanine
    monophosphate monophosphate monophosphate CF3 S 6-(N,N-diacetyl)-
    adenine
    monophosphate monophosphate monophosphate CF3 S 2-fluoroadenine
    monophosphate monophosphate monophosphate CF3 S 8-fluoroadenine
    monophosphate monophosphate monophosphate CF3 S 2,8-difluoro-
    adenine
    monophosphate monophosphate monophosphate CF3 S adenine
    acetyl acetyl acetyl CF3 O guanine
    acetyl acetyl acetyl CF3 S guanine
    acetyl acetyl acetyl 2-bromo- O guanine
    vinyl
    acetyl acetyl acetyl 2-bromo- S guanine
    vinyl
  • Alternatively, the following nucleosides of Formula XI are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00046
  • wherein:
  • R1 R2 R7 R6 X Base
    H H H CH3 O 2,4-O-Diacetyluracil
    H H H CH3 O Hypoxanthine
    H H H CH3 O 2,4-O-Diacetylthymine
    H H H CH3 O Thymine
    H H H CH3 O Cytosine
    H H H CH3 O 4-(N-mono-
    acetyl)cytosine
    H H H CH3 O 4-(N,N-diacetyl)cytosine
    H H H CH3 O Uracil
    H H H CH3 O 5-Fluorouracil
    H H H CH3 S 2,4-O-Diacetyluracil
    H H H CH3 S Hypoxanthine
    H H H CH3 S 2,4-O-Diacetylthymine
    H H H CH3 S Thymine
    H H H CH3 S Cytosine
    H H H CH3 S 4-(N-mono-acetyl)cytosin
    H H H CH3 S 4-(N,N-diacetyl)cytosine
    H H H CH3 S Uracil
    H H H CH3 S 5-Fluorouracil
    CH3
    monophosphate H H CH3 O 2,4-O-Diacetyluracil
    monophosphate H H CH3 O Hypoxanthine
    monophosphate H H CH3 O 2,4-O-Diacetylthymine
    monophosphate H H CH3 O Thymine
    monophosphate H H CH3 O Cytosine
    monophosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    monophosphate H H CH3 O 4-(N,N-diacetyl)cytosine
    monophosphate H H CH3 O Uracil
    monophosphate H H CH3 O 5-Fluorouracil
    monophosphate H H CH3 S 2,4-O-Diacetyluracil
    monophosphate H H CH3 S Hypoxanthine
    monophosphate H H CH3 S 2,4-O-Diacetylthymine
    monophosphate H H CH3 S Thymine
    monophosphate H H CH3 S Cytosine
    monophosphate H H CH3 S 4-(N-mono-
    acetyl)cytosine
    monophosphate H H CH3 S 4-(N,N-diacetyl)cytosine
    monophosphate H H CH3 S Uracil
    monophosphate H H CH3 S 5-Fluorouracil
    diphosphate H H CH3 O 2,4-O-Diacetylurac
    diphosphate H H CH3 O Hypoxanthine
    diphosphate H H CH3 O 2,4-O-Diacetylthymine
    diphosphate H H CH3 O Thymine
    diphosphate H H CH3 O Cytosine
    diphosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    diphosphate H H CH3 O 4-(N,N-diacetyl)cytosine
    diphosphate H H CH3 O Uracil
    diphosphate H H CH3 O 5-Fluorouracil
    diphosphate H H CH3 S 2,4-O-Diacetyluracil
    diphosphate H H CH3 S Hypoxanthine
    diphosphate H H CH3 S 2,4-O-Diacetylthym
    diphosphate H H CH3 S Thymine
    diphosphate H H CH3 S Cytosine
    triphosphate H H CH3 O 2,4-O-Diacetyluracil
    triphosphate H H CH3 O Hypoxanthine
    triphosphate H H CH3 O 2,4-O-Diacetylthymine
    triphosphate H H CH3 O Thymine
    triphosphate H H CH3 O Cytosine
    triphosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    triphosphate H H CH3 O 4-(N,N-diacetyl)cytos
    triphosphate H H CH3 O Uracil
    triphosphate H H CH3 O 5-Fluorouracil
    triphosphate H H CH3 S 2,4-O-Diacetyluracil
    triphosphate H H CH3 S Hypoxanthine
    triphosphate H H CH3 S 2,4-O-Diacetylthym
    triphosphate H H CH3 S Thymine
    triphosphate H H CH3 S Cytosine
    monophosphate monophosphate Br CF3 O 2,4-O-Diacetyluracil
    monophosphate monophosphate Br CF3 O Hypoxanthine
    monophosphate monophosphate Br CF3 O 2,4-O-Diacetylthymine
    monophosphate monophosphate Br CF3 O Thymine
    monophosphate monophosphate Br CF3 O Cytosine
    monophosphate monophosphate Br CF3 O 4-(N-mono-
    acetyl)cytosine
    monophosphate monophosphate Br CF3 O 4-(N,N-diacetyl)cytosine
    monophosphate monophosphate Br CF3 O Uracil
    monophosphate monophosphate Br CF3 O 5-Fluorouracil
    monophosphate monophosphate Br CF3 S 2,4-O-Diacetyluracil
    monophosphate monophosphate Br CF3 S Hypoxanthine
    monophosphate monophosphate Br CF3 S 2,4-O-Diacetylthymine
    monophosphate monophosphate Br CF3 S Thymine
    monophosphate monophosphate Br CF3 S Cytosine
    monophosphate monophosphate Br CF3 S 4-(N-mono-
    acetyl)cytosine
    monophosphate monophosphate Br CF3 S 4-(N,N-diacetyl)cytos
    monophosphate monophosphate Br CF3 S Uracil
    monophosphate monophosphate Br CF3 S 5-Fluorouracil
    acetyl acetyl NO2 CF3 O 4-(N,N-diacetyl)cytosine
    acetyl acetyl NO2 CF3 S 4-(N,N-diacetyl)cytosine
    acetyl acetyl NO2 CF3 O 4-(N,N-diacetyl)cytosine
    acetyl acetyl NO2 2-bromo- S 4-(N,N-diacetyl)cytosine
    vinyl
  • Alternatively, the following nucleosides of Formula XII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00047
  • wherein:
  • R1 R6 X Base
    H CH3 O 2,4-O-Diacetyluracil
    H CH3 O Hypoxanthine
    H CH3 O 2,4-O-Diacetylthymine
    H CH3 O Thymine
    H CH3 O Cytosine
    H CH3 O 4-(N-mono-acetyl)cytosine
    H CH3 O 4-(N,N-diacetyl)cytosine
    H CH3 O Uracil
    H CH3 O 5-Fluorouracil
    H CH3 S 2,4-O-Diacetyluracil
    H CH3 S Hypoxanthine
    H CH3 S 2,4-O-Diacetylthymine
    H CH3 S Thymine
    H CH3 S Cytosine
    H CH3 S 4-(N-mono-acetyl)cytosine
    H CH3 S 4-(N,N-diacetyl)cytosine
    H CH3 S Uracil
    H CH3 S 5-Fluorouracil
    monophosphate CH3 O 2,4-O-Diacetyluracil
    monophosphate CH3 O Hypoxanthine
    monophosphate CH3 O 2,4-O-Diacetylthymine
    monophosphate CH3 O Thymine
    monophosphate CH3 O Cytosine
    monophosphate CH3 O 4-(N-mono-acetyl)cytosine
    monophosphate CH3 O 4-(N,N-diacetyl)cytosine
    monophosphate CH3 O Uracil
    monophosphate CH3 O 5-Fluorouracil
    monophosphate CH3 S 2,4-O-Diacetyluracil
    monophosphate CH3 S Hypoxanthine
    monophosphate CH3 S 2,4-O-Diacetylthymine
    monophosphate CH3 S Thymine
    monophosphate CH3 S Cytosine
    monophosphate CH3 S 4-(N-mono-acetyl)cytosine
    monophosphate CH3 S 4-(N,N-diacetyl)cytosine
    monophosphate CH3 S Uracil
    monophosphate CH3 S 5-Fluorouracil
    diphosphate CH3 O 2,4-O-Diacetyluracil
    diphosphate CH3 O Hypoxanthine
    diphosphate CH3 O 2,4-O-Diacetylthymine
    diphosphate CH3 O Thymine
    diphosphate CH3 O Cytosine
    diphosphate CH3 O 4-(N-mono-acetyl)cytosine
    diphosphate CH3 O 4-(N,N-diacetyl)cytosine
    diphosphate CH3 O Uracil
    diphosphate CH3 O 5-Fluorouracil
    diphosphate CH3 S 2,4-O-Diacetyluracil
    diphosphate CH3 S Hypoxanthine
    diphosphate CH3 S 2,4-O-Diacetylthymine
    diphosphate CH3 S Thymine
    diphosphate CH3 S Cytosine
    triphosphate CH3 O 2,4-O-Diacetyluracil
    triphosphate CH3 O Hypoxanthine
    triphosphate CH3 O 2,4-O-Diacetylthymine
    triphosphate CH3 O Thymine
    triphosphate CH3 O Cytosine
    triphosphate CH3 O 4-(N-mono-acetyl)cytosine
    triphosphate CH3 O 4-(N,N-diacetyl)cytosine
    triphosphate CH3 O Uracil
    triphosphate CH3 O 5-Fluorouracil
    triphosphate CH3 S 2,4-O-Diacetyluracil
    triphosphate CH3 S Hypoxanthine
    triphosphate CH3 S 2,4-O-Diacetylthymine
    triphosphate CH3 S Thymine
    triphosphate CH3 S Cytosine
    monophosphate CF3 O 2,4-O-Diacetyluracil
    monophosphate CF3 O Hypoxanthine
    monophosphate CF3 O 2,4-O-Diacetylthymine
    monophosphate CF3 O Thymine
    monophosphate CF3 O Cytosine
    monophosphate CF3 O 4-(N-mono-acetyl)cytosine
    monophosphate CF3 O 4-(N,N-diacetyl)cytosine
    monophosphate CF3 O Uracil
    monophosphate CF3 O 5-Fluorouracil
    monophosphate CF3 S 2,4-O-Diacetyluracil
    monophosphate CF3 S Hypoxanthine
    monophosphate CF3 S 2,4-O-Diacetylthymine
    monophosphate CF3 S Thymine
    monophosphate CF3 S Cytosine
    monophosphate CF3 S 4-(N-mono-acetyl)cytosine
    monophosphate CF3 S 4-(N,N-diacetyl)cytosine
    monophosphate CF3 S Uracil
    monophosphate CF3 S 5-Fluorouracil
    acetyl CF3 O 4-(N,N-diacetyl)cytosine
    acetyl CF3 S 4-(N,N-diacetyl)cytosine
    acetyl 2-bromo-vinyl O 4-(N,N-diacetyl)cytosine
    acetyl 2-bromo-vinyl S 4-(N,N-diacetyl)cytosine
  • Alternatively, the following nucleosides of Formula XVII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00048
  • wherein:
  • R1 R6 R7 X Base R9 R10
    H CH3 H O 2,4-O-Diacetyluracil NHAc Me
    H CH3 H O Hypoxanthine NH2 Me
    H CH3 H O 2,4-O-Diacetylthymine NHAc Me
    H CH3 H O Thymine NH2 Me
    H CH3 H O Cytosine NH2 Me
    H CH3 H O 4-(N-mono-acetyl)cytosine NHAc Me
    H CH3 H O 4-(N,N-diacetyl)cytosine NHAc Me
    H CH3 H O Uracil NH2 Me
    H CH3 H O 5-Fluorouracil NH2 Me
    H CH3 H S 2,4-O-Diacetyluracil NHAc Me
    H CH3 H S Hypoxanthine NH2 Me
    H CH3 H S 2,4-O-Diacetylthymine NHAc Me
    H CH3 H S Thymine NH2 Me
    H CH3 H S Cytosine NH2 Me
    H CH3 H S 4-(N-mono-acetyl)cytosine NHAc Me
    H CH3 H S 4-(N,N-diacetyl)cytosine NHAc Me
    H CH3 H S Uracil NH2 Me
    H CH3 H S 5-Fluorouracil NH2 Me
    monophosphate CH3 H O 2,4-O-Diacetyluracil NHAc Me
    monophosphate CH3 H O Hypoxanthine NH2 Me
    monophosphate CH3 H O 2,4-O-Diacetylthymine NHAc Me
    monophosphate CH3 H O Thymine NH2 Me
    monophosphate CH3 H O Cytosine NH2 Me
    monophosphate CH3 H O 4-(N-mono-acetyl)cytosine NHAC Me
    monophosphate CH3 H O 4-(N,N-diacetyl)cytosine NHAc Me
    monophosphate CH3 H O Uracil NH2 Me
    monophosphate CH3 H O 5-Fluorouracil NH2 Me
    monophosphate CH3 H S 2,4-O-Diacetyluracil NHAc Me
    monophosphate CH3 H S Hypoxanthine NH2 Me
    monophosphate CH3 H S 2,4-O-Diacetylthymine NHAc Me
    monophosphate CH3 H S Thymine NH2 Me
    monophosphate CH3 H S Cytosine NH2 Me
    monophosphate CH3 H S 4-(N-mono-acetyl)cytosine NHAc Me
    monophosphate CH3 H S 4-(N,N-diacetyl)cytosine NHAc Me
    monophosphate CH3 H S Uracil NH2 Me
    monophosphate CH3 H S 5-Fluorouracil NH2 Me
    diphosphate CH3 H O 2,4-O-Diacetyluracil NHAc Me
    diphosphate CH3 H O Hypoxanthine NH2 Me
    diphosphate CH3 H O 2,4-O-Diacetylthymine NH2 Me
    diphosphate CH3 H O Thymine NH2 Me
    diphosphate CH3 H O Cytosine NH2 Me
    diphosphate CH3 H O 4-(N-mono-acetyl)cytosine NHAc Me
    diphosphate CH3 H O 4-(N,N-diacetyl)cytos NHAc Me
    diphosphate CH3 H O Uracil NH2 Me
    diphosphate CH3 H O 5-Fluorouracil NH2 Me
    diphosphate CH3 H S 2,4-O-Diacetyluracil NH2 Me
    diphosphate CH3 H S Hypoxanthine NH2 Me
    diphosphate CH3 H S 2,4-O-Diacetylthymine NHAc Me
    diphosphate CH3 H S Thymine NH2 Me
    diphosphate CH3 H S Cytosine NH2 Me
    triphosphate CH3 H O 2,4-O-Diacetyluracil NHAc Me
    triphosphate CH3 H O Hypoxanthine NHAc Me
    triphosphate CH3 H O 2,4-O-Diacetylthymine NHAc Me
    triphosphate CH3 H O Thymine NH2 Me
    triphosphate CH3 H O Cytosine NH2 Me
    triphosphate CH3 H O 4-(N-mono-acetyl)cytosine NHAc Me
    triphosphate CH3 H O 4-(N,N-diacetyl)cytosine NH2 Me
    triphosphate CH3 H O Uracil NH2 Me
    triphosphate CH3 H O 5-Fluorouracil NH2 Me
    triphosphate CH3 H S 2,4-O-Diacetyluracil NH2 Me
    triphosphate CH3 H S Hypoxanthine NH2 Me
    triphosphate CH3 H S 2,4-O-Diacetylthymine NH2 Me
    triphosphate CH3 H S Thymine NH2 Me
    triphosphate CH3 H S Cytosine NH2 Me
    monophosphate CF3 H O 2,4-O-Diacetyluracil NH2 Me
    monophosphate CF3 H O Hypoxanthine NH2 Me
    monophosphate CF3 H O 2,4-O-Diacetylthymine NH2 Me
    monophosphate CF3 H O Thymine NH2 Me
    monophosphate CF3 H O Cytosine NH2 Me
    monophosphate CF3 H O 4-(N-mono-acetyl)cytosine NH2 Me
    monophosphate CF3 H O 4-(N,N-diacetyl)cytosine NH2 Me
    monophosphate CF3 H O Uracil NH2 Me
    monophosphate CF3 H O 5-Fluorouracil NH2 Me
    monophosphate CF3 H S 2,4-O-Diacetyluracil NH2 Me
    monophosphate CF3 H S Hypoxanthine NH2 Me
    monophosphate CF3 H S 2,4-O-Diacetylthymine NH2 Me
    monophosphate CF3 H S Thymine NH2 Me
    monophosphate CF3 H S Cytosine NH2 Me
    monophosphate CF3 H S 4-(N-mono-acetyl)cytosme NH2 Me
    monophosphate CF3 H S 4-(N,N-diacetyl)cytosine NH2 Me
    monophosphate CF3 H S Uracil NH2 Me
    monophosphate CF3 H S 5-Fluorouracil NH2 Me
    acetyl CH3 H O 4-(N,N-diacetyl)cytosine H Br
    acetyl CH3 H S 4-(N,N-diacetyl)cytosine H Br
    acetyl CH3 OH O 4-(N,N-diacetyl)cytosine H Br
    acetyl CH3 OH S 4-(N,N-diacetyl)cytosine H Br
  • Example 3: Preparation of 3′-C-methylriboadenine
  • The title compound can be prepared according to a published procedure (R. F. Nutt, M. J. Dickinson, F. W. Holly, and E. Walton, “Branched-chain sugar nucleosides. III. 3′-C-methyladenine”, J. Org. Chem. 1968, 33, 1789-1795) (Scheme 9).
  • Figure US20180235993A1-20180823-C00049
  • (a) RuO2/NaIO4; (b) MeMe/TiCl4; (c) HCl/MeOH/H2O; (d) BzCl/pyridine; (e) AcBr, HBr/AcOH; (f) chloromercuri-6-benzamidopurine; (g) NH3/MeOH.
  • In a similar manner, but using the appropriate sugar and pyrimidine or purine bases, the following nucleosides of Formula III are prepared.
  • Figure US20180235993A1-20180823-C00050
  • wherein:
  • R1 R2 R3 X1 X2 Y
    H H H H H H
    H H H H H NH2
    H H H H H NH-cyclopropyl
    H H H H H NH-methyl
    H H H H H NH-ethyl
    H H H H H NH-acetyl
    H H H H H OH
    H H H H H OMe
    H H H H H OEt
    H H H H H O-cyclopropyl
    H H H H H O-acetyl
    H H H H H SH
    H H H H H SMe
    H H H H H SEt
    H H H H H S-cyclopropyl
    H H H H H F
    H H H H H Cl
    H H H H H Br
    H H H H H I
    monophosphate H H H H NH2
    monophosphate H H H H NH-acetyl
    monophosphate H H H H NH-cyclopropyl
    monophosphate H H H H NH-methyl
    monophosphate H H H H NH-ethyl
    monophosphate H H H H OH
    monophosphate H H H H O-acetyl
    monophosphate H H H H OMe
    monophosphate H H H H OEt
    monophosphate H H H H O-cyclopropyl
    monophosphate H H H H SH
    monophosphate H H H H SMe
    monophosphate H H H H SEt
    monophosphate H H H H S-cyclopropyl
    monophosphate H H H H F
    monophosphate H H H H Cl
    monophosphate H H H H Br
    monophosphate H H H H I
    diphosphate H H H H NH2
    diphosphate H H H H NH-acetyl
    diphosphate H H H H NH-cyclopropyl
    diphosphate H H H H NH-methyl
    diphosphate H H H H NH-ethyl
    diphosphate H H H H OH
    diphosphate H H H H O-acetyl
    diphosphate H H H H OMe
    diphosphate H H H H OEt
    diphosphate H H H H O-cyclopropyl
    diphosphate H H H H SH
    diphosphate H H H H SMe
    diphosphate H H H H SEt
    diphosphate H H H H S-cyclopropyl
    diphosphate H H H H F
    diphosphate H H H H Cl
    diphosphate H H H H Br
    diphosphate H H H H I
    triphosphate H H H H NH2
    triphosphate H H H H NH-acetyl
    triphosphate H H H H NH-cyclopropyl
    triphosphate H H H H NH-methyl
    triphosphate H H H H NH-ethyl
    triphosphate H H H H OH
    triphosphate H H H H OMe
    triphosphate H H H H OEt
    triphosphate H H H H O-cyclopropyl
    triphosphate H H H H O-acetyl
    triphosphate H H H H SH
    triphosphate H H H H SMe
    triphosphate H H H H SEt
    triphosphate H H H H S-cyclopropyl
    triphosphate H H H H F
    triphosphate H H H H Cl
    triphosphate H H H H Br
    triphosphate H H H H I
    monophosphate monophosphate monophosphate H H NH2
    monophosphate monophosphate monophosphate H H NH-cyclopropyl
    monophosphate monophosphate monophosphate H H OH
    monophosphate monophosphate monophosphate H H F
    monophosphate monophosphate monophosphate H H Cl
    diphosphate diphosphate diphosphate H H NH2
    diphosphate diphosphate diphosphate H H NH-cyclopropyl
    diphosphate diphosphate diphosphate H H OH
    diphosphate diphosphate diphosphate H H F
    diphosphate diphosphate diphosphate H H Cl
    triphosphate triphosphate triphosphate H H NH2
    triphosphate triphosphate triphosphate H H NH-cyclopropyl
    triphosphate triphosphate triphosphate H H OH
    triphosphate triphosphate triphosphate H H F
    triphosphate triphosphate triphosphate H H Cl
    H H H F H NH2
    H H H F H NH-cyclopropyl
    H H H F H OH
    H H H F H F
    H H H F H Cl
    H H H Cl H NH2
    H H H Cl H NH-cyclopropyl
    H H H Cl H OH
    H H H Cl H F
    H H H Cl H Cl
    H H H Br H NH2
    H H H Br H NH-cyclopropyl
    H H H Br H OH
    H H H Br H F
    H H H Br H Cl
    H H H NH2 H NH2
    H H H NH2 H NH-cyclopropyl
    H H H NH2 H OH
    H H H NH2 H F
    H H H NH2 H Cl
    H H H SH H NH2
    H H H SH H NH-cyclopropyl
    H H H SH H OH
    H H H SH H F
    H H H SH H Cl
    acetyl H H H H NH2
    acetyl H H H H NH-cyclopropyl
    acetyl H H H H OH
    acetyl H H H H F
    acetyl H H H H Cl
    acetyl H H F H NH2
    acetyl H H F H NH-cyclopropyl
    acetyl H H F H OH
    acetyl H H F H F
    acetyl H H F H Cl
    H acetyl acetyl H H NH2
    H acetyl acetyl H H NH-cyclopropyl
    H acetyl acetyl H H OH
    H acetyl acetyl H H F
    H acetyl acetyl H H Cl
    acetyl acetyl acetyl H H NH2
    acetyl acetyl acetyl H H NH-cyclopropyl
    acetyl acetyl acetyl H H OH
    acetyl acetyl acetyl H H F
    acetyl acetyl acetyl H H Cl
    monophosphate acetyl acetyl H H NH2
    monophosphate acetyl acetyl H H NH-cyclopropyl
    monophosphate acetyl acetyl H H OH
    monophosphate acetyl acetyl H H F
    monophosphate acetyl acetyl H H Cl
    diphosphate acetyl acetyl H H NH2
    diphosphate acetyl acetyl H H NH-cyclopropyl
    diphosphate acetyl acetyl H H OH
    diphosphate acetyl acetyl H H F
    diphosphate acetyl acetyl H H Cl
    triphosphate acetyl acetyl H H NH2
    triphosphate acetyl acetyl H H NH-cyclopropyl
    triphosphate acetyl acetyl H H OH
    triphosphate acetyl acetyl H H F
    triphosphate acetyl acetyl H H Cl
    H H H H NH2 H
    H H H H NH2 NH2
    H H H H NH2 NH-cyclopropyl
    H H H H NH2 NH-methyl
    H H H H NH2 NH-ethyl
    H H H H NH2 NH-acetyl
    H H H H NH2 OH
    H H H H NH2 OMe
    H H H H NH2 OEt
    H H H H NH2 O-cyclopropyl
    H H H H NH2 O-acetyl
    H H H H NH2 SH
    H H H H NH2 SMe
    H H H H NH2 SEt
    H H H H NH2 S-cyclopropyl
    H H H H NH2 F
    H H H H NH2 Cl
    H H H H NH2 Br
    H H H H NH2 I
    monophosphate H H H NH2 NH2
    monophosphate H H H NH2 NH-acetyl
    monophosphate H H H NH2 NH-cyclopropyl
    monophosphate H H H NH2 NH-methyl
    monophosphate H H H NH2 NH-ethyl
    monophosphate H H H NH2 OH
    monophosphate H H H NH2 O-acetyl
    monophosphate H H H NH2 OMe
    monophosphate H H H NH2 OEt
    monophosphate H H H NH2 O-cyclopropyl
    monophosphate H H H NH2 SH
    monophosphate H H H NH2 SMe
    monophosphate H H H NH2 SEt
    monophosphate H H H NH2 S-cyclopropyl
    monophosphate H H H NH2 F
    monophosphate H H H NH2 Cl
    monophosphate H H H NH2 Br
    monophosphate H H H NH2 I
    diphosphate H H H NH2 NH2
    diphosphate H H H NH2 NH-acetyl
    diphosphate H H H NH2 NH-cyclopropyl
    diphosphate H H H NH2 NH-methyl
    diphosphate H H H NH2 NH-ethyl
    diphosphate H H H NH2 OH
    diphosphate H H H NH2 O-acetyl
    diphosphate H H H NH2 OMe
    diphosphate H H H NH2 OEt
    diphosphate H H H NH2 O-cyclopropyl
    diphosphate H H H NH2 SH
    diphosphate H H H NH2 SMe
    diphosphate H H H NH2 SEt
    diphosphate H H H NH2 S-cyclopropyl
    diphosphate H H H NH2 F
    diphosphate H H H NH2 Cl
    diphosphate H H H NH2 Br
    diphosphate H H H NH2 I
    triphosphate H H H NH2 NH2
    triphosphate H H H NH2 NH-acetyl
    triphosphate H H H MH2 NH-cyclopropyl
    triphosphate H H H NH2 NH-methyl
    triphosphate H H H NH2 NH-ethyl
    triphosphate H H H NH2 OH
    triphosphate H H H NH2 OMe
    triphosphate H H H NH2 OEt
    triphosphate H H H NH2 O-cyclopropyl
    triphosphate H H H NH2 O-acetyl
    triphosphate H H H NH2 SH
    triphosphate H H H NH2 SMe
    triphosphate H H H NH2 SEt
    triphosphate H H H NH2 S-cyclopropyl
    triphosphate H H H NH2 F
    triphosphate H H H NH2 Cl
    triphosphate H H H NH2 Br
    triphosphate H H H NH2 I
    monophosphate monophosphate monophosphate H NH2 NH2
    monophosphate monophosphate monophosphate H NH2 NH-cyclopropyl
    monophosphate monophosphate monophosphate H NH2 OH
    monophosphate monophosphate monophosphate H NH2 F
    monophosphate monophosphate monophosphate H NH2 Cl
    diphosphate diphosphate diphosphate H NH2 NH2
    diphosphate diphosphate diphosphate H NH2 NH-cyclopropyl
    diphosphate diphosphate diphosphate H NH2 OH
    diphosphate diphosphate diphosphate H NH2 F
    diphosphate diphosphate diphosphate H NH2 Cl
    triphosphate triphosphate triphosphate H NH2 NH2
    triphosphate triphosphate triphosphate H NH2 NH-cyclopropyl
    triphosphate triphosphate triphosphate H NH2 OH
    triphosphate triphosphate triphosphate H NH2 F
    triphosphate triphosphate triphosphate H NH2 Cl
    H H H F NH2 NH2
    H H H F NH2 NH-cyclopropyl
    H H H F NH2 OH
    H H H F NH2 F
    H H H F NH2 Cl
    H H H Cl NH2 NH2
    H H H Cl NH2 NH-cyclopropyl
    H H H Cl NH2 OH
    H H H Cl NH2 F
    H H H Cl NH2 Cl
    H H H Br NH2 NH2
    H H H Br NH2 NH-cyclopropyl
    H H H Br NH2 OH
    H H H Br NH2 F
    H H H Br NH2 Cl
    H H H NH2 NH2 NH2
    H H H NH2 NH2 NH-cyclopropyl
    H H H NH2 NH2 OH
    H H H NH2 NH2 F
    H H H NH2 NH2 Cl
    H H H SH NH2 NH2
    H H H SH NH2 NH-cyclopropyl
    H H H SH NH2 OH
    H H H SH NH2 F
    H H H SH NH2 Cl
    acetyl H H H NH2 NH2
    acetyl H H H NH2 NH-cyclopropyl
    acetyl H H H NH2 OH
    acetyl H H H NH2 F
    acetyl H H H NH2 Cl
    acetyl H H F NH2 NH2
    acetyl H H F NH2 NH-cyclopropyl
    acetyl H H F NH2 OH
    acetyl H H F NH2 F
    acetyl H H F NH2 Cl
    H acetyl acetyl H NH2 NH2
    H acetyl acetyl H NH2 NH-cyclopropyl
    H acetyl acetyl H NH2 OH
    H acetyl acetyl H NH2 F
    H acetyl acetyl H NH2 Cl
    acetyl acetyl acetyl H NH2 NH2
    acetyl acetyl acetyl H NH2 NH-cyclopropyl
    acetyl acetyl acetyl H NH2 OH
    acetyl acetyl acetyl H NH2 F
    acetyl acetyl acetyl H NH2 Cl
    monophosphate acetyl acetyl H NH2 NH2
    monophosphate acetyl acetyl H NH2 NH-cyclopropyl
    monophosphate acetyl acetyl H NH2 OH
    monophosphate acetyl acetyl H NH2 F
    monophosphate acetyl acetyl H NH2 Cl
    diphosphate acetyl acetyl H NH2 NH2
    diphosphate acetyl acetyl H NH2 NH-cyclopropyl
    diphosphate acetyl acetyl H NH2 OH
    diphosphate acetyl acetyl H NH2 F
    diphosphate acetyl acetyl H NH2 Cl
    triphosphate acetyl acetyl H NH2 NH2
    triphosphate acetyl acetyl H NH2 NH-cyclopropyl
    triphosphate acetyl acetyl H NH2 OH
    triphosphate acetyl acetyl H NH2 F
    triphosphate acetyl acetyl H NH2 Cl
    H H H H Cl H
    H H H H Cl H
    H H H H Cl NH2
    H H H H Cl NH-cyclopropyl
    H H H H Cl NH-methyl
    H H H H Cl NH-ethyl
    H H H H Cl NH-acetyl
    H H H H Cl OH
    H H H H Cl OMe
    H H H H Cl OEt
    H H H H Cl O-cyclopropyl
    H H H H Cl O-acetyl
    H H H H Cl SH
    H H H H Cl SMe
    H H H H Cl SEt
    H H H H Cl S-cyclopropyl
    monophosphate H H H Cl NH2
    monophosphate H H H Cl NH-acetyl
    monophosphate H H H Cl NH-cyclopropyl
    monophosphate H H H Cl NH-methyl
    monophosphate H H H Cl NH-ethyl
    monophosphate H H H Cl OH
    monophosphate H H H Cl O-acetyl
    monophosphate H H H Cl OMe
    monophosphate H H H Cl OEt
    monophosphate H H H Cl O-cyclopropyl
    monophosphate H H H Cl SH
    monophosphate H H H Cl SMe
    monophosphate H H H Cl SEt
    monophosphate H H H Cl S-cyclopropyl
    diphosphate H H H Cl NH2
    diphosphate H H H Cl NH-acetyl
    diphosphate H H H Cl NH-cyclopropyl
    diphosphate H H H Cl NH-methyl
    diphosphate H H H Cl NH-ethyl
    diphosphate H H H Cl OH
    diphosphate H H H Cl O-acetyl
    diphosphate H H H Cl OMe
    diphosphate H H H Cl OEt
    diphosphate H H H Cl O-cyclopropyl
    diphosphate H H H Cl SH
    diphosphate H H H Cl SMe
    diphosphate H H H Cl SEt
    diphosphate H H H Cl S-cyclopropyl
    triphosphate H H H Cl NH2
    triphosphate H H H Cl NH-acetyl
    triphosphate H H H Cl NH-cyclopropyl
    triphosphate H H H Cl NH-methyl
    triphosphate H H H Cl NH-ethyl
    triphosphate H H H Cl OH
    triphosphate H H H Cl OMe
    triphosphate H H H Cl OEt
    triphosphate H H H Cl O-cyclopropyl
    triphosphate H H H Cl O-acetyl
    triphosphate H H H Cl SH
    triphosphate H H H Cl SMe
    triphosphate H H H Cl SEt
    triphosphate H H H Cl S-cyclopropyl
    monophosphate monophosphate monophosphate H Cl NH2
    monophosphate monophosphate monophosphate H Cl NH-cyclopropyl
    monophosphate monophosphate monophosphate H Cl OH
    diphosphate diphosphate diphosphate H Cl NH2
    diphosphate diphosphate diphosphate H Cl NH-cyclopropyl
    diphosphate diphosphate diphosphate H Cl OH
    triphosphate triphosphate triphosphate H Cl NH2
    triphosphate triphosphate triphosphate H Cl NH-cyclopropyl
    triphosphate triphosphate triphosphate H Cl OH
    H H H F Cl NH2
    H H H F Cl NH-cyclopropyl
    H H H F Cl OH
    H H H Cl Cl NH2
    H H H Cl Cl NH-cyclopropyl
    H H H Cl Cl OH
    H H H Br Cl NH2
    H H H Br Cl NH-cyclopropyl
    H H H Br Cl OH
    H H H NH2 Cl NH2
    H H H NH2 Cl NH-cyclopropyl
    H H H NH2 Cl OH
    H H H SH Cl NH2
    H H H SH Cl NH-cyclopropyl
    H H H SH Cl OH
    acetyl H H H Cl NH2
    acetyl H H H Cl NH-cyclopropyl
    acetyl H H H Cl OH
    acetyl H H F Cl NH2
    acetyl H H F Cl NH-cyclopropyl
    acetyl H H F Cl OH
    H acetyl acetyl H Cl NH2
    H acetyl acetyl H Cl NH-cyclopropyl
    H acetyl acetyl H Cl OH
    acetyl acetyl acetyl H Cl NH2
    acetyl acetyl acetyl H Cl NH-cyclopropyl
    acetyl acetyl acetyl H Cl OH
    monophosphate acetyl acetyl H Cl NH2
    monophosphate acetyl acetyl H Cl NH-cyclopropyl
    monophosphate acetyl acetyl H Cl OH
    diphosphate acetyl acetyl H Cl NH2
    diphosphate acetyl acetyl H Cl NH-cyclopropyl
    diphosphate acetyl acetyl H Cl OH
    triphosphate acetyl acetyl H Cl NH2
    triphosphate acetyl acetyl H Cl NH-cyclopropyl
    triphosphate acetyl acetyl H Cl OH
    H H H H Cl NH2
    H H H H Cl NH-cyclopropyl
    H H H H Cl OH
    H H H H Br NH2
    H H H H Br NH-cyclopropyl
    H H H H Br OH
  • Alternatively, the following nucleosides of Formula VI are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00051
  • wherein:
  • R1 R2 R3 X1 Y
    H H H H H
    H H H H NH2
    H H H H NH-cyclopropyl
    H H H H NH-methyl
    H H H H NH-ethyl
    H H H H NH-acetyl
    H H H H OH
    H H H H OMe
    H H H H OEt
    H H H H O-cyclopropyl
    H H H H O-acetyl
    H H H H SH
    H H H H SMe
    H H H H SEt
    H H H H S-cyclopropyl
    monophosphate H H H NH2
    monophosphate H H H NH-acetyl
    monophosphate H H H NH-cyclopropyl
    monophosphate H H H NH-methyl
    monophosphate H H H NH-ethyl
    monophosphate H H H OH
    monophosphate H H H O-acetyl
    monophosphate H H H OMe
    monophosphate H H H OEt
    monophosphate H H H O-cyclopropyl
    monophosphate H H H SH
    monophosphate H H H SMe
    monophosphate H H H SEt
    monophosphate H H H S-cyclopropyl
    diphosphate H H H NH2
    diphosphate H H H NH-acetyl
    diphosphate H H H NH-cyclopropyl
    diphosphate H H H NH-methyl
    diphosphate H H H NH-ethyl
    diphosphate H H H OH
    diphosphate H H H O-acetyl
    diphosphate H H H OMe
    diphosphate H H H OEt
    diphosphate H H H O-cyclopropyl
    diphosphate H H H SH
    diphosphate H H H SMe
    diphosphate H H H SEt
    diphosphate H H H S-cyclopropyl
    triphosphate H H H NH2
    triphosphate H H H NH-acetyl
    triphosphate H H H NH-cyclopropyl
    triphosphate H H H NH-methyl
    triphosphate H H H NH-ethyl
    triphosphate H H H OH
    triphosphate H H H OMe
    triphosphate H H H OEt
    triphosphate H H H O-cyclopropyl
    triphosphate H H H O-acetyl
    triphosphate H H H SH
    triphosphate H H H SMe
    triphosphate H H H SEt
    triphosphate H H H S-cyclopropyl
    monophosphate monophosphate monophosphate H NH2
    monophosphate monophosphate monophosphate H NH-cyclopropyl
    monophosphate monophosphate monophosphate H OH
    diphosphate diphosphate diphosphate H NH2
    diphosphate diphosphate diphosphate H NH-cyclopropyl
    diphosphate diphosphate diphosphate H OH
    triphosphate triphosphate triphosphate H NH2
    triphosphate triphosphate triphosphate H NH-cyclopropyl
    triphosphate triphosphate triphosphate H OH
    H H H F NH2
    H H H F NH-cyclopropyl
    H H H F OH
    H H H Cl NH2
    H H H Cl NH-cyclopropyl
    H H H Cl OH
    H H H Br NH2
    H H H Br NH-cyclopropyl
    H H H Br OH
    H H H NH2 NH2
    H H H NH2 NH-cyclopropyl
    H H H NH2 OH
    H H H SH NH2
    H H H SH NH-cyclopropyl
    H H H SH OH
    acetyl H H H NH2
    acetyl H H H NH-cyclopropyl
    acetyl H H H OH
    acetyl H H F NH2
    acetyl H H F NH-cyclopropyl
    acetyl H H F OH
    H acetyl acetyl H NH2
    H acetyl acetyl H NH-cyclopropyl
    H acetyl acetyl H OH
    acetyl acetyl acetyl H NH2
    acetyl acetyl acetyl H NH-cyclopropyl
    acetyl acetyl acetyl H OH
    monophosphate acetyl acetyl H NH2
    monophosphate acetyl acetyl H NH-cyclopropyl
    monophosphate acetyl acetyl H OH
    diphosphate acetyl acetyl H NH2
    diphosphate acetyl acetyl H NH-cyclopropyl
    diphosphate acetyl acetyl H OH
    triphosphate acetyl acetyl H NH2
    triphosphate acetyl acetyl H NH-cyclopropyl
    triphosphate acetyl acetyl H OH
  • Alternatively, the following nucleosides of Formula XIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00052
  • wherein:
  • R1 R2 R3 R6 X Base
    H H H CH3 O 2,4-O-
    Diacetyluracil
    H H H CH3 O Hypoxanthine
    H H H CH3 O 2,4-O-
    Diacetylthymine
    H H H CH3 O Thymine
    H H H CH3 O Cytosine
    H H H CH3 O 4-(N-mono-
    acetyl)cytosine
    H H H CH3 O 4-(N,N-
    diacetyl)cytosine
    H H H CH3 O Uracil
    H H H CH3 O 5-Fluorouracil
    H H H CH3 S 2,4-O-
    Diacetyluraci
    H H H CH3 S Hypoxanthine
    H H H CH3 S 2,4-O-
    Diacetylthymine
    H H H CH3 S Thymine
    H H H CH3 S Cytosine
    H H H CH3 S 4-(N-mono-
    acetyl)cytosine
    H H H CH3 S 4-(N,N-
    diacetyl)cytosine
    H H H CH3 S Uracil
    H H H CH3 S 5-Fluorouracil
    monophosphate H H CH3 O 2,4-O-
    Diacetyluracil
    monophosphate H H CH3 O Hypoxanthine
    monophosphate H H CH3 O 2,4-O-
    Diacetylthym
    monophosphate H H CH3 O Thymine
    monophosphate H H CH3 O Cytosine
    monophosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    monophosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    monophosphate H H CH3 O Uracil
    monophosphate H H CH3 O 5-Fluorouracil
    monophosphate H H CH3 S 2,4-O-
    Diacetyluracil
    monophosphate H H CH3 S Hypoxanthine
    monophosphate H H CH3 S 2,4-O-
    Diacetylthym
    monophosphate H H CH3 S Thymine
    monophosphate H H CH3 S Cytosine
    monophosphate H H CH3 S 4-(N-mono-
    acetyl)cytosine
    monophosphate H H CH3 S 4-(N,N-
    diacetyl)cytosine
    monophosphate H H CH3 S Uracil
    monophosphate H H CH3 S 5-Fluorouracil
    diphosphate H H CH3 O 2,4-O-
    Diacetyluracil
    diphosphate H H CH3 O Hypoxanthine
    diphosphate H H CH3 O 2,4-O-
    Diacetylthymine
    diphosphate H H CH3 O Thymine
    diphosphate H H CH3 O Cytosine
    diphosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    diphosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    diphosphate H H CH3 O Uracil
    diphosphate H H CH3 O 5-Fluorouracil
    diphosphate H H CH3 S 2,4-O-
    Diacetyluracil
    diphosphate H H CH3 S Hypoxanthine
    diphosphate H H CH3 S 2,4-O-
    Diacetylthym
    diphosphate H H CH3 S Thymine
    diphosphate H H CH3 S Cytosine
    triphosphate H H CH3 O 2,4-O-
    Diacetyluracil
    triphosphate H H CH3 O Hypoxanthine
    triphosphate H H CH3 O 2,4-O-
    Diacetylthymine
    triphosphate H H CH3 O Thymine
    triphosphate H H CH3 O Cytosine
    triphosphate H H CH3 O 4-(N-mono-
    acetyl)cytosine
    triphosphate H H CH3 O 4-(N,N-
    diacetyl)cytosine
    triphosphate H H CH3 O Uracil
    triphosphate H H CH3 O 5-Fluorouracil
    triphosphate H H CH3 S 2,4-O-
    Diacetyluracil
    triphosphate H H CH3 S Hypoxanthine
    triphosphate H H CH3 S 2,4-O-
    Diacetylthymine
    triphosphate H H CH3 S Thymine
    triphosphate H H CH3 S Cytosine
    monophosphate monophosphate monophosphate CF3 O 2,4-O-
    Diacetyluracil
    monophosphate monophosphate monophosphate CF3 O Hypoxanthine
    monophosphate monophosphate monophosphate CF3 O 2,4-O-
    Diacetylthymine
    monophosphate monophosphate monophosphate CF3 O Thymine
    monophosphate monophosphate monophosphate CF3 O Cytosine
    monophosphate monophosphate monophosphate CF3 O 4-(N-mono-
    acetyl)cytosine
    monophosphate monophosphate monophosphate CF3 O 4-(N,N-
    diacetyl)cytosine
    monophosphate monophosphate monophosphate CF3 O Uracil
    monophosphate monophosphate monophosphate CF3 O 5-Fluorouracil
    monophosphate monophosphate monophosphate CF3 S 2,4-O-
    Diacetyluracil
    monophosphate monophosphate monophosphate CF3 S Hypoxanthine
    monophosphate monophosphate monophosphate CF3 S 2,4-O-
    Diacetylthymine
    monophosphate monophosphate monophosphate CF3 S Thymine
    monophosphate monophosphate monophosphate CF3 S Cytosine
    monophosphate monophosphate monophosphate CF3 S 4-(N-mono-
    acetyl)cytosine
    monophosphate monophosphate monophosphate CF3 S 4-(N,N-
    diacetyl)cytosine
    monophosphate monophosphate monophosphate CF3 S Uracil
    monophosphate monophosphate monophosphate CF3 S 5-Fluorouracil
    acetyl acetyl acetyl CF3 O 4-(N,N-
    diacetyl)cytosine
    acetyl acetyl acetyl CF3 S 4-(N,N-
    diacetyl)cytosine
    acetyl acetyl acetyl 2-bromo- O 4-(N,N-
    vinyl diacetyl)cytosine
    acetyl acetyl acetyl 2-bromo- S 4-(N,N-
    vinyl diacetyl)cytosine
    H H H CH3 O 2-(N,N-diacetyl)-
    guanine
    H H H CH3 O 6-O-acetyl
    guanine
    H H H CH3 O 8-fluoroguanine
    H H H CH3 O guanine
    H H H CH3 O 6-(N,N-diacetyl)-
    adenine
    H H H CH3 O 2-fluoroadenine
    H H H CH3 O 8-fluoroadenine
    H H H CH3 O 2,8-difluoro-
    adenine
    H H H CH3 O adenine
    H H H CH3 S 2-(N,N-diacetyl)-
    guanine
    H H H CH3 S 6-O-acetyl
    guanine
    H H H CH3 S 8-fluoroguanine
    H H H CH3 S guanine
    H H H CH3 S 6-(N,N-diacetyl)-
    adenine
    H H H CH3 S 2-fluoroadenine
    H H H CH3 S 8-fluoroadenine
    H H H CH3 S 2,8-difluoro-
    adenine
    H H H CH3 S adenine
    monophosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    monophosphate H H CH3 O 6-O-acetyl
    guanine
    monophosphate H H CH3 O 8-fluoroguanine
    monophosphate H H CH3 O guanine
    monophosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    monophosphate H H CH3 O 2-fluoroadenine
    monophosphate H H CH3 O 8-fluoroadenine
    monophosphate H H CH3 O 2,8-difluoro-
    adenine
    monophosphate H H CH3 O adenine
    monophosphate H H CH3 S 2-(N,N-diacetyl)-
    guanine
    monophosphate H H CH3 S 6-O-acetyl
    guanine
    monophosphate H H CH3 S 8-fluoroguanine
    monophosphate H H CH3 S guanine
    monophosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    monophosphate H H CH3 S 2-fluoroadenine
    monophosphate H H CH3 S 8-fluoroadenine
    monophosphate H H CH3 S 2,8-difluoro-
    adenine
    monophosphate H H CH3 S adenine
    diphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    diphosphate H H CH3 O 6-O-acetyl
    guanine
    diphosphate H H CH3 O 8-fluoroguanine
    diphosphate H H CH3 O guanine
    diphosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    diphosphate H H CH3 O 2-fluoroadenine
    diphosphate H H CH3 O 8-fluoroadenine
    diphosphate H H CH3 O 2,8-difluoro-
    adenine
    diphosphate H H CH3 O adenine
    diphosphate H H CH3 S 2-(N,N-diacetyl)-
    guanine
    diphosphate H H CH3 S 6-O-acetyl
    guanine
    diphosphate H H CH3 S 8-fluoroguanine
    diphosphate H H CH3 S guanine
    diphosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    diphosphate H H CH3 S 2-fluoroadenine
    diphosphate H H CH3 S 8-fluoroadenine
    diphosphate H H CH3 S 2,8-difluoro-
    adenine
    diphosphate H H CH3 S adenine
    triphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    triphosphate H H CH3 O 6-O-acetyl
    guanine
    triphosphate H H CH3 O 8-fluoroguanine
    triphosphate H H CH3 O guanine
    triphosphate H H CH3 O 6-(N,N-diacetyl)-
    adenine
    triphosphate H H CH3 O 2-fluoroadenine
    triphosphate H H CH3 O 8-fluoroadenine
    triphosphate H H CH3 O 2,8-difluoro-
    adenine
    triphosphate H H CH3 O 2-(N,N-diacetyl)-
    guanine
    triphosphate H H CH3 S 6-O-acetyl
    guanine
    triphosphate H H CH3 S 8-fluoroguanine
    triphosphate H H CH3 S guanine
    triphosphate H H CH3 S 6-(N,N-diacetyl)-
    adenine
    triphosphate H H CH3 S 2-fluoroadenine
    triphosphate H H CH3 S 8-fluoroadenine
    triphosphate H H CH3 S 2,8-difluoro-
    adenine
    triphosphate H H CH3 S adenine
    monophosphate monophosphate monophosphate CF3 O 2-(N,N-diacetyl)-
    guanine
    monophosphate monophosphate monophosphate CF3 O 6-O-acetyl
    guanine
    monophosphate monophosphate monophosphate CF3 O 8-fluoroguanine
    monophosphate monophosphate monophosphate CF3 O guanine
    monophosphate monophosphate monophosphate CF3 O 6-(N,N-diacetyl)-
    adenine
    monophosphate monophosphate monophosphate CF3 O 2-fluoroadenine
    monophosphate monophosphate monophosphate CF3 O 8-fluoroadenine
    monophosphate monophosphate monophosphate CF3 O 2,8-difluoro-
    adenine
    monophosphate monophosphate monophosphate CF3 O adenine
    monophosphate monophosphate monophosphate CF3 S 2-(N,N-diacetyl)-
    guanine
    monophosphate monophosphate monophosphate CF3 S 6-O-acetyl
    guanine
    monophosphate monophosphate monophosphate CF3 S 8-fluoroguanine
    monophosphate monophosphate monophosphate CF3 S guanine
    monophosphate monophosphate monophosphate CF3 S 6-(N,N-diacetyl)-
    adenine
    monophosphate monophosphate monophosphate CF3 S 2-fluoroadenine
    monophosphate monophosphate monophosphate CF3 S 8-fluoroadenine
    monophosphate monophosphate monophosphate CF3 S 2,8-difluoro-
    adenine
    monophosphate monophosphate monophosphate CF3 S adenine
    acetyl acetyl acetyl CF3 O guanine
    acetyl acetyl acetyl CF3 S guanine
    acetyl acetyl acetyl 2-bromo- O guanine
    vinyl
    acetyl acetyl acetyl 2-bromo- S guanine
    vinyl
  • Alternatively, the following nucleosides of Formula XIV are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00053
  • wherein:
  • R1 R2 R6 X Base
    H H CH3 O 2,4-O-Diacetyluracil
    H H CH3 O Hypoxanthine
    H H CH3 O 2,4-O-Diacetylthymine
    H H CH3 O Thymine
    H H CH3 O Cytosine
    H H CH3 O 4-(N-mono-acetyl)cytosine
    H H CH3 O 4-(N,N-diacetyl)cytosine
    H H CH3 O Uracil
    H H CH3 O 5-Fluorouracil
    H H CH3 S 2,4-O-Diacetyluracil
    H H CH3 S Hypoxanthine
    H H CH3 S 2,4-O-Diacetylthymine
    H H CH3 S Thymine
    H H CH3 S Cytosine
    H H CH3 S 4-(N-mono-acetyl)cytosin
    H H CH3 S 4-(N,N-diacetyl)cytosine
    H H CH3 S Uracil
    H H CH3 S 5-Fluorouracil
    monophosphate H CH3 O 2,4-O-Diacetyluracil
    monophosphate H CH3 O Hypoxanthine
    monophosphate H CH3 O 2,4-O-Diacetylthym
    monophosphate H CH3 O Thymine
    monophosphate H CH3 O Cytosine
    monophosphate H CH3 O 4-(N-mono-acetyl)cytosine
    monophosphate H CH3 O 4-(N,N-diacetyl)cytos
    monophosphate H CH3 O Uracil
    monophosphate H CH3 O 5-Fluorouracil
    monophosphate H CH3 S 2,4-O-Diacetyluracil
    monophosphate H CH3 S Hypoxanthine
    monophosphate H CH3 S 2,4-O-Diacetylthyrn
    monophosphate H CH3 S Thymine
    monophosphate H CH3 S Cytosine
    monophosphate H CH3 S 4-(N-mono-acetyl)cytosine
    monophosphate H CH3 S 4-(N,N-diacetyl)cytosine
    monophosphate H CH3 S Uracil
    monophosphate H CH3 S 5-Fluorouracil
    diphosphate H CH3 O 2,4-O-Diacetyluracil
    diphosphate H CH3 O Hypoxanthine
    diphosphate H CH3 O 2,4-O-Diacetylthymine
    diphosphate H CH3 O Thymine
    diphosphate H CH3 O Cytosine
    diphosphate H CH3 O 4-(N-mono-acetyl)cytosine
    diphosphate H CH3 O 4-(N,N-diacetyl)cytosine
    diphosphate H CH3 O Uracil
    diphosphate H CH3 O 5-Fluorouracil
    diphosphate H CH3 S 2,4-O-Diacetyluracil
    diphosphate H CH3 S Hypoxanthine
    diphosphate H CH3 S 2,4-O-Diacetylthymine
    diphosphate H CH3 S Thymine
    diphosphate H CH3 S Cytosine
    triphosphate H CH3 O 2,4-O-Diacetyluracil
    triphosphate H CH3 O Hypoxanthine
    triphosphate H CH3 O 2,4-O-Diacetylthymine
    triphosphate H CH3 O Thymine
    triphosphate H CH3 O Cytosine
    triphosphate H CH3 O 4-(N-mono-acetyl)cytosine
    triphosphate H CH3 O 4-(N,N-diacetyl)cytosine
    triphosphate H CH3 O Uracil
    triphosphate H CH3 O 5-Fluorouracil
    triphosphate H CH3 S 2,4-O-Diacetyluracil
    triphosphate H CH3 S Hypoxanthine
    triphosphate H CH3 S 2,4-O-Diacetylthymine
    triphosphate H CH3 S Thymine
    triphosphate H CH3 S Cytosine
    monophosphate monophosphate CF3 O 2,4-O-Diacetyluracil
    monophosphate monophosphate CF3 O Hypoxanthine
    monophosphate monophosphate CF3 O 2,4-O-Diacetylthymine
    monophosphate monophosphate CF3 O Thymine
    monophosphate monophosphate CF3 O Cytosine
    monophosphate monophosphate CF3 O 4-(N-mono-acetyl)cytosine
    monophosphate monophosphate CF3 O 4-(N,N-diacetyl)cytosine
    monophosphate monophosphate CF3 O Uracil
    monophosphate monophosphate CF3 O 5-Fluorouracil
    monophosphate monophosphate CF3 S 2,4-O-Diacetyluracil
    monophosphate monophosphate CF3 S Hypoxanthine
    monophosphate monophosphate CF3 S 2,4-O-Diacetylthymine
    monophosphate monophosphate CF3 S Thymine
    monophosphate monophosphate CF3 S Cytosine
    monophosphate monophosphate CF3 S 4-(N-mono-acetyl)cytosine
    monophosphate monophosphate CF3 S 4-(N,N-diacetyl)cytosine
    monophosphate monophosphate CF3 S Uracil
    monophosphate monophosphate CF3 S 5-Fluorouracil
    acetyl acetyl CF 3 0 4-(N,N-diacetyl)cytosine
    acetyl acetyl CF3 S 4-(N,N-diacetyl)cytosine
    acetyl acetyl 2-bromo- O 4-(N,N-diacetyl)cytosine
    vinyl
    acetyl acetyl 2-bromo- S 4-(N,N-diacetyl)cytosine
    vinyl
  • Alternatively, the following nucleosides of Formula XV are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00054
  • wherein:
  • R1 R6 X Base
    H CH3 O 2,4-O-Diacetyluracil
    H CH3 O Hypoxanthine
    H CH3 O 2,4-O-Diacetylthymine
    H CH3 O Thymine
    H CH3 O Cytosine
    H CH3 O 4-(N-mono-acetyl)cytosine
    H CH3 O 4-(N,N-diacetyl)cytosine
    H CH3 O Uracil
    H CH3 O 5-Fluorouracil
    H CH3 S 2,4-O-Diacetyluracil
    H CH3 S Hypoxanthine
    H CH3 S 2,4-O-Diacetylthymine
    H CH3 S Thymine
    H CH3 S Cytosine
    H CH3 S 4-(N-mono-acetyl)cytosine
    H CH3 S 4-(N,N-diacetyl)cytosine
    H CH3 S Uracil
    H CH3 S 5-Fluorouracil
    monophosphate CH3 O 2,4-O-Diacetyluracil
    monophosphate CH3 O Hypoxanthine
    monophosphate CH3 O 2,4-O-Diacetylthymine
    monophosphate CH3 O Thymine
    monophosphate CH3 O Cytosine
    monophosphate CH3 O 4-(N-mono-acetyl)cytosine
    monophosphate CH3 O 4-(N,N-diacetyl)cytosine
    monophosphate CH3 O Uracil
    monophosphate CH3 O 5-Fluorouracil
    monophosphate CH3 S 2,4-O-Diacetyluracil
    monophosphate CH3 S Hypoxanthine
    monophosphate CH3 S 2,4-O-Diacetylthymine
    monophosphate CH3 S Thymine
    monophosphate CH3 S Cytosine
    monophosphate CH3 S 4-(N-mono-acety1)cytosine
    monophosphate CH3 S 4-(N,N-diacetyl)cytosine
    monophosphate CH3 S Uracil
    monophosphate CH3 S 5-Fluorouracil
    diphosphate CH3 O 2,4-O-Diacetyluracil
    diphosphate CH3 O Hypoxanthine
    diphosphate CH3 O 2,4-O-Diacetylthymine
    diphosphate CH3 O Thymine
    diphosphate CH3 O Cytosine
    diphosphate CH3 O 4-(N-mono-acetyl)cytosine
    diphosphate CH3 O 4-(N,N-diacetyl)cytosine
    diphosphate CH3 O Uracil
    diphosphate CH3 O 5-Fluorouracil
    diphosphate CH3 S 2,4-O-Diacetyluracil
    diphosphate CH3 S Hypoxanthine
    diphosphate CH3 S 2,4-O-Diacetylthymine
    diphosphate CH3 S Thymine
    diphosphate CH3 S Cytosine
    triphosphate CH3 O 2,4-O-Diacetyluracil
    triphosphate CH3 O Hypoxanthine
    triphosphate CH3 O 2,4-O-Diacetylthymine
    triphosphate CH3 O Thymine
    triphosphate CH3 O Cytosine
    triphosphate CH3 O 4-(N-mono-acetyl)cytosine
    triphosphate CH3 O 4-(N,N-diacetyl)cytosine
    triphosphate CH3 O Uracil
    triphosphate CH3 O 5-Fluorouracil
    triphosphate CH3 S 2,4-O-Diacetyluracil
    triphosphate CH3 S Hypoxanthine
    triphosphate CH3 S 2,4-O-Diacetylthymine
    triphosphate CH3 S Thymine
    triphosphate CH3 S Cytosine
    monophosphate CF3 O 2,4-O-Diacetyluracil
    monophosphate CF3 O Hypoxanthine
    monophosphate CF3 O 2,4-0-Diacetylthymine
    monophosphate CF3 O Thymine
    monophosphate CF3 O Cytosine
    monophosphate CF3 O 4-(N-mono-acetyl)cytosine
    monophosphate CF3 O 4-(N,N-diacetyl)cytosine
    monophosphate CF3 O Uracil
    monophosphate CF3 O 5-Fluorouracil
    monophosphate CF3 S 2,4-O-Diacetyluracil
    monophosphate CF3 S Hypoxanthine
    monophosphate CF3 S 2,4-O-Diacetylthymine
    monophosphate CF3 S Thymine
    monophosphate CF3 S Cytosine
    monophosphate CF3 S 4-(N-mono-acetyl)cytosine
    monophosphate CF3 S 4-(N,N-diacetyl)cytosine
    monophosphate CF3 S Uracil
    monophosphate CF3 S 5-Fluorouracil
    acetyl CF3 O 4-(N,N-diacetyl)cytosine
    acetyl CF3 S 4-(N,N-diacetyl)cytosine
    acetyl 2-bromo-vinyl O 4-(N,N-diacetyl)cytosine
    acetyl 2-bromo-vinyl S 4-(N,N-diacetyl)cytosine
  • Alternatively, the following nucleosides of Formula XVIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Figure US20180235993A1-20180823-C00055
  • wherein:
  • R1 R6 R7 X Base R8 R9
    H CH3 OH O 2,4-O-Diacetyluracil H Me
    H CH3 OH O Hypoxanthine H Me
    H CH3 OH O 2,4-O-Diacetylthymine H Me
    H CH3 OH O Thymine H Me
    H CH3 OH O Cytosine H Me
    H CH3 OH O 4-(N-mono-acetyl)cytosine H Me
    H CH3 OH O 4-(N,N-diacetyl)cytosine H Me
    H CH3 OH O Uracil H Me
    H CH3 OH O 5-Fluorouracil H Me
    H CH3 OH S 2,4-O-Diacetyluracil H Me
    H CH3 OH S Hypoxanthine H Me
    H CH3 OH S 2,4-O-Diacetylthymine H Me
    H CH3 OH S Thymine H Me
    H CH3 OH S Cytosine H Me
    H CH3 OH S 4-(N-mono-acetyl)cytosine H Me
    H CH3 OH S 4-(N,N-diacetyl)cytosine H Me
    H CH3 OH S Uracil H Me
    H CH3 OH S 5-Fluorouracil H Me
    monophosphate CH3 OH O 2,4-O-Diacetyluracil H Me
    monophosphate CH3 OH O Hypoxanthine H Me
    monophosphate CH3 OH O 2,4-O-Diacetylthymine H Me
    monophosphate CH3 OH O Thymine H Me
    monophosphate CH3 OH O Cytosine H Me
    monophosphate CH3 OH O 4-(N-mono-acetyl)cytosine H Me
    monophosphate CH3 OH O 4-(N,N-diacetyl)cytosine H Me
    monophosphate CH3 OH O Uracil H Me
    monophosphate CH3 OH O 5-Fluorouracil H Me
    monophosphate CH3 OH S 2,4-O-Diacetyluracil H Me
    monophosphate CH3 OH S Hypoxanthine H Me
    monophosphate CH3 OH S 2,4-O-Diacetylthymine H Me
    monophosphate CH3 OH S Thymine H Me
    monophosphate CH3 OH S Cytosine H Me
    monophosphate CH3 OH S 4-(N-mono-acetyl)cytosine H Me
    monophosphate CH3 OH S 4-(N,N-diacetyl)cytosine H Me
    monophosphate CH3 OH S Uracil H Me
    monophosphate CH3 OH S 5-Fluorouracil H Me
    diphosphate CH3 OH O 2,4-O-Diacetyluracil H Me
    diphosphate CH3 OH O Hypoxanthine H Me
    diphosphate CH3 OH O 2,4-O-Diacetylthymine H Me
    diphosphate CH3 OH O Thymine H Me
    diphosphate CH3 OH O Cytosine H Me
    diphosphate CH3 OH O 4-(N-mono-acetyl)cytosine H Me
    diphosphate CH3 OH O 4-(N,N-diacetyl)cytosine H Me
    diphosphate CH3 OH O Uracil H Me
    diphosphate CH3 OH O 5-Fluorouracil H Me
    diphosphate CH3 OH S 2,4-O-Diacetyluracil H Me
    diphosphate CH3 OH S Hypoxanthine H Me
    diphosphate CH3 OH S 2,4-O-Diacetylthymine H Me
    diphosphate CH3 OH S Thymine H Me
    diphosphate CH3 OH S Cytosine H Me
    triphosphate CH3 OH O 2,4-O-Diacetyluracil H Me
    triphosphate CH3 OH O Hypoxanthine H Me
    triphosphate CH3 OH O 2,4-O-Diacetylthymine H Me
    triphosphate CH3 OH O Thymine H Me
    triphosphate CH3 OH O Cytosine H Me
    triphosphate CH3 OH O 4-(N-mono-acetyl)cytosine H Me
    triphosphate CH3 OH O 4-(N,N-diacetyl)cytosine H Me
    triphosphate CH3 OH O Uracil H Me
    triphosphate CH3 OH O 5-Fluorouracil H Me
    triphosphate CH3 OH S 2,4-O-Diacetyluracil H Me
    triphosphate CH3 OH S Hypoxanthine H Me
    triphosphate CH3 OH S 2,4-O-Diacetylthymine H Me
    triphosphate CH3 OH S Thymine H Me
    triphosphate CH3 OH S Cytosine H Me
    monophosphate CF3 OH O 2,4-O-Diacetyluracil H Me
    monophosphate CF3 OH O Hypoxanthine H Me
    monophosphate CF3 OH O 2,4-O-Diacetylthymine H Me
    monophosphate CF3 OH O Thymine H Me
    monophosphate CF3 OH O Cytosine H Me
    monophosphate CF3 OH O 4-(N-mono-acetyl)cytosine H Me
    monophosphate CF3 OH O 4-(N,N-diacetyl)cytosine H Me
    monophosphate CF3 OH O Uracil H Me
    monophosphate CF3 OH O 5-Fluorouracil H Me
    monophosphate CF3 OH S 2,4-O-Diacetyluracil H Me
    monophosphate CF3 OH S Hypoxanthine H Me
    monophosphate CF3 OH S 2,4-O-Diacetylthymine H Me
    monophosphate CF3 OH S Thymine H Me
    monophosphate CF3 OH S Cytosine H Me
    monophosphate CF3 OH S 4-(N-mono-acetyl)cytosine H Me
    monophosphate CF3 OH S 4-(N,N-diacetyl)cytosine H Me
    monophosphate CF3 OH S Uracil H Me
    monophosphate CF3 OH S 5-Fluorouracil H Me
    acetyl CH3 OH O 4-(N,N-diacetyl)cytosine H Br
    acetyl CH3 OH S 4-(N,N-diacetyl)cytosine H Br
  • VII. Anti-Flavivirus or Pestivirus Activity
  • Compounds can exhibit anti-flavivirus or pestivirus activity by inhibiting flavivirus or pestivirus polymerase, by inhibiting other enzymes needed in the replication cycle, or by other pathways.
  • Examples
  • The test compounds were dissolved in DMSO at an initial concentration of 200 and then were serially diluted in culture medium.
  • Unless otherwise stated, baby hamster kidney (BHK-21) (ATCC CCL-10) and Bos Taurus (BT) (ATCC CRL 1390) cells were grown at 37° C. in a humidified CO2 (5%) atmosphere. BHK-21 cells were passaged in Eagle MEM additioned of 2 mM L-glutamine, 10% fetal bovine serum (FBS, Gibco) and Earle's BSS adjusted to contain 1.5 g/L sodium bicarbonate and 0.1 mM non-essential amino acids. BT cells were passaged in Dulbecco's modified Eagle's medium with 4 mM L-glutamine and 10% horse serum (HS, Gibco), adjusted to contain 1.5 g/L sodium bicarbonate, 4.5 g/L glucose and 1.0 mM sodium pyruvate. The vaccine strain 17D (YFV-17D) (Stamaril®, Pasteur Merieux) and Bovine Viral Diarrhea virus (BVDV) (ATCC VR-534) were used to infect BHK and BT cells, respectively, in 75 cm2 bottles. After a 3 day incubation period at 37° C., extensive cytopathic effect was observed. Cultures were freeze-thawed three times, cell debris were removed by centrifugation and the supernatant was aliquoted and stored at −70° C. YFV-17D and BVDV were titrated in BHK-21 and BT cells, respectively, that were grown to confluency in 24-well plates.
  • Example 4: Phosphorylation Assay of Nucleoside to Active Triphosphate
  • To determine the cellular metabolism of the compounds, HepG2 cells were obtained from the American Type Culture Collection (Rockville, Md.), and were grown in 225 cm2 tissue culture flasks in minimal essential medium supplemented with non-essential amino acids, 1% penicillin-streptomycin. The medium was renewed every three days, and the cells were subcultured once a week. After detachment of the adherent monolayer with a 10 minute exposure to 30 mL of trypsin-EDTA and three consecutive washes with medium, confluent HepG2 cells were seeded at a density of 2.5×106 cells per well in a 6-well plate and exposed to 10 μM of [3H] labeled active compound (500 dpm/pmol) for the specified time periods. The cells were maintained at 37° C. under a 5% CO2 atmosphere. At the selected time points, the cells were washed three times with ice-cold phosphate-buffered saline (PBS). Intracellular active compound and its respective metabolites were extracted by incubating the cell pellet overnight at −20° C. with 60% methanol followed by extraction with an additional 20 μL of cold methanol for one hour in an ice bath. The extracts were then combined, dried under gentle filtered air flow and stored at −20° C. until HPLC analysis. The preliminary results of the HPLC analysis are tabulated in Table 1.
  • TABLE 1
    [pmol/million cells]
    Time β-D-2′-CH3- β-D-2′-CH3- β-D-2′-CH3- β-D-2′-CH3-
    (h) riboA-TP riboU-TP riboC-TP riboG-TP
    2 33.1 0.40 2.24 ND
    4 67.7 1.21 3.99 ND
    8 147 1.57 9.76 2.85
    24 427 6.39 34.9 0.91
    30 456 7.18 36.2 3.22
    48 288 9.42 56.4 6.26
  • Example 5: Bioavailability Assay in Cynomolgus Monkeys
  • Within 1 week prior to the study initiation, the cynomolgus monkey was surgically implanted with a chronic venous catheter and subcutaneous venous access port (VAP) to facilitate blood collection and underwent a physical examination including hematology and serum chemistry evaluations and the body weight was recorded. Each monkey (six total), received approximately 250 uCi of 3H activity with each dose of active compound, namely β-D-2′-CH3-riboG at a dose level of 10 mg/kg at a dose concentration of 5 mg/mL, either via an intravenous bolus (3 monkeys, IV), or via oral gavage (3 monkeys, PO). Each dosing syringe was weighed before dosing to gravimetrically determine the quantity of formulation administered. Urine samples were collected via pan catch at the designated intervals (approximately 18-0 hours pre-dose, 0-4, 4-8 and 8-12 hours post-dosage) and processed. Blood samples were collected as well (pre-dose, 0.25, 0.5, 1, 2, 3, 6, 8, 12 and 24 hours post-dosage) via the chronic venous catheter and VAP or from a peripheral vessel if the chronic venous catheter procedure should not be possible. The blood and urine samples were analyzed for the maximum concentration (Cmax), time when the maximum concentration was achieved (Tmax), area under the curve (AUC), half life of the dosage concentration (T1/2), clearance (CL), steady state volume and distribution (Vss) and bioavailability (F), which are tabulated in Tables 2 and 3, and graphically illustrated in FIGS. 2 and 3, respectively.
  • TABLE 2
    Oral Bioavailability in Monkeys
    Mean Norm
    Dose AUC Norm AUC AUC F
    (mg) (ng/mL × h) (ng/mL × h/mg) (ng/mL × h/mg) (%)
    IV Monkey 1 46.44 13614 293.2
    IV Monkey 2 24.53 6581 268.3
    IV Monkey 3 20.72 6079 293.4 284.9
    PO Monkey 1 29.04 758 26.1
    PO Monkey 2 30.93 898 29.0
    PO Monkey 3 30.04 1842 61.3 38.8 13.6
  • TABLE 3
    Experimental Pharmacokinetics of β-D-2′-CH3-riboG
    in Cynomolgus Monkeys
    IV PO
    Dose/Route (mg/kg) 10 10
    Cmax (ng/mL) 6945.6 ± 1886.0 217.7 ± 132.1
    Tmax (hr) 0.25 ± 0.00 2.00 ± 1.00
    AUC (ng/mL × hr) 8758.0 ± 4212.9 1166.0 ± 589.6 
    T1/2 (hr) 7.9 ± 5.4 10.3 ± 4.1 
    CL (L/hr/kg) 1.28 ± 0.48
    Vss (L/kg) 2.09 ± 0.54
    F (%) 13.8
  • Example 6: Bone Marrow Toxicity Assay
  • Human bone marrow cells were collected from normal healthy volunteers and the mononuclear population was separated by Ficoll-Hypaque gradient centrifugation as described previously by Sommadossi J-P, Carlisle R. “Toxicity of 3′-azido-3′-deoxythymidine and 9-(1,3-dihydroxy-2-propoxymethyl)guanine for normal human hematopoietic progenitor cells in vitro” Antimicrobial Agents and Chemotherapy 1987; 31:452-454; and Sommadossi J-P, Schinazi R F, Chu C K, Xie M-Y. “Comparison of cytotoxicity of the (−)- and (+)-enantiomer of 2′,3′-dideoxy-3′-thiacytidine in normal human bone marrow progenitor cells” Biochemical Pharmacology 1992; 44:1921-1925. The culture assays for CFU-GM and BFU-E were performed using a bilayer soft agar or methylcellulose method. Drugs were diluted in tissue culture medium and filtered. After 14 to 18 days at 37° C. in a humidified atmosphere of 5% CO2 in air, colonies of greater than 50 cells were counted using an inverted microscope. The results in Table 4 are presented as the percent inhibition of colony formation in the presence of drug compared to solvent control cultures.
  • TABLE 4
    Human Bone Marrow Toxicity CFU-GM
    and BFU-E Clonogenic Assays
    IC50 in μM
    Treatment CFU-GM BFU-E
    ribavirin ~5 ~1
    β-D-2′-CH3-riboA >100 >100
    β-D-2′-CH3-riboU >100 >100
    β-D-2′-CH3-riboC >10 >10
    β-D-2′-CH3-riboG >10 >100
  • Example 7: Mitochondria Toxicity Assay
  • HepG2 cells were cultured in 12-well plates as described above and exposed to various concentrations of drugs as taught by Pan-Zhou X-R, Cui L, Zhou X-J, Sommadossi J-P, Darley-Usmer V M. “Differential effects of antiretroviral nucleoside analogs on mitochondrial function in HepG2 cells” Antimicrob Agents Chemother 2000; 44:496-503. Lactic acid levels in the culture medium after 4 day drug exposure was measured using a Boehringer lactic acid assay kit. Lactic acid levels were normalized by cell number as measured by hemocytometer count. The preliminary results from this assay are tabulated in Table 5.
  • TABLE 5
    Mitochondrial Toxicity Study (L-lactic acid assay)
    Conc. (μM) lactate (mg/106 cell) % of Control
    Control 2.18
    FIAU 10 3.73 170.4
    β-D-2′-CH3-riboC 1 2.52 115.3
    10 2.36 107.9
    50 2.26 103.4
    100 2.21 101.2
    Figure US20180235993A1-20180823-C00056
    Figure US20180235993A1-20180823-C00057
  • Example 8: Cytotoxicity Assay
  • Cells were seeded at a rate of between 5×103 and 5×104/well into 96-well plates in growth medium overnight at 37° C. in a humidified CO2 (5%) atmosphere. New growth medium containing serial dilutions of the drugs was then added. After incubation for 4 days, cultures were fixed in 50% TCA and stained with sulforhodamineB. The optical density was read at 550 nm. The cytotoxic concentration was expressed as the concentration required to reduce the cell number by 50% (CC50). The data is tabulated in Table 6.
  • TABLE 6
    MDBK versus Human Hepatoma
    CC50, μM
    Compound MDBK Huh7 HepG2
    β-D-2'-CH3-riboA 20 40 50-60
    β-D-2'-CH3-riboU >250 >250 >250
    β-D-2'-CH3-riboC 100 >250 150
    β-D-2'-CH3-riboG 100 >250 >250
    Ribavirin 5 25 150
  • Example 9: Cell Protection Assay (CPA)
  • The assay was performed essentially as described by Baginski, S. G.; Pevear, D. C.; Seipel, M.; Sun, S. C. C.; Benetatos, C. A.; Chunduru, S. K.; Rice, C. M. and M. S. Collett “Mechanism of action of a pestivirus antiviral compound” PNAS USA 2000, 97(14), 7981-7986. MDBK cells (ATCC) were seeded onto 96-well culture plates (4,000 cells per well) 24 hours before use. After infection with BVDV (strain NADL, ATCC) at a multiplicity of infection (MOI) of 0.02 plaque forming units (PFU) per cell, serial dilutions of test compounds were added to both infected and uninfected cells in a final concentration of 0.5% DMSO in growth medium. Each dilution was tested in quadruplicate. Cell densities and virus inocula were adjusted to ensure continuous cell growth throughout the experiment and to achieve more than 90% virus-induced cell destruction in the untreated controls after four days post-infection. After four days, plates were fixed with 50% TCA and stained with sulforhodamine B. The optical density of the wells was read in a microplate reader at 550 nm. The 50% effective concentration (EC50) values were defined as the compound concentration that achieved 50% reduction of cytopathic effect of the virus. The results are tabulated in Table 7. FIGS. 4 and 5 provide a graphical illustration of the methodology used to arrive at the 50% effective concentration (EC50) values for β-D-2′-CH3-riboG and ribavirin. FIG. 6 compares the results of the CPA for β-D-2′-CH3-riboG, β-D-2′-CH3-riboC, riboU, β-D-2′-CH3-riboA and ribavirin
  • TABLE 7
    Cell Protection Assay
    EC50, μM CC50, μM
    β-D-2′-CH3-riboA 2 20
    β-D-2′-CH3-riboU 20 >250
    β-D-2′-CH3-riboC 2 100
    β-D-2′-CH3-riboG 4 100
    Ribavirin >3 5
  • Example 10: Plaque Reduction Assay
  • For each compound the effective concentration was determined in duplicate 24-well plates by plaque reduction assays. Cell monolayers were infected with 100 PFU/well of virus. Then, serial dilutions of test compounds in MEM supplemented with 2% inactivated serum and 0.75% of methyl cellulose were added to the monolayers. Cultures were further incubated at 37° C. for 3 days, then fixed with 50% ethanol and 0.8% Crystal Violet, washed and air-dried. Then plaques were counted to determine the concentration to obtain 90% virus suppression and tabulated in Table 8. FIG. 7 is a graphical illustration of the results from the Plaque Reduction Assay. FIG. 8 is an image of BVDV plaque formation in the presence of increasing concentrations of β-D-2′-CH3-riboU.
  • TABLE 8
    Viral Suppression via Plaque Reduction Assay
    EC90, μM
    β-D-2′-CH3-riboA <3
    β-D-2′-CH3-riboU <81
    β-D-2′-CH3-riboC <9
    β-D-2′-CH3-riboG <9
  • Example 11: Yield Reduction Assay
  • For each compound the concentration to obtain a 6-log reduction in viral load was determined in duplicate 24-well plates by yield reduction assays. The assay was performed as described by Baginski, S. G.; Pevear, D. C.; Seipel, M.; Sun, S. C. C.; Benetatos, C. A.; Chunduru, S. K.; Rice, C. M. and M. S. Collett “Mechanism of action of a pestivirus antiviral compound” PNAS USA 2000, 97(14), 7981-7986, with minor modifications. Briefly, MDBK cells were seeded onto 24-well plates (2×105 cells per well) 24 hours before infection with BVDV (NADL strain) at a multiplicity of infection (MOI) of 0.1 PFU per cell. Serial dilutions of test compounds were added to cells in a final concentration of 0.5% DMSO in growth medium. Each dilution was tested in triplicate. After three days, cell cultures (cell monolayers and supernatants) were lysed by three freeze-thaw cycles, and virus yield was quantified by plaque assay. Briefly, MDBK cells were seeded onto 6-well plates (5×105 cells per well) 24 h before use. Cells were inoculated with 0.2 mL of test lysates for 1 hour, washed and overlaid with 0.5% agarose in growth medium. After 3 days, cell monolayers were fixed with 3.5% formaldehyde and stained with 1% crystal violet (w/v in 50% ethanol) to visualize plaques. The plaques were counted to determine the concentration to obtain a 6-log reduction in viral load as tabulated in Table 9. FIG. 9 is a graphical illustration of the results from the Yield Reduction Assay. FIG. 8 is an image of BVDV yield reduction in the presence of increasing concentrations of β-D-2′-CH3-riboC.
  • TABLE 9
    Concentration to Obtain 6-log Reduction
    Conc. for 6-log Reduction
    (μM)
    β-D-2′-CH3-riboU 120
    β-D-2′-CH3-riboG 20
    β-D-2′-CH3-riboC 20
    β-D-2′-CH3-riboA 9
  • Example 12: Comparative Cytotoxicity
  • Table 10 summarizes the cytoxicity of two compounds of this invention, β-D-1′-CH3-riboA and β-D-2′-CH3-riboA, in comparison to RBV (“ribavirin”), in various cell systems.
  • TABLE 10
    Comparative Cytotoxicity* (CC50)
    BD BHK VERO MT-4
    β-D-1′-CH3-riboA >100 200 >100 18
    β-D-2′-CH3-riboA 75 22 22 6.6
    RBV ND 50 11 ND
    *Compound concentration (μM) required to reduce the viability of cells by 50%.
  • The chemical structures for β-D-1′-CH3-riboA and β-D-2′-CH3-riboA are as follows:
  • Figure US20180235993A1-20180823-C00058
  • Table 11 summarizes the antiviral activity of β-D-1′-CH3-riboA and β-D-2′-CH3-riboA against several viruses within the flavivirus and pestivirus genuses.
  • TABLE 11
    Comparative Antiviral Activity* (EC50)
    BVDV YFV PICO VSV HIV-1
    β-D-1′-CH3-riboA 10 7.0 51 >100 >18
    β-D-2′-CH3-riboA 0.1 0.2 5.0 >100 >6.6
    RBV ND 30 >30 ND ND
    *Compound concentration (μM) required to reduce the plaque number by 50%. The following virus-cell system were used: BVDC-BT, YFV-BHK, PICO (Cosxackie B1 and Polio Sabin)/VSV-Vero.
  • Table 12 summarizes the antiviral activity and toxicity of β-D-2′-methyl-riboG, β-D-2′-methyl-riboC and β-D-2′-methyl-riboU, against a couple of viruses within the flavivirus and pestivirus genuses.
  • TABLE 12
    Comparative Antiviral Activity* (EC50)
    BVDV YFV
    EC50* CC50** EC50* CC50**
    β-D-2′-CH3-riboG 2 >100 1.2 20
    β-D-2′-CH3-riboC 3.7 >100 70 >100
    β-D-2′-CH3-riboU 20 >100 33 >100
    *Compound concentration (μM) required to reduce the plaque number by 50%. The following virus-cell system were used: BVDC-BT and YFV-BHK.
    *Compound concentration (μM) required to reduce the viability of cells by 50%.
  • The chemical structures for β-D-2′-CH3-riboG, β-D-2′-CH3-riboC and β-D-2′-CH3-riboU are as follows:
  • Figure US20180235993A1-20180823-C00059
  • Table 13 summarizes the anti-viral activity of several compounds of this invention against BVDV in three different assays.
  • TABLE 13
    for BVDV
    Yield Reduction
    Cell Plaque 6 log10 Cytotoxicity
    Protection Reduction reduction Huh7 cells
    Compound (EC50, μM) (EC90, μM) EC90, μM (μM) (EC50, μM)
    β-D-2′-CH3-riboA 2 <3 <2 9 50
    β-D-2′-CH3-riboT >250 ND ND ND >250
    β-D-2′-CH3-riboU 20 <81 24 120 >250
    β-D-2′-CH3-riboC 2 <9 <4 20 >250
    β-D-2′-CH3-riboG 4 <9 3 20 >250
    β-D-2′-CH3-riboI 45 ND ND ND >250
    Ribavirin >3 >200 >20 toxic 20
  • This invention has been described with reference to its preferred embodiments. Variations and modifications of the invention, will be obvious to those skilled in the art from the foregoing detailed description of the invention.

Claims (28)

1-129. (canceled)
130. A method for the treatment of a flavivirus or pestivirus infection in a host, comprising administering to a host infected with a flavivirus or a pestivirus an anti-virally effective amount of a compound of Formula X:
Figure US20180235993A1-20180823-C00060
or a phosphate thereof, or a pharmaceutically acceptable salt or ester thereof, wherein:
Base is a purine or pyrimidine base;
R1, R2, and R3 are independently H; phosphate; mono phosphate; diphosphate;
triphosphate; a stabilized phosphate prodrug; acyl; lower acyl; alkyl; lower alkyl; sulfonate ester; alkyl sulfonyl; arylalkyl sulfonyl; methanesulfonyl; benzyl, wherein the phenyl group is optionally substituted with one or more substituents selected from the group consisting of hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate. phosphonic acid, phosphate or phosphonate; a lipid; a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo provides a compound wherein R1, R2, or R3 is independently H or phosphate;
R6 is alkyl; and
X is O, S, SO2 or CH2.
131. The method of claim 130, wherein R1 and R2 are independently H;
phosphate; monophosphate; diphosphate; triphosphate; or a stabilized phosphate prodrug.
132. The method of claim 130, wherein R1 and R2 are each hydrogen.
133. The method of claim 130, wherein X is O.
134. The method of claim 130, wherein R6 is methyl.
135. The method of claim 130, wherein Base is a purine or pyrimidine base selected from adenine, N6-alkyl purine, N6-acyl purine, N6-benzylpurine, N6-halopurine, N6-vinylpurine, N6-acetylenic purine, N6-acyl purine, N6-hydroxyalkyl purine, N6-thioalkyl purine, N2-alkyl purine, N2-alkyl-6-thiopurine, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6-azapyrimidine, 6-azacytosine, 2- and/or 4-mercaptopyrimidine, uracil, 5-halouracil, 5-fluorouracil, C5-alkylpyrimidine, C5-benzyl pyrimidine, C5-halopyrimidine, C5-vinyl pyrimidine, C5-acetylenic pyrimidine, C5-acyl pyrimidine, C5-hydroxyalkyl purine, C5-amidopyrimidine, C5-cyanopyrimidine, C5-nitropyrimidine, C5-aminopyrimidine, N2-alkyl purine, N2-alkyl-6-thiopurine, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, guanine, hypoxanthine, 2,6-diaminopurine, or 6-chloropurine.
136. The method of claim 130, wherein Base is a purine or pyrimidine base selected from adenine, thymine, cytosine, uracil or guanine.
137. The method of claim 130, wherein Base is cytosine or uracil.
138. The method of claim 130, wherein the flavivirus or pestivirus is selected from dengue hemorrhagic fever virus, yellow fever virus, shock syndrome or Japanese encephalitis virus.
139. A method for the treatment of a flavivirus or pestivirus infection in a host, comprising contacting a cell infected with a flavivirus or a pestivirus with an anti-virally effective amount of a compound of Formula X:
Figure US20180235993A1-20180823-C00061
or a phosphate thereof, or a pharmaceutically acceptable salt or ester thereof, wherein:
Base is a purine or pyrimidine base;
R1, R2, and R3 are independently H; phosphate; mono phosphate; diphosphate;
triphosphate; a stabilized phosphate prodrug; acyl; lower acyl; alkyl; lower alkyl; sulfonate ester; alkyl sulfonyl; arylalkyl sulfonyl; methanesulfonyl; benzyl, wherein the phenyl group is optionally substituted with one or more substituents selected from the group consisting of hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate. phosphonic acid, phosphate or phosphonate; a lipid; a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo provides a compound wherein R1, R2, or R3 is independently H or phosphate;
R6 is alkyl; and
X is O, S, SO2 or CH2.
140. The method of claim 139, wherein R1 and R2 are independently H; phosphate; monophosphate; diphosphate; triphosphate; or a stabilized phosphate prodrug.
141. The method of claim 139, wherein R1 and R2 are each hydrogen.
142. The method of claim 139, wherein X is O.
143. The method of claim 139, wherein R6 is methyl.
144. The method of claim 139, wherein Base is a purine or pyrimidine base selected from adenine, N6-alkyl purine, N6-acyl purine, N6-benzyl purine, N6-halopurine, N6-vinyl purine, N6-acetylenic purine, N6-acyl purine, N6-hydroxyalkyl purine, N6-thioalkyl purine, N2-alkyl purine, N2-alkyl-6-thiopurine, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6-azapyrimidine, 6-azacytosine, 2- and/or 4-mercaptopyrimidine, uracil, 5-halouracil, 5-fluorouracil, C5-alkylpyrimidine, C5-benzyl pyrimidine, C5-halopyrimidine, C5-vinyl pyrimidine, C5-acetylenic pyrimidine, C5-acyl pyrimidine, C5-hydroxyalkyl purine, C5-amidopyrimidine, C5-cyanopyrimidine, C5-nitropyrimidine, C5-aminopyrimidine, N2-alkylpurine, N2-alkyl-6-thiopurine, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, guanine, hypoxanthine, 2,6-diaminopurine, or 6-chloropurine.
145. The method of claim 139, wherein Base is a purine or pyrimidine base selected from adenine, thymine, cytosine, uracil or guanine.
146. The method of claim 139, wherein Base is cytosine or uracil.
147. The method of claim 139, wherein the flavivirus or pestivirus is selected from dengue hemorrhagic fever virus, yellow fever virus, shock syndrome or Japanese encephalitis virus.
148. A method for the treatment of a flavivirus or pestivirus infection in a host, comprising contacting a flavivirus or pestivirus polymerase with an anti-virally effective amount of a compound of Formula X:
Figure US20180235993A1-20180823-C00062
or a phosphate thereof, or a pharmaceutically acceptable salt or ester thereof, wherein:
Base is a purine or pyrimidine base;
R1, R2, and R3 are independently H; phosphate; mono phosphate; diphosphate;
triphosphate; a stabilized phosphate prodrug; acyl; lower acyl; alkyl; lower alkyl; sulfonate ester; alkyl sulfonyl; arylalkyl sulfonyl; methanesulfonyl; benzyl, wherein the phenyl group is optionally substituted with one or more substituents selected from the group consisting of hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nitro, cyano, sulfonic acid, sulfate. phosphonic acid, phosphate or phosphonate; a lipid; a phospholipid; an amino acid; a carbohydrate; a peptide; a cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo provides a compound wherein R1, R2, or R3 is independently H or phosphate;
R6 is alkyl; and
X is O, S, SO2 or CH2.
149. The method of claim 148, wherein R1 and R2 are independently H; phosphate; monophosphate; diphosphate; triphosphate; or a stabilized phosphate prodrug.
150. The method of claim 148, wherein R1 is triphosphate and R2 is hydrogen.
151. The method of claim 148, wherein X is O.
152. The method of claim 148, wherein R6 is methyl.
153. The method of claim 148, wherein Base is a purine or pyrimidine base selected from adenine, N6-alkyl purine, N6-acyl purine, N6-benzyl purine, N6-halopurine, N6-vinyl purine, N6-acetylenic purine, N6-acyl purine, N6-hydroxyalkyl purine, N6-thioalkyl purine, N2-alkyl purine, N2-alkyl-6-thiopurine, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6-azapyrimidine, 6-azacytosine, 2- and/or 4-mercaptopyrimidine, uracil 5-halouracil, 5-fluorouracil C5-alkyl pyrimidine, C5-benzylpyrimidine, C5-halopyrimidine, C5-vinyl pyrimidine, C5-acetylenic pyrimidine, C5-acyl pyrimidine, C5-hydroxyalkyl purine, C5-amidopyrimidine, C5-cyanopyrimidine, C5-nitropyrimidine, C5-aminopyrimidine, N2-alkylpurine, N2-alkyl-6-thiopurine, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, guanine, hypoxanthine, 2,6-diaminopurine, or 6-chloropurine.
154. The method of claim 148, wherein Base is a purine or pyrimidine base selected from adenine, thymine, cytosine, uracil or guanine.
155. The method of claim 148, wherein Base is cytosine or uracil.
156. The method of claim 148, wherein the flavivirus or pestivirus is selected from dengue hemorrhagic fever virus, yellow fever virus, shock syndrome or Japanese encephalitis virus.
US15/952,117 2000-05-26 2018-04-12 Methods and compositions for treating flaviviruses and pestiviruses Abandoned US20180235993A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/952,117 US20180235993A1 (en) 2000-05-26 2018-04-12 Methods and compositions for treating flaviviruses and pestiviruses

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US20767400P 2000-05-26 2000-05-26
US28327601P 2001-04-11 2001-04-11
US09/863,816 US6812219B2 (en) 2000-05-26 2001-05-23 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,135 US7163929B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US11/527,124 US8343937B2 (en) 2000-05-26 2006-09-25 Methods and compositions for treating flaviviruses and pestiviruses
US13/675,937 US9968628B2 (en) 2000-05-26 2012-11-13 Methods and compositions for treating flaviviruses and pestiviruses
US15/952,117 US20180235993A1 (en) 2000-05-26 2018-04-12 Methods and compositions for treating flaviviruses and pestiviruses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/675,937 Division US9968628B2 (en) 2000-05-26 2012-11-13 Methods and compositions for treating flaviviruses and pestiviruses

Publications (1)

Publication Number Publication Date
US20180235993A1 true US20180235993A1 (en) 2018-08-23

Family

ID=26902464

Family Applications (9)

Application Number Title Priority Date Filing Date
US09/863,816 Expired - Lifetime US6812219B2 (en) 2000-05-26 2001-05-23 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,135 Expired - Lifetime US7163929B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,693 Expired - Lifetime US7148206B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,694 Expired - Lifetime US7105493B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,692 Expired - Lifetime US7101861B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US11/527,124 Active 2025-06-11 US8343937B2 (en) 2000-05-26 2006-09-25 Methods and compositions for treating flaviviruses and pestiviruses
US13/675,937 Expired - Fee Related US9968628B2 (en) 2000-05-26 2012-11-13 Methods and compositions for treating flaviviruses and pestiviruses
US13/956,287 Abandoned US20130315863A1 (en) 2000-05-26 2013-07-31 Methods and compositions for treating flaviviridae infections
US15/952,117 Abandoned US20180235993A1 (en) 2000-05-26 2018-04-12 Methods and compositions for treating flaviviruses and pestiviruses

Family Applications Before (8)

Application Number Title Priority Date Filing Date
US09/863,816 Expired - Lifetime US6812219B2 (en) 2000-05-26 2001-05-23 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,135 Expired - Lifetime US7163929B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,693 Expired - Lifetime US7148206B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,694 Expired - Lifetime US7105493B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US10/602,692 Expired - Lifetime US7101861B2 (en) 2000-05-26 2003-06-20 Methods and compositions for treating flaviviruses and pestiviruses
US11/527,124 Active 2025-06-11 US8343937B2 (en) 2000-05-26 2006-09-25 Methods and compositions for treating flaviviruses and pestiviruses
US13/675,937 Expired - Fee Related US9968628B2 (en) 2000-05-26 2012-11-13 Methods and compositions for treating flaviviruses and pestiviruses
US13/956,287 Abandoned US20130315863A1 (en) 2000-05-26 2013-07-31 Methods and compositions for treating flaviviridae infections

Country Status (24)

Country Link
US (9) US6812219B2 (en)
EP (2) EP1736478B1 (en)
JP (2) JP5230052B2 (en)
KR (2) KR20080021797A (en)
CN (2) CN1315862C (en)
AP (2) AP1727A (en)
AR (1) AR032883A1 (en)
AU (2) AU2001272923A1 (en)
BR (1) BR0111196A (en)
CA (1) CA2410579C (en)
CZ (1) CZ301182B6 (en)
EA (2) EA007867B1 (en)
IL (1) IL153020A0 (en)
MA (1) MA26916A1 (en)
MX (1) MXPA02011691A (en)
NO (1) NO327249B1 (en)
NZ (2) NZ536570A (en)
OA (1) OA12382A (en)
PE (1) PE20020051A1 (en)
PL (1) PL359169A1 (en)
TW (1) TW200425898A (en)
WO (1) WO2001092282A2 (en)
YU (1) YU92202A (en)
ZA (2) ZA200210112B (en)

Families Citing this family (230)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6444652B1 (en) 1998-08-10 2002-09-03 Novirio Pharmaceuticals Limited β-L-2'-deoxy-nucleosides for the treatment of hepatitis B
DE122007000062I1 (en) * 1998-08-10 2007-12-20 Idenix Cayman Ltd Beta-L-2'-deoxynucleosides for the treatment of hepatitis B virus
AU2001245575A1 (en) * 2000-03-09 2001-09-17 Videoshare, Inc. Sharing a streaming video
MY164523A (en) 2000-05-23 2017-12-29 Univ Degli Studi Cagliari Methods and compositions for treating hepatitis c virus
EP1736478B1 (en) * 2000-05-26 2015-07-22 IDENIX Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
CN101862345B (en) * 2000-10-18 2014-06-04 吉利德制药有限责任公司 Modified nucleosides for treatment of viral infections and abnormal cellular proliferation
US7105499B2 (en) * 2001-01-22 2006-09-12 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
WO2002057287A2 (en) * 2001-01-22 2002-07-25 Merck & Co., Inc. Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
US8481712B2 (en) 2001-01-22 2013-07-09 Merck Sharp & Dohme Corp. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
GB0114286D0 (en) * 2001-06-12 2001-08-01 Hoffmann La Roche Nucleoside Derivatives
WO2003000200A2 (en) 2001-06-22 2003-01-03 Pharmasset Ltd. β-2'-OR 3'-HALONUCLEOSIDES
US20040006002A1 (en) * 2001-09-28 2004-01-08 Jean-Pierre Sommadossi Methods and compositions for treating flaviviruses and pestiviruses using 4'-modified nucleoside
WO2003051898A1 (en) * 2001-12-17 2003-06-26 Ribapharm Inc. Unusual nucleoside libraries, compounds, and preferred uses as antiviral and anticancer agents
AU2002341942A1 (en) * 2002-01-17 2003-09-02 Ribapharm Inc. Sugar modified nucleosides as viral replication inhibitors
WO2003062256A1 (en) * 2002-01-17 2003-07-31 Ribapharm Inc. 2'-beta-modified-6-substituted adenosine analogs and their use as antiviral agents
US7217815B2 (en) * 2002-01-17 2007-05-15 Valeant Pharmaceuticals North America 2-beta -modified-6-substituted adenosine analogs and their use as antiviral agents
WO2003068244A1 (en) * 2002-02-13 2003-08-21 Merck & Co., Inc. Methods of inhibiting orthopoxvirus replication with nucleoside compounds
US7247621B2 (en) * 2002-04-30 2007-07-24 Valeant Research & Development Antiviral phosphonate compounds and methods therefor
EP1501850A2 (en) * 2002-05-06 2005-02-02 Genelabs Technologies, Inc. Nucleoside derivatives for treating hepatitis c virus infection
EP1572945A2 (en) * 2002-06-27 2005-09-14 Merck & Co., Inc. Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
PT1523489E (en) * 2002-06-28 2014-06-24 Centre Nat Rech Scient Modified 2' and 3' -nucleoside produgs for treating flaviridae infections
CN101172993A (en) * 2002-06-28 2008-05-07 埃迪尼克斯(开曼)有限公司 2'-c-methyl-3'-o-l-valine ester ribofuranosyl cytidine for treatment of flaviviridae infections
US7608600B2 (en) * 2002-06-28 2009-10-27 Idenix Pharmaceuticals, Inc. Modified 2′ and 3′-nucleoside prodrugs for treating Flaviviridae infections
AP2005003213A0 (en) * 2002-06-28 2005-03-31 Univ Cagliari 2'-C-methyl-3'-O-L-valine ester ribofuranosyl cytidine for treatment of flaviviridae infections.
AU2003263412A1 (en) 2002-06-28 2004-01-19 Centre National De La Recherche Scientifique (Cnrs) 1'-, 2'- and 3'- modified nucleoside derivatives for treating flaviviridae infections
US20060264389A1 (en) * 2002-07-16 2006-11-23 Balkrishen Bhat Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
WO2004009020A2 (en) 2002-07-24 2004-01-29 Merck & Co., Inc. Pyrrolopyrimidine thionucleoside analogs as antivirals
IL166640A0 (en) * 2002-08-01 2006-01-15 Pharmasset Ltd Compounds with the bicyclo Ä4.2.1Ü nonane system for the treatment of flaviviridae infections
US20040067877A1 (en) * 2002-08-01 2004-04-08 Schinazi Raymond F. 2', 3'-Dideoxynucleoside analogues for the treatment or prevention of Flaviviridae infections
US7151092B2 (en) 2002-10-31 2006-12-19 Metabasis Therapeutics, Inc. Cytarabine monophosphate prodrugs
HUE033832T2 (en) 2002-11-15 2018-01-29 Idenix Pharmaceuticals Llc 2'-methyl nucleosides in combination with interferon and flaviviridae mutation
US7034167B2 (en) 2002-12-06 2006-04-25 Merck & Co., Inc. Process to ribofuranose sugar derivatives as intermediates to branched-chain nucleosides
CA2509687C (en) * 2002-12-12 2012-08-14 Idenix (Cayman) Limited Process for the production of 2'-branched nucleosides
PL377608A1 (en) * 2002-12-23 2006-02-06 Idenix (Cayman) Limited Process for the production of 3'-nucleoside prodrugs
AR043006A1 (en) 2003-02-12 2005-07-13 Merck & Co Inc PROCESS TO PREPARE RAMIFIED RIBONUCLEOSIDS
MXPA05010419A (en) * 2003-03-28 2006-05-31 Pharmasset Inc Compounds for the treatment of flaviviridae infections.
US20040259934A1 (en) * 2003-05-01 2004-12-23 Olsen David B. Inhibiting Coronaviridae viral replication and treating Coronaviridae viral infection with nucleoside compounds
CN100503628C (en) 2003-05-30 2009-06-24 法莫赛特股份有限公司 Modified fluorinated nucleoside analogues
AU2004258750A1 (en) 2003-07-25 2005-02-03 Centre National De La Recherche Scientifique -Cnrs Purine nucleoside analogues for treating diseases caused by flaviviridae including hepatitis C
WO2005018330A1 (en) * 2003-08-18 2005-03-03 Pharmasset, Inc. Dosing regimen for flaviviridae therapy
BRPI0414019A (en) 2003-08-27 2006-10-24 Biota Inc tricyclic nucleosides or nucleotides as therapeutic agents
GB0500020D0 (en) 2005-01-04 2005-02-09 Novartis Ag Organic compounds
US20050182252A1 (en) * 2004-02-13 2005-08-18 Reddy K. R. Novel 2'-C-methyl nucleoside derivatives
EP1758453B1 (en) 2004-06-15 2014-07-16 Merck Sharp & Dohme Corp. C-purine nucleoside analogs as inhibitors of rna-dependent rna viral polymerase
RU2007102281A (en) * 2004-06-23 2008-07-27 Айденикс (Кайман) Лимитед (Ky) 5-AZA-7-DEAZAPURINE DERIVATIVES FOR THE TREATMENT OF DISEASES ASSOCIATED WITH FLAVIVIRIDAE
CN1972696B (en) * 2004-06-24 2010-08-11 默沙东公司 Nucleoside aryl phosphoramidates for the treatment of RNA-dependent RNA viral infection
CN101023094B (en) * 2004-07-21 2011-05-18 法莫赛特股份有限公司 Preparation of alkyl-substituted 2-deoxy-2-fluoro-d-ribofuranosyl pyrimidines and purines and their derivatives
PL3109244T3 (en) * 2004-09-14 2019-09-30 Gilead Pharmasset Llc Preparation of 2'fluoro-2'-alkyl-substituted or other optionally substituted ribofuranosyl pyrimidines and purines and their derivatives
US7524831B2 (en) 2005-03-02 2009-04-28 Schering Corporation Treatments for Flaviviridae virus infection
EP1858889A1 (en) 2005-03-08 2007-11-28 Biota Scientific Management Pty. Ltd. Bicyclic nucleosides and nucleotides as therapeutic agents
US20090156545A1 (en) * 2005-04-01 2009-06-18 Hostetler Karl Y Substituted Phosphate Esters of Nucleoside Phosphonates
CA2606195C (en) 2005-05-02 2015-03-31 Merck And Co., Inc. Hcv ns3 protease inhibitors
ATE524183T1 (en) 2005-06-17 2011-09-15 Novartis Ag USE OF SANGLIFEHRIN IN HCV THERAPY
TWI387603B (en) 2005-07-20 2013-03-01 Merck Sharp & Dohme Hcv ns3 protease inhibitors
BRPI0614205A2 (en) 2005-08-01 2016-11-22 Merck & Co Inc compound, pharmaceutical composition and compound use
AR056191A1 (en) * 2005-08-23 2007-09-26 Idenix Phatmaceuticals Inc RING OF NUCLEOSIDS OF SEVEN MEMBERS AS INHIBITORS OF THE VIRAL REPLICA, METHODS FOR THEIR SYNTHESIS, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND THEIR USE IN THE TREATMENT OF VIRUS INFECTION OF THE FLAVIVIRIDAE FAMILY
US20090047338A1 (en) * 2005-10-05 2009-02-19 Immune Disease Institute, Inc. Method to Treat Flavivirus Infection with siRNA
WO2007065829A1 (en) 2005-12-09 2007-06-14 F. Hoffmann-La Roche Ag Antiviral nucleosides
CA2634749C (en) 2005-12-23 2014-08-19 Idenix Pharmaceuticals, Inc. Process for preparing a synthetic intermediate for preparation of branched nucleosides
US8895531B2 (en) 2006-03-23 2014-11-25 Rfs Pharma Llc 2′-fluoronucleoside phosphonates as antiviral agents
EP2004634A1 (en) 2006-04-04 2008-12-24 F.Hoffmann-La Roche Ag 3',5'-di-o-acylated nucleosides for hcv treatment
EP2007789B1 (en) 2006-04-11 2015-05-20 Novartis AG Spirocyclic HCV/HIV inhibitors and their uses
GB0609492D0 (en) 2006-05-15 2006-06-21 Angeletti P Ist Richerche Bio Therapeutic agents
US8058260B2 (en) * 2006-05-22 2011-11-15 Xenoport, Inc. 2′-C-methyl-ribofuranosyl cytidine prodrugs, pharmaceutical compositions and uses thereof
GB0612423D0 (en) 2006-06-23 2006-08-02 Angeletti P Ist Richerche Bio Therapeutic agents
PT2084174E (en) 2006-10-10 2013-10-08 Hoffmann La Roche Preparation of nucleosides ribofuranosyl pyrimidines
AP2009004812A0 (en) 2006-10-10 2009-04-30 Medivir Ab HCV nucleoside inhibitor
JP5345541B2 (en) 2006-10-24 2013-11-20 メルク・シャープ・アンド・ドーム・コーポレーション HCV NS3 protease inhibitor
EP2076278B1 (en) 2006-10-24 2015-05-06 Merck Sharp & Dohme Corp. Macrocyclic HCV NS3 protease inhibitors
CN101583372A (en) 2006-10-24 2009-11-18 默克公司 HCV NS3 protease inhibitors
EP2083844B1 (en) 2006-10-27 2013-11-27 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors
AU2007318165B2 (en) 2006-10-27 2011-11-17 Msd Italia S.R.L. HCV NS3 protease inhibitors
ES2367455T3 (en) 2006-11-09 2011-11-03 F. Hoffmann-La Roche Ag ARILAMIDES REPLACED BY TIAZOL OR OXAZOL.
GB0623493D0 (en) 2006-11-24 2007-01-03 Univ Cardiff Chemical compounds
US7897737B2 (en) 2006-12-05 2011-03-01 Lasergen, Inc. 3′-OH unblocked, nucleotides and nucleosides, base modified with photocleavable, terminating groups and methods for their use in DNA sequencing
GB0625283D0 (en) 2006-12-19 2007-01-24 Cyclacel Ltd Combination
US8101595B2 (en) 2006-12-20 2012-01-24 Istituto di Ricerche di Biologia Molecolare P. Angletti SpA Antiviral indoles
GB0625345D0 (en) 2006-12-20 2007-01-31 Angeletti P Ist Richerche Bio Therapeutic compounds
GB0625349D0 (en) 2006-12-20 2007-01-31 Angeletti P Ist Richerche Bio Therapeutic compounds
US7951789B2 (en) 2006-12-28 2011-05-31 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
AU2007342367B2 (en) 2007-01-05 2012-12-06 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Nucleoside aryl phosphoramidates for the treatment of RNA-dependent RNA viral infection
CA2674589A1 (en) * 2007-01-12 2008-07-24 Biocryst Pharmaceuticals, Inc. Antiviral nucleoside analogs
PL2144604T3 (en) 2007-02-28 2012-02-29 Conatus Pharmaceuticals Inc Methods for the treatment of chronic viral hepatitis C using RO 113-0830
ES2426684T3 (en) 2007-03-23 2013-10-24 To-Bbb Holding B.V. Conjugates for the targeted delivery of drugs through the blood brain barrier
US7964580B2 (en) 2007-03-30 2011-06-21 Pharmasset, Inc. Nucleoside phosphoramidate prodrugs
AU2008277442A1 (en) 2007-07-17 2009-01-22 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Macrocyclic indole derivatives for the treatment of hepatitis C infections
JP5433573B2 (en) 2007-07-19 2014-03-05 イステイチユート・デイ・リチエルケ・デイ・ビオロジア・モレコラーレ・ピ・アンジエレツテイ・エツセ・エルレ・エルレ Macrocyclic compounds as antiviral agents
CN101903355B (en) 2007-12-17 2014-05-14 霍夫曼-拉罗奇有限公司 Imidazole-substituted arylamides
ES2417182T3 (en) 2007-12-17 2013-08-06 F. Hoffmann-La Roche Ag New arylamides substituted with pyrazole
CA2708228C (en) 2007-12-17 2016-06-21 F. Hoffmann-La Roche Ag Tetrazole-substituted arylamide derivatives and their use as p2x3 and/or p2x2/3 purinergic receptor antagonists
KR101405746B1 (en) 2007-12-17 2014-06-10 에프. 호프만-라 로슈 아게 Triazole-substituted arylamide derivatives and their use as p2x3 and /or p2x2/3 purinergic receptor antagonists
WO2009115927A2 (en) * 2008-03-18 2009-09-24 Institut De Recherches Cliniques De Montreal Nucleotide analogues with quaternary carbon stereogenic centers and methods of use
TW200946541A (en) 2008-03-27 2009-11-16 Idenix Pharmaceuticals Inc Solid forms of an anti-HIV phosphoindole compound
CN104262345B (en) 2008-04-23 2017-06-23 吉利德科学公司 For the CARBA nucleoside analogs of 1 ' substitution of antiviral therapy
EP2271345B1 (en) 2008-04-28 2015-05-20 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors
NZ589719A (en) 2008-06-09 2012-08-31 Cyclacel Ltd Combinations of sapacitabine or cndac with dna methyltransferase inhibitors such as decitabine and procaine
WO2009152353A2 (en) 2008-06-11 2009-12-17 Lasergen, Inc. Nucleotides and nucleosides and methods for their use in dna sequencing
US8173621B2 (en) * 2008-06-11 2012-05-08 Gilead Pharmasset Llc Nucleoside cyclicphosphates
EP2307434B1 (en) * 2008-07-02 2014-02-12 IDENIX Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
EP2313102A2 (en) 2008-07-03 2011-04-27 Biota Scientific Management Bycyclic nucleosides and nucleotides as therapeutic agents
CA2731177C (en) 2008-07-22 2013-10-29 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Macrocyclic quinoxaline compounds as hcv ns3 protease inhibitors
NZ603854A (en) 2008-08-29 2015-03-27 Boehringer Ingelheim Vetmed West nile virus vaccine
NZ617066A (en) 2008-12-23 2015-02-27 Gilead Pharmasset Llc Nucleoside analogs
EA019295B1 (en) * 2008-12-23 2014-02-28 Джилид Фармассет, Ллс. Synthesis of purine nucleosides and process for preparing them
AU2009329867B2 (en) 2008-12-23 2015-01-29 Gilead Pharmasset Llc Nucleoside phosphoramidates
AU2010203416C1 (en) 2009-01-09 2013-07-25 Inhibitex, Inc. Phosphoramidate derivatives of guanosine nucleoside compounds for treatment of viral infections
WO2010082050A1 (en) 2009-01-16 2010-07-22 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Macrocyclic and 7-aminoalkyl-substituted benzoxazocines for treatment of hepatitis c infections
GB0900914D0 (en) 2009-01-20 2009-03-04 Angeletti P Ist Richerche Bio Antiviral agents
US8193372B2 (en) 2009-03-04 2012-06-05 Idenix Pharmaceuticals, Inc. Phosphothiophene and phosphothiazole HCV polymerase inhibitors
EA201190178A1 (en) * 2009-03-20 2012-06-29 Алиос Биофарма, Инк. REPLACED NUCLEOSIDE AND NUCLEOTIC ANALOGUES
US20110182850A1 (en) 2009-04-10 2011-07-28 Trixi Brandl Organic compounds and their uses
US8512690B2 (en) 2009-04-10 2013-08-20 Novartis Ag Derivatised proline containing peptide compounds as protease inhibitors
US20100297079A1 (en) * 2009-05-20 2010-11-25 Chimerix, Inc. Compounds, compositions and methods for treating viral infection
TWI576352B (en) 2009-05-20 2017-04-01 基利法瑪席特有限責任公司 Nucleoside phosphoramidates
US8618076B2 (en) 2009-05-20 2013-12-31 Gilead Pharmasset Llc Nucleoside phosphoramidates
US9676797B2 (en) 2015-09-02 2017-06-13 Abbvie Inc. Anti-viral compounds
US8476457B2 (en) 2009-06-22 2013-07-02 Roche Palo Alto Llc Indole, indazole and benzimidazole arylamides as P2X3 and P2X2/3 antagonists
WO2010149578A2 (en) 2009-06-22 2010-12-29 F. Hoffmann-La Roche Ag Novel biphenyl and phenyl-pyridine amides
ES2593405T3 (en) 2009-06-22 2016-12-09 F. Hoffmann-La Roche Ag New arylamides substituted by benzoxazolone
EP2459582B1 (en) 2009-07-30 2015-05-27 Merck Sharp & Dohme Corp. Hepatitis c virus ns3 protease inhibitors
AR077712A1 (en) 2009-08-05 2011-09-14 Idenix Pharmaceuticals Inc SERINA PROTEASA MACROCICLICA INHIBITORS
SI2480559T1 (en) 2009-09-21 2013-10-30 Gilead Sciences, Inc. Processes and intermediates for the preparation of 1'-cyano-carbanucleoside analogs
AU2010317996A1 (en) 2009-11-14 2012-05-10 F. Hoffmann-La Roche Ag Biomarkers for predicting rapid response to HCV treatment
US20110117055A1 (en) 2009-11-19 2011-05-19 Macdonald James E Methods of Treating Hepatitis C Virus with Oxoacetamide Compounds
KR20120085877A (en) 2009-12-02 2012-08-01 에프. 호프만-라 로슈 아게 Biomarkers for predicting sustained response to hcv treatment
CN102822175A (en) 2009-12-18 2012-12-12 埃迪尼克斯医药公司 5,5-fused arylene or heteroarylene hepatitis C virus inhibitors
AU2011235044A1 (en) 2010-03-31 2012-11-22 Gilead Pharmasset Llc Stereoselective synthesis of phosphorus containing actives
US8563530B2 (en) 2010-03-31 2013-10-22 Gilead Pharmassel LLC Purine nucleoside phosphoramidate
CA2795054A1 (en) 2010-04-01 2011-10-06 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
KR102108864B1 (en) 2010-07-19 2020-05-12 길리애드 사이언시즈, 인코포레이티드 Methods for the preparation of diasteromerically pure phosphoramidate prodrugs
ES2524356T3 (en) 2010-07-22 2014-12-05 Gilead Sciences, Inc. Methods and compounds to treat infections caused by Paramyxoviridae virus
US8871737B2 (en) 2010-09-22 2014-10-28 Alios Biopharma, Inc. Substituted nucleotide analogs
JP5909495B2 (en) 2010-10-08 2016-04-26 ノバルティス アーゲー Vitamin E formulation of sulfamide NS3 inhibitor
WO2012075140A1 (en) 2010-11-30 2012-06-07 Pharmasset, Inc. Compounds
WO2012080050A1 (en) 2010-12-14 2012-06-21 F. Hoffmann-La Roche Ag Solid forms of a phenoxybenzenesulfonyl compound
US9351989B2 (en) * 2010-12-29 2016-05-31 Inhibitex, Inc. Substituted purine nucleosides, phosphoroamidate and phosphorodiamidate derivatives for treatment of viral infections
TW201309690A (en) 2011-02-10 2013-03-01 Idenix Pharmaceuticals Inc Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating HCV infections
US9187730B2 (en) 2011-03-14 2015-11-17 Boehringer Ingelheim Vetmedica, Inc. Equine rhinitis vaccine
WO2012154321A1 (en) 2011-03-31 2012-11-15 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US20120252721A1 (en) 2011-03-31 2012-10-04 Idenix Pharmaceuticals, Inc. Methods for treating drug-resistant hepatitis c virus infection with a 5,5-fused arylene or heteroarylene hepatitis c virus inhibitor
US9156872B2 (en) 2011-04-13 2015-10-13 Merck Sharp & Dohme Corp. 2′-azido substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
EP2696679B1 (en) 2011-04-13 2017-08-02 Merck Sharp & Dohme Corp. 2'-cyano substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
KR20130138840A (en) 2011-04-13 2013-12-19 머크 샤프 앤드 돔 코포레이션 2'-substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
WO2012140436A1 (en) 2011-04-14 2012-10-18 Cyclacel Limited Dosage regimen for sapacitabine and decitabine in combination for treating acute myeloid leukemia
CN102775458B (en) * 2011-05-09 2015-11-25 中国人民解放军军事医学科学院毒物药物研究所 The preparation of β-D-(2 ' R)-2 '-deoxidation-2 '-fluoro-2'CmeC derivative and purposes
EP2731433A4 (en) 2011-07-13 2014-12-31 Merck Sharp & Dohme 5'-substituted nucleoside analogs and methods of use thereof for the treatment of viral diseases
US9416154B2 (en) 2011-07-13 2016-08-16 Merck Sharp & Dohme Corp. 5′-substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
JP2014526474A (en) 2011-09-12 2014-10-06 アイディニックス ファーマシューティカルズ インコーポレイテッド Compounds and pharmaceutical compositions for the treatment of viral infections
AR088441A1 (en) 2011-09-12 2014-06-11 Idenix Pharmaceuticals Inc SUBSTITUTED CARBONYLOXYMETHYLPHOSPHORAMIDATE COMPOUNDS AND PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF VIRAL INFECTIONS
US8889860B2 (en) 2011-09-13 2014-11-18 Lasergen, Inc. 3′-OH unblocked, fast photocleavable terminating nucleotides and methods for nucleic acid sequencing
CA2840242C (en) 2011-09-16 2019-03-26 Gilead Sciences, Inc. Methods for treating hcv
US8507460B2 (en) 2011-10-14 2013-08-13 Idenix Pharmaceuticals, Inc. Substituted 3′,5′-cyclic phosphates of purine nucleotide compounds and pharmaceutical compositions for the treatment of viral infections
EP2780026B1 (en) 2011-11-15 2019-10-23 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors
US8889159B2 (en) 2011-11-29 2014-11-18 Gilead Pharmasset Llc Compositions and methods for treating hepatitis C virus
CA2856722C (en) 2011-11-30 2022-11-22 Emory University Antiviral jak inhibitors useful in treating or preventing retroviral and other viral infections
WO2013096680A1 (en) 2011-12-22 2013-06-27 Alios Biopharma, Inc. Substituted phosphorothioate nucleotide analogs
US20140356325A1 (en) 2012-01-12 2014-12-04 Ligand Pharmaceuticals Incorporated Novel 2'-c-methyl nucleoside derivative compounds
WO2013133927A1 (en) 2012-02-13 2013-09-12 Idenix Pharmaceuticals, Inc. Pharmaceutical compositions of 2'-c-methyl-guanosine, 5'-[2-[(3-hydroxy-2,2-dimethyl-1-oxopropyl)thio]ethyl n-(phenylmethyl)phosphoramidate]
CA2864098A1 (en) 2012-02-14 2013-08-22 University Of Georgia Research Foundation, Inc. Spiro[2.4]heptanes for treatment of flaviviridae infections
EP2828277A1 (en) 2012-03-21 2015-01-28 Vertex Pharmaceuticals Incorporated Solid forms of a thiophosphoramidate nucleotide prodrug
WO2013142157A1 (en) 2012-03-22 2013-09-26 Alios Biopharma, Inc. Pharmaceutical combinations comprising a thionucleotide analog
WO2013177219A1 (en) 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. D-amino acid compounds for liver disease
US9109001B2 (en) 2012-05-22 2015-08-18 Idenix Pharmaceuticals, Inc. 3′,5′-cyclic phosphoramidate prodrugs for HCV infection
WO2013177195A1 (en) 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. 3',5'-cyclic phosphate prodrugs for hcv infection
SG11201407336PA (en) 2012-05-25 2015-03-30 Janssen Sciences Ireland Uc Uracyl spirooxetane nucleosides
US9206412B2 (en) 2012-05-31 2015-12-08 Colorado State University Research Foundation Thioxothiazolidine inhibitors
WO2014052638A1 (en) 2012-09-27 2014-04-03 Idenix Pharmaceuticals, Inc. Esters and malonates of sate prodrugs
US10513534B2 (en) 2012-10-08 2019-12-24 Idenix Pharmaceuticals Llc 2′-chloro nucleoside analogs for HCV infection
EP2909223B1 (en) 2012-10-19 2017-03-22 Idenix Pharmaceuticals LLC Dinucleotide compounds for hcv infection
WO2014066239A1 (en) 2012-10-22 2014-05-01 Idenix Pharmaceuticals, Inc. 2',4'-bridged nucleosides for hcv infection
US20140140951A1 (en) 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. D-Alanine Ester of Rp-Nucleoside Analog
WO2014078436A1 (en) 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. D-alanine ester of sp-nucleoside analog
EP2935304A1 (en) 2012-12-19 2015-10-28 IDENIX Pharmaceuticals, Inc. 4'-fluoro nucleosides for the treatment of hcv
KR102532198B1 (en) 2012-12-21 2023-05-11 얀센 바이오파마, 인코퍼레이트. Substituted nucleosides, nucleotides and analogs thereof
CN105748499B (en) 2013-01-31 2018-12-28 吉利德制药有限责任公司 The combination preparation of two antiviral compounds
US10034893B2 (en) 2013-02-01 2018-07-31 Enanta Pharmaceuticals, Inc. 5, 6-D2 uridine nucleoside/tide derivatives
WO2014121417A1 (en) 2013-02-07 2014-08-14 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds and methods of use thereof for the treatment of hepatitis c
WO2014121418A1 (en) 2013-02-07 2014-08-14 Merck Sharp & Dohme Corp. Tetracyclic heterocycle compounds and methods of use thereof for the treatment of hepatitis c
US9309275B2 (en) 2013-03-04 2016-04-12 Idenix Pharmaceuticals Llc 3′-deoxy nucleosides for the treatment of HCV
WO2014137930A1 (en) 2013-03-04 2014-09-12 Idenix Pharmaceuticals, Inc. Thiophosphate nucleosides for the treatment of hcv
EP2970357A1 (en) 2013-03-13 2016-01-20 IDENIX Pharmaceuticals, Inc. Amino acid phosphoramidate pronucleotides of 2'-cyano, azido and amino nucleosides for the treatment of hcv
RU2534613C2 (en) 2013-03-22 2014-11-27 Александр Васильевич Иващенко Alkyl2-{[(2r,3s,5r)-5-(4-amino-2-oxo-2h-pyrimidine-1-yl)- -hydroxy- tetrahydro-furan-2-ylmethoxy]-phenoxy-phosphorylamino}-proptonates, nucleoside inhibitors of rna-polymerase hcv ns5b, methods for producing and using them
US9187515B2 (en) 2013-04-01 2015-11-17 Idenix Pharmaceuticals Llc 2′,4′-fluoro nucleosides for the treatment of HCV
BR112015025716A2 (en) 2013-04-12 2017-07-18 Achillion Pharmaceuticals Inc Deuterium Nucleoside Prodrugs Useful for Hcv Treatment
EP3004130B1 (en) 2013-06-05 2019-08-07 Idenix Pharmaceuticals LLC. 1',4'-thio nucleosides for the treatment of hcv
EP3027636B1 (en) 2013-08-01 2022-01-05 Idenix Pharmaceuticals LLC D-amino acid phosphoramidate pronucleotides of halogeno pyrimidine compounds for liver disease
MX2016002185A (en) 2013-08-27 2016-06-06 Gilead Pharmasset Llc Combination formulation of two antiviral compounds.
JP6762873B2 (en) 2013-09-11 2020-09-30 エモリー・ユニバーシテイ Nucleotide and nucleoside compositions and related uses
WO2015042375A1 (en) 2013-09-20 2015-03-26 Idenix Pharmaceuticals, Inc. Hepatitis c virus inhibitors
WO2015061683A1 (en) 2013-10-25 2015-04-30 Idenix Pharmaceuticals, Inc. D-amino acid phosphoramidate and d-alanine thiophosphoramidate pronucleotides of nucleoside compounds useful for the treatment of hcv
EP3063165A1 (en) 2013-11-01 2016-09-07 Idenix Pharmaceuticals LLC D-alanine phosphoramidate pronucleotides of 2'-methyl 2'-fluoro guanosine nucleoside compounds for the treatment of hcv
EP3074399A1 (en) 2013-11-27 2016-10-05 Idenix Pharmaceuticals LLC 2'-dichloro and 2'-fluoro-2'-chloro nucleoside analogues for hcv infection
EP3083654A1 (en) 2013-12-18 2016-10-26 Idenix Pharmaceuticals LLC 4'-or nucleosides for the treatment of hcv
EP3623364A1 (en) 2014-02-13 2020-03-18 Ligand Pharmaceuticals, Inc. Prodrug compounds and their uses
WO2015134561A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Pharmaceutical compositions comprising a 5,5-fused heteroarylene flaviviridae inhibitor and their use for treating or preventing flaviviridae infection
EP3114122A1 (en) 2014-03-05 2017-01-11 Idenix Pharmaceuticals LLC Solid forms of a flaviviridae virus inhibitor compound and salts thereof
EP3113763A1 (en) 2014-03-05 2017-01-11 Idenix Pharmaceuticals LLC Solid prodrug forms of 2'-chloro-2'-methyl uridine for hcv
US10202411B2 (en) 2014-04-16 2019-02-12 Idenix Pharmaceuticals Llc 3′-substituted methyl or alkynyl nucleosides nucleotides for the treatment of HCV
JP2017520545A (en) 2014-07-02 2017-07-27 リガンド・ファーマシューティカルズ・インコーポレイテッド Prodrug compounds and their use
US9675632B2 (en) 2014-08-26 2017-06-13 Enanta Pharmaceuticals, Inc. Nucleoside and nucleotide derivatives
TWI740546B (en) 2014-10-29 2021-09-21 美商基利科學股份有限公司 Methods for the preparation of ribosides
US9718851B2 (en) 2014-11-06 2017-08-01 Enanta Pharmaceuticals, Inc. Deuterated nucleoside/tide derivatives
US9732110B2 (en) 2014-12-05 2017-08-15 Enanta Pharmaceuticals, Inc. Nucleoside and nucleotide derivatives
US20190022116A1 (en) 2014-12-26 2019-01-24 Emory University N4-Hydroxycytidine and Derivatives and Anti-Viral Uses Related Thereto
CA3182565A1 (en) 2015-03-06 2016-09-15 Atea Pharmaceuticals, Inc. .beta.-d-2'-deoxy-2'-.alpha.-fluoro-2'-.beta.-c-substituted-2-modified-n6-substituted purine nucleotides for hcv treatment
MX2021005087A (en) 2015-09-16 2022-08-18 Gilead Sciences Inc Methods for treating arenaviridae and coronaviridae virus infections.
CN109562113A (en) 2016-05-10 2019-04-02 C4医药公司 Loop coil degron body for target protein degradation
EP3455218A4 (en) 2016-05-10 2019-12-18 C4 Therapeutics, Inc. C3-carbon linked glutarimide degronimers for target protein degradation
EP3454856A4 (en) 2016-05-10 2019-12-25 C4 Therapeutics, Inc. Heterocyclic degronimers for target protein degradation
US10202412B2 (en) 2016-07-08 2019-02-12 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-substituted-4′-substituted-2-substituted-N6-substituted-6-aminopurinenucleotides for the treatment of paramyxovirus and orthomyxovirus infections
US10711029B2 (en) 2016-07-14 2020-07-14 Atea Pharmaceuticals, Inc. Beta-d-2′-deoxy-2′-alpha-fluoro-2′-beta-c-substituted-4′fluoro-n6-substituted-6-amino-2-substituted purine nucleotides for the treatment of hepatitis c virus infection
US20190169221A1 (en) * 2016-08-12 2019-06-06 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
PT3512863T (en) 2016-09-07 2022-03-09 Atea Pharmaceuticals Inc 2'-substituted-n6-substituted purine nucleotides for rna virus treatment
CA3197567A1 (en) 2017-02-01 2018-08-09 Atea Pharmaceuticals, Inc. Nucleotide hemi-sulfate salt for the treatment of hepatitis c virus
EP3595672B1 (en) 2017-03-14 2023-09-06 Gilead Sciences, Inc. Compounds for use in methods of treating feline coronavirus infections
AR111490A1 (en) 2017-05-01 2019-07-17 Gilead Sciences Inc CRYSTALLINE FORMS OF PROPANOATE OF (S) -2-ETILBUTIL 2 - (((S) - (((2R, 3S, 4R, 5R) -5- (4-AMINOPIRROLO [2,1-F] [1,2, 4] TRIAZIN-7-IL) -5-CIANO-3,4-DIHYDROXITETRAHIDROFURAN-2-IL) METOXI) (FENOXI) FOSFORIL) AMINO)
EP3645836A4 (en) 2017-06-26 2021-04-07 HRL Laboratories, LLC System and method for generating output of a downhole inertial measurement unit
EP3651734A1 (en) 2017-07-11 2020-05-20 Gilead Sciences, Inc. Compositions comprising an rna polymerase inhibitor and cyclodextrin for treating viral infections
CA3082191C (en) 2017-12-07 2021-09-21 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
CN112351799A (en) 2018-04-10 2021-02-09 阿堤亚制药公司 Treatment of HCV infected patients with cirrhosis
CA3115712A1 (en) * 2018-10-17 2020-04-23 Xibin Liao 6-mercaptopurine nucleoside analogues
WO2021154687A1 (en) 2020-01-27 2021-08-05 Gilead Sciences, Inc. Methods for treating sars cov-2 infections
US10874687B1 (en) 2020-02-27 2020-12-29 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
WO2021183750A2 (en) 2020-03-12 2021-09-16 Gilead Sciences, Inc. Methods of preparing 1'-cyano nucleosides
EP4132651A1 (en) 2020-04-06 2023-02-15 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carbanucleoside analogs
US20210393653A1 (en) 2020-05-29 2021-12-23 Gilead Sciences, Inc. Remdesivir treatment methods
BR112022026321A2 (en) 2020-06-24 2023-01-17 Gilead Sciences Inc 1'-CYAN NUCLEOSIDE ANALOGS AND USES THEREOF
US11926645B2 (en) 2020-08-27 2024-03-12 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US20240002442A1 (en) * 2020-11-25 2024-01-04 Northeastern University Cyclic peptides with antimicrobial properties
EP4320128A1 (en) 2022-03-02 2024-02-14 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6812219B2 (en) * 2000-05-26 2004-11-02 Idenix Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
US6914054B2 (en) * 2000-05-23 2005-07-05 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus
US7582618B2 (en) * 2002-06-28 2009-09-01 Idenix Pharmaceuticals, Inc. 2′-C-methyl-3′-O-L-valine ester ribofuranosyl cytidine for treatment of flaviviridae infections
US7625875B2 (en) * 2002-06-28 2009-12-01 Idenix Pharmaceuticals, Inc. 2′ and 3′-nucleoside prodrugs for treating Flaviviridae infections
US7824851B2 (en) * 2002-11-15 2010-11-02 Idenix Pharmaceuticals, Inc. 2′-branched nucleosides and Flaviviridae mutation

Family Cites Families (335)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8841A (en) * 1852-03-30 Sice-httxieb
US28013A (en) * 1860-04-24 Improved bullet-ladle
US83307A (en) * 1868-10-20 Improvement in wash-boilehs
US87873A (en) * 1869-03-16 Perry prettyman
US6458772B1 (en) 1909-10-07 2002-10-01 Medivir Ab Prodrugs
US3074929A (en) 1955-08-11 1963-01-22 Burroughs Wellcome Co Glycosides of 6-mercaptopurine
GB924246A (en) 1958-12-23 1963-04-24 Wellcome Found Purine derivatives and their preparation
US3116282A (en) 1960-04-27 1963-12-31 Upjohn Co Pyrimidine nucleosides and process
GB984877A (en) 1962-08-16 1965-03-03 Waldhof Zellstoff Fab Improvements in and relating to 6-halonucleosides
GB1163103A (en) 1965-11-15 1969-09-04 Merck & Co Inc Ribofuranosyl Purine Derivatives
FR1498856A (en) 1965-11-15 1968-01-10
DE1695411A1 (en) 1966-05-02 1971-04-15 Merck & Co Inc Substituted purine nucleosides and processes for their preparation
FR1521076A (en) 1966-05-02 1968-04-12 Merck & Co Inc Substituted purine nucleosides
US3480613A (en) * 1967-07-03 1969-11-25 Merck & Co Inc 2-c or 3-c-alkylribofuranosyl - 1-substituted compounds and the nucleosides thereof
DE2122991C2 (en) 1971-05-04 1982-06-09 Schering Ag, 1000 Berlin Und 4619 Bergkamen Process for the preparation of cytosine and 6-azacytosine nucleosides
US3798209A (en) 1971-06-01 1974-03-19 Icn Pharmaceuticals 1,2,4-triazole nucleosides
USRE29835E (en) 1971-06-01 1978-11-14 Icn Pharmaceuticals 1,2,4-Triazole nucleosides
JPS4848495A (en) 1971-09-21 1973-07-09
US4022889A (en) 1974-05-20 1977-05-10 The Upjohn Company Therapeutic compositions of antibiotic U-44,590 and methods for using the same
DE2508312A1 (en) 1975-02-24 1976-09-02 Schering Ag NEW PROCESS FOR THE PRODUCTION OF NUCLEOSIDES
US4058602A (en) 1976-08-09 1977-11-15 The United States Of America As Represented By The Department Of Health, Education And Welfare Synthesis, structure, and antitumor activity of 5,6-dihydro-5-azacytidine
DE2757365A1 (en) 1977-12-20 1979-06-21 Schering Ag NEW PROCESS FOR THE PRODUCTION OF NUCLEOSIDES
DD140254A1 (en) 1978-12-04 1980-02-20 Dieter Baerwolff METHOD OF PREPARING 4-SUBSTITUTED PYRIMIDIN NUCLEOSIDES
DE2852721A1 (en) 1978-12-06 1980-06-26 Basf Ag METHOD FOR REPRESENTING POTASSIUM RIBONATE AND RIBONOLACTONE
US4239753A (en) 1978-12-12 1980-12-16 The Upjohn Company Composition of matter and process
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
FR2562543B1 (en) 1984-04-10 1987-09-25 Elf Aquitaine NOVEL CYCLIC PHOSPHONITES, THEIR PREPARATION AND APPLICATIONS
NL8403224A (en) 1984-10-24 1986-05-16 Oce Andeno Bv DIOXAPHOSPHORINANS, THEIR PREPARATION AND THE USE FOR SPLITTING OF OPTICALLY ACTIVE COMPOUNDS.
KR880000094B1 (en) 1984-12-07 1988-02-23 보령제약 주식회사 Preparation process for nucleoside derivative
JPS61204193A (en) 1985-03-05 1986-09-10 Takeda Chem Ind Ltd Production of cytosine nuceoside
US6448392B1 (en) 1985-03-06 2002-09-10 Chimerix, Inc. Lipid derivatives of antiviral nucleosides: liposomal incorporation and method of use
US5223263A (en) 1988-07-07 1993-06-29 Vical, Inc. Liponucleotide-containing liposomes
JPS61212592A (en) 1985-03-19 1986-09-20 Tokyo Tanabe Co Ltd Production of d-ribose
US4605659A (en) 1985-04-30 1986-08-12 Syntex (U.S.A.) Inc. Purinyl or pyrimidinyl substituted hydroxycyclopentane compounds useful as antivirals
JPS61263995A (en) 1985-05-16 1986-11-21 Takeda Chem Ind Ltd Production of cytosine nucleoside
US4754026A (en) 1985-06-04 1988-06-28 Takeda Chemical Industries, Ltd. Conversion of uracil derivatives to cytosine derivatives
US5455339A (en) 1986-05-01 1995-10-03 University Of Georgia Research Foundation, Inc. Method for the preparation of 2',3'-dideoxy and 2',3'-dideoxydide-hydro nucleosides
JPS63215894A (en) 1987-03-04 1988-09-08 Hitachi Ltd Motor pump with intensifier
JPH0699467B2 (en) 1987-03-04 1994-12-07 ヤマサ醤油株式会社 2 ▲ '▼ -Deoxy-2 ▲' ▼ (S) -alkylpyrimidine nucleoside derivative
DE3714473A1 (en) 1987-04-30 1988-11-10 Basf Ag CONTINUOUS PROCESS FOR EPIMERIZING SUGAR, ESPECIALLY FROM D-ARABINOSE TO D-RIBOSE
GB8719367D0 (en) 1987-08-15 1987-09-23 Wellcome Found Therapeutic compounds
US5246924A (en) 1987-09-03 1993-09-21 Sloan-Kettering Institute For Cancer Research Method for treating hepatitis B virus infections using 1-(2'-deoxy-2'-fluoro-beta-D-arabinofuranosyl)-5-ethyluracil
EP0380558A4 (en) 1987-09-22 1991-07-31 The Regents Of The University Of California Liposomal nucleoside analogues for treating aids
US4880784A (en) 1987-12-21 1989-11-14 Brigham Young University Antiviral methods utilizing ribofuranosylthiazolo[4,5-d]pyrimdine derivatives
US4996308A (en) * 1988-03-25 1991-02-26 Merrell Dow Pharmaceuticals Inc. Derivatives with unsaturated substitutions for the 5'-hydroxymethyl group
NZ229453A (en) 1988-06-10 1991-08-27 Univ Minnesota & Southern Rese A pharmaceutical composition containing purine derivatives with nucleosides such as azt, as antiviral agents
US5122517A (en) 1988-06-10 1992-06-16 Regents Of The University Of Minnesota Antiviral combination comprising nucleoside analogs
GB8815265D0 (en) 1988-06-27 1988-08-03 Wellcome Found Therapeutic nucleosides
US6252060B1 (en) 1988-07-07 2001-06-26 Nexstar Pharmaceuticals, Inc. Antiviral liponucleosides: treatment of hepatitis B
US6599887B2 (en) 1988-07-07 2003-07-29 Chimerix, Inc. Methods of treating viral infections using antiviral liponucleotides
SE8802687D0 (en) 1988-07-20 1988-07-20 Astra Ab NUCLEOSIDE DERIVATIVES
US5744600A (en) 1988-11-14 1998-04-28 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Phosphonomethoxy carbocyclic nucleosides and nucleotides
US5616702A (en) * 1988-11-15 1997-04-01 Merrell Pharmaceuticals Inc. 2-'-ethenylidene cytidine, uridine and guanosine derivatives
US5705363A (en) 1989-03-02 1998-01-06 The Women's Research Institute Recombinant production of human interferon τ polypeptides and nucleic acids
US5411947A (en) 1989-06-28 1995-05-02 Vestar, Inc. Method of converting a drug to an orally available form by covalently bonding a lipid to the drug
US5194654A (en) 1989-11-22 1993-03-16 Vical, Inc. Lipid derivatives of phosphonoacids for liposomal incorporation and method of use
US5118672A (en) 1989-07-10 1992-06-02 University Of Georgia Research Foundation 5'-diphosphohexose nucleoside pharmaceutical compositions
US5463092A (en) 1989-11-22 1995-10-31 Vestar, Inc. Lipid derivatives of phosphonacids for liposomal incorporation and method of use
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
US6060592A (en) 1990-01-11 2000-05-09 Isis Pharmaceuticals, Inc. Pyrimidine nucleoside compounds and oligonucleoside compounds containing same
US5200514A (en) 1990-01-19 1993-04-06 University Of Georgia Research Foundation, Inc. Synthesis of 2'-deoxypyrimidine nucleosides
US5204466A (en) 1990-02-01 1993-04-20 Emory University Method and compositions for the synthesis of bch-189 and related compounds
WO1991016920A1 (en) 1990-05-07 1991-11-14 Vical, Inc. Lipid prodrugs of salicylate and nonsteroidal anti-inflammatory drugs
FR2662165B1 (en) 1990-05-18 1992-09-11 Univ Paris Curie BRANCHED NUCLEOSIDE DERIVATIVES, THEIR PREPARATION PROCESS AND THEIR USE AS MEDICAMENTS.
EP0531452A4 (en) 1990-05-29 1993-06-09 Vical, Inc. Synthesis of glycerol di- and triphosphate derivatives
CA2083961A1 (en) 1990-05-29 1991-11-30 Henk Van Den Bosch Synthesis of glycerol di- and triphosphate derivatives
DK0533833T3 (en) 1990-06-13 1996-04-22 Arnold Glazier Phosphorus prodrugs
US5627165A (en) 1990-06-13 1997-05-06 Drug Innovation & Design, Inc. Phosphorous prodrugs and therapeutic delivery systems using same
JP2559917B2 (en) * 1990-06-15 1996-12-04 三共株式会社 Pyrimidine nucleoside derivative
US5372808A (en) 1990-10-17 1994-12-13 Amgen Inc. Methods and compositions for the treatment of diseases with consensus interferon while reducing side effect
US5543390A (en) 1990-11-01 1996-08-06 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
US5256641A (en) 1990-11-01 1993-10-26 State Of Oregon Covalent polar lipid-peptide conjugates for immunological targeting
US5543389A (en) 1990-11-01 1996-08-06 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education On Behalf Of The Oregon Health Sciences University, A Non Profit Organization Covalent polar lipid-peptide conjugates for use in salves
US5827819A (en) 1990-11-01 1998-10-27 Oregon Health Sciences University Covalent polar lipid conjugates with neurologically active compounds for targeting
US5149794A (en) 1990-11-01 1992-09-22 State Of Oregon Covalent lipid-drug conjugates for drug targeting
IL100502A (en) 1991-01-03 1995-12-08 Iaf Biochem Int Pharmaceutical compositions containing cis-4-amino-1(hydroxymethyl-1,3-oxathiolan-5-yl)-1H-pyrimid-2-one nucleoside or its derivatives
JPH04266880A (en) 1991-02-22 1992-09-22 Japan Tobacco Inc Production of 3-dpa-lactone
ATE361293T1 (en) 1991-03-06 2007-05-15 Univ Emory SALTS AND AMIDES OF (-)CIS 5-FLUORO-2'-DEOXY-3'- THIACYTIDINES SUITABLE FOR THE TREATMENT OF HEPATITIS B
WO1992018517A1 (en) 1991-04-17 1992-10-29 Yale University Method of treating or preventing hepatitis b virus
US5157027A (en) 1991-05-13 1992-10-20 E. R. Squibb & Sons, Inc. Bisphosphonate squalene synthetase inhibitors and method
WO1993000910A1 (en) 1991-07-12 1993-01-21 Vical, Inc. Antiviral liponucleosides: treatment of hepatitis b
JPH0525152A (en) 1991-07-22 1993-02-02 Japan Tobacco Inc Production of 3-dpa-lactone
US5554728A (en) 1991-07-23 1996-09-10 Nexstar Pharmaceuticals, Inc. Lipid conjugates of therapeutic peptides and protease inhibitors
TW224053B (en) 1991-09-13 1994-05-21 Paul B Chretien
US5676942A (en) 1992-02-10 1997-10-14 Interferon Sciences, Inc. Composition containing human alpha interferon species proteins and method for use thereof
DE4207363A1 (en) 1992-03-04 1993-09-09 Max Delbrueck Centrum ANTIVIRAL NUCLEOSIDE ANALOGS, THEIR PRODUCTION AND THEIR PHARMACEUTICAL USE
US5371210A (en) 1992-06-22 1994-12-06 Eli Lilly And Company Stereoselective fusion glycosylation process for preparing 2'-deoxy-2',2'-difluoronucleosides and 2'-deoxy-2'-fluoronucleosides
US5606048A (en) 1992-06-22 1997-02-25 Eli Lilly And Company Stereoselective glycosylation process for preparing 2'-Deoxy-2', 2'-difluoronucleosides and 2'-deoxy-2'-fluoronucleosides
US5401861A (en) 1992-06-22 1995-03-28 Eli Lilly And Company Low temperature process for preparing alpha-anomer enriched 2-deoxy-2,2-difluoro-D-ribofuranosyl sulfonates
US5821357A (en) 1992-06-22 1998-10-13 Eli Lilly And Company Stereoselective glycosylation process for preparing 2'-deoxy-2',2'-difluoropurine and triazole nucleosides
US5256797A (en) 1992-06-22 1993-10-26 Eli Lilly And Company Process for separating 2-deoxy-2,2-difluoro-D-ribofuranosyl alkylsulfonate anomers
WO1994001117A1 (en) 1992-07-02 1994-01-20 The Wellcome Foundation Limited Therapeutic nucleosides
DE4224737A1 (en) 1992-07-27 1994-02-03 Herbert Prof Dr Schott New cytosine analogues with lipophilic protected amino gps. - for treatment of cancer and virus diseases e.g. AIDS, are more protected against enzymatic des-amination and can be used in higher doses than unprotected cpds.
US5256924A (en) * 1992-08-10 1993-10-26 Allied-Signal Inc. Superconducting commutator for DC machines
CA2105112C (en) 1992-09-01 2005-08-02 Thomas C. Britton A process for anomerizing nucleosides
ATE202383T1 (en) 1992-09-10 2001-07-15 Isis Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR THE TREATMENT OF HEPATITIS C VIRUS-ASSOCIATED DISEASES
US6423489B1 (en) 1992-09-10 2002-07-23 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus-associated diseases
US6174868B1 (en) 1992-09-10 2001-01-16 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of hepatitis C virus-associated diseases
US6433159B1 (en) 1992-09-10 2002-08-13 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus associated diseases
US6391542B1 (en) 1992-09-10 2002-05-21 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus-associated diseases
JPH06135988A (en) 1992-10-22 1994-05-17 Toagosei Chem Ind Co Ltd Nucleotide derivative
GB9226729D0 (en) 1992-12-22 1993-02-17 Wellcome Found Therapeutic combination
JPH06211890A (en) 1993-01-12 1994-08-02 Yamasa Shoyu Co Ltd 2'-deoxy-2'@(3754/24)s)-substituted alkylcytidine derivative
JPH06228186A (en) 1993-01-29 1994-08-16 Yamasa Shoyu Co Ltd 2'-deoxy-@(3754/24)2's)-alkylpyrimidine nucleoside derivative
CA2156394A1 (en) 1993-02-24 1994-09-01 Jui H. Wang Compositions and methods of application of reactive antiviral polymers
GB9307043D0 (en) 1993-04-05 1993-05-26 Norsk Hydro As Chemical compounds
JP3312951B2 (en) 1993-04-08 2002-08-12 三菱化学株式会社 Olefin-based thermoplastic resin composition
JPH08510236A (en) 1993-05-12 1996-10-29 カール ワイ. ホステトラー Acyclovir derivative for topical use
WO1995003056A1 (en) 1993-07-19 1995-02-02 Tokyo Tanabe Company Limited Hepatitis c virus proliferation inhibitor
US6156501A (en) 1993-10-26 2000-12-05 Affymetrix, Inc. Arrays of modified nucleic acid probes and methods of use
US5587362A (en) 1994-01-28 1996-12-24 Univ. Of Ga Research Foundation L-nucleosides
US5908821A (en) 1994-05-11 1999-06-01 Procter & Gamble Company Dye transfer inhibiting compositions with specifically selected metallo catalysts
WO1996001115A1 (en) 1994-07-01 1996-01-18 Pro-Neuron, Inc. Pyrimidine nucleotide precursors for treatment of systemic inflammation and inflammatory hepatitis
DE4432623A1 (en) 1994-09-14 1996-03-21 Huels Chemische Werke Ag Process for bleaching aqueous surfactant solutions
US5696277A (en) 1994-11-15 1997-12-09 Karl Y. Hostetler Antiviral prodrugs
DK0747389T3 (en) 1994-12-13 2004-01-26 Taiho Pharmaceutical Co Ltd 3'-substituted nucleoside derivatives
GB9505025D0 (en) 1995-03-13 1995-05-03 Medical Res Council Chemical compounds
JP3786447B2 (en) 1995-03-31 2006-06-14 エーザイ株式会社 Preventive and therapeutic agent for hepatitis C
DE19513330A1 (en) 1995-04-03 1996-10-10 Schering Ag New process for the production of nucleosides
US5977061A (en) * 1995-04-21 1999-11-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic N6 - substituted nucleotide analagues and their use
JPH0959292A (en) 1995-08-25 1997-03-04 Yamasa Shoyu Co Ltd Production of 4-aminopyrimidine nucleoside
CA2231442A1 (en) 1995-09-07 1997-03-13 University Of Georgia Research Foundation, Inc. Therapeutic azide compounds
ATE318896T1 (en) 1995-09-27 2006-03-15 Univ Emory RECOMBINANT RNA REPLICASE OF HEPATITIS C VIRUS
US5908621A (en) 1995-11-02 1999-06-01 Schering Corporation Polyethylene glycol modified interferon therapy
US5980884A (en) 1996-02-05 1999-11-09 Amgen, Inc. Methods for retreatment of patients afflicted with Hepatitis C using consensus interferon
US5633388A (en) 1996-03-29 1997-05-27 Viropharma Incorporated Compounds, compositions and methods for treatment of hepatitis C
US5830905A (en) 1996-03-29 1998-11-03 Viropharma Incorporated Compounds, compositions and methods for treatment of hepatitis C
GB9609932D0 (en) 1996-05-13 1996-07-17 Hoffmann La Roche Use of IL-12 and IFN alpha for the treatment of infectious diseases
US5891874A (en) 1996-06-05 1999-04-06 Eli Lilly And Company Anti-viral compound
JP3927630B2 (en) 1996-09-27 2007-06-13 エーザイ・アール・アンド・ディー・マネジメント株式会社 Preventive and therapeutic agents for viral infections
US5922757A (en) 1996-09-30 1999-07-13 The Regents Of The University Of California Treatment and prevention of hepatic disorders
KR100412480B1 (en) * 1996-10-16 2003-12-31 아이씨엔 파마슈티컬스, 인코포레이티드 Purine l-nucleosides, analogs and uses thereof
CA2267279A1 (en) * 1996-10-16 1998-04-23 Devron Averett Monocyclic l-nucleosides, analogs and uses thereof
AP1019A (en) 1996-10-18 2001-10-16 Vertex Pharma Inhibitors of serinre proteases, particularly hepatitis C virus NS3 protease.
CA2269213A1 (en) * 1996-10-28 1998-05-07 The University Of Washington Induction of viral mutation by incorporation of miscoding ribonucleoside analogs into viral rna
GB9623908D0 (en) 1996-11-18 1997-01-08 Hoffmann La Roche Amino acid derivatives
US6248878B1 (en) 1996-12-24 2001-06-19 Ribozyme Pharmaceuticals, Inc. Nucleoside analogs
BR9807473A (en) 1997-01-17 2000-03-21 Icn Pharmaceuticals Cytokine-related disease treatments
EA001920B1 (en) 1997-03-19 2001-10-22 Эмори Юниверсити Synthesis, anti-human immunodeficiency virus and anti-hepatitus b virus activities of 1,3-oxaselenolane nucleosides
UA61962C2 (en) 1997-06-30 2003-12-15 Мерц Фарма Гмбх Унд Ко. Кгаа 1-amino-alkylcyclohexane nmda receptors antagonists
HUP0100100A3 (en) 1997-08-11 2001-12-28 Boehringer Ingelheim Ca Ltd Hepatitis c inhibitor peptide analogues, pharmaceutical compositions comprising thereof and their use
US6172046B1 (en) 1997-09-21 2001-01-09 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in patients having chronic Hepatitis C infection
US6472373B1 (en) 1997-09-21 2002-10-29 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in antiviral treatment naive patients having chronic hepatitis C infection
ES2186660T3 (en) 1997-09-21 2003-05-16 Schering Corp COMBINATION THERAPY TO ERADICATE HCV-RNA DETECTABLE IN PATIENTS WITH CHRONIC HEPATITIS C INFECTION.
AU758426B2 (en) 1997-10-30 2003-03-20 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Nucleosides for imaging and treatment applications
WO1999040916A1 (en) 1998-02-12 1999-08-19 G.D. Searle & Co. Use of n-substituted-1,5-dideoxy-1,5-imino-d-glucitol compounds for treating hepatitis virus infections
WO1999043690A1 (en) 1998-02-25 1999-09-02 Rational Drug Design Laboratories L-4'-arabinofuranonucleoside compound and medicine composition comprising the same
CA2322008C (en) 1998-02-25 2011-06-28 Emory University 2'-fluoronucleosides
US6312662B1 (en) 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds
ES2401070T3 (en) 1998-03-06 2013-04-16 Metabasis Therapeutics, Inc. New prodrugs for phosphorus-containing compounds
KR100389853B1 (en) 1998-03-06 2003-08-19 삼성전자주식회사 Method for recording and reproducing catalog information
GB9806815D0 (en) 1998-03-30 1998-05-27 Hoffmann La Roche Amino acid derivatives
TW466112B (en) 1998-04-14 2001-12-01 Lilly Co Eli Novel use of 2'-deoxy-2',2'-difluorocytidine for immunosuppressive therapy and pharmaceutical composition comprising the same
WO1999059621A1 (en) 1998-05-15 1999-11-25 Schering Corporation Combination therapy comprising ribavirin and interferon alpha in antiviral treatment naive patients having g chronic hepatitis c infection
US6833361B2 (en) 1998-05-26 2004-12-21 Ribapharm, Inc. Nucleosides having bicyclic sugar moiety
JP3839667B2 (en) 1998-06-08 2006-11-01 エフ.ホフマン−ラ ロシュ アーゲー Use of Peg-INF-alpha and ribavirin for the treatment of chronic hepatitis C
US6444652B1 (en) 1998-08-10 2002-09-03 Novirio Pharmaceuticals Limited β-L-2'-deoxy-nucleosides for the treatment of hepatitis B
DE122007000062I1 (en) * 1998-08-10 2007-12-20 Idenix Cayman Ltd Beta-L-2'-deoxynucleosides for the treatment of hepatitis B virus
US6277830B1 (en) 1998-10-16 2001-08-21 Schering Corporation 5′-amino acid esters of ribavirin and the use of same to treat hepatitis C with interferon
CA2252144A1 (en) 1998-10-16 2000-04-16 University Of Alberta Dual action anticancer prodrugs
WO2000025799A1 (en) 1998-11-05 2000-05-11 Centre National De La Recherche Scientifique β-L-2'-DEOXY-NUCLEOSIDES FOR THE TREATMENT OF HIV INFECTION
US6458773B1 (en) 1998-11-05 2002-10-01 Emory University Nucleoside with anti-hepatitis B virus activity
JP2003507322A (en) 1998-12-18 2003-02-25 シェリング・コーポレーション Ribavirin-PEGylated interferon-α-induced HCV combination therapy
EP1175425A2 (en) 1999-01-21 2002-01-30 The Board Of Regents, The University Of Texas System Inhibitors of intestinal apical membrane na/phosphate co-transportation
BR0008135A (en) 1999-02-12 2002-02-19 Glaxo Group Ltd Compound, methods for treating a virus infection in a human and for treating a hepatitis B virus infection in a human, pharmaceutical composition, use of a compound, method for releasing a compound, patient package, process for preparation of a compound
JP2003505501A (en) 1999-02-12 2003-02-12 ジー・ディー・サール・アンド・カンパニー Glucamine compounds for the treatment of hepatitis virus infection
EP1155017B1 (en) * 1999-02-22 2003-01-15 Shire Biochem Inc. [1,8] naphthyridine derivatives having antiviral activity
EP1724276A1 (en) 1999-03-05 2006-11-22 Metabasis Therapeutics, Inc. Novel phosphorus-containing prodrugs
US6831069B2 (en) * 1999-08-27 2004-12-14 Ribapharm Inc. Pyrrolo[2,3-d]pyrimidine nucleoside analogs
BR0007050A (en) 1999-09-01 2001-07-31 Matsushita Electric Ind Co Ltd Digital data copyright protection system
WO2001018013A1 (en) 1999-09-08 2001-03-15 Metabasis Therapeutics, Inc. Prodrugs for liver specific drug delivery
US6566365B1 (en) 1999-11-04 2003-05-20 Biochem Pharma Inc. Method for the treatment of Flaviviridea viral infection using nucleoside analogues
MXPA02006156A (en) 1999-12-22 2003-09-22 Metabasis Therapeutics Inc Novel bisamidate phosphonate prodrugs.
WO2001049700A1 (en) 1999-12-30 2001-07-12 Biochem Pharma Inc. Imidazopyrimidine nucleoside analogues with anti-hiv activity
US20050119310A1 (en) 2000-02-14 2005-06-02 Mueller Richard A. Use of n-substituted-1,5-dideoxy-1,5-imino-d-glucitol compounds for treating hepatitis virus infections
US6455508B1 (en) 2000-02-15 2002-09-24 Kanda S. Ramasamy Methods for treating diseases with tirazole and pyrrolo-pyrimidine ribofuranosyl nucleosides
US7056895B2 (en) * 2000-02-15 2006-06-06 Valeant Pharmaceuticals International Tirazole nucleoside analogs and methods for using same
US6495677B1 (en) 2000-02-15 2002-12-17 Kanda S. Ramasamy Nucleoside compounds
EA200200778A1 (en) * 2000-02-18 2003-06-26 Шайре Байокем Инк. METHOD OF TREATMENT OR PREVENTION OF HEPATITIS C INFECTION IN THE ORGANISM ORGANISM, PHARMACEUTICAL COMPOSITION AGAINST FLAVIVIRUS, CONNECTION OF FORMULA Ib - AN ACTIVE AGENT FOR THE TREATMENT OR PREVENTION OF EFFECTECH EFFECTURES Ib - AN ACTIVE AGENT FOR THE TREATMENT OR PREVENTION INEKEKHEKUSA Ib - AN ACTIVE AGENT FOR THE TREATMENT OR PROTECTION
FR2806095A1 (en) 2000-03-10 2001-09-14 Gentech New polynucleotides for producing transgenic plants resistant to geminivirus infection comprising polynucleotides encoding proteins which interact with at least one of the products of the geminivirus genome
DE60136620D1 (en) * 2000-04-13 2009-01-02 Pharmasset Inc 3 OR 2 HYDROXYMETHYL SUBSTITUTED NUCLEOSIDE DERIVATIVES AND THEIR USE FOR THE TREATMENT OF VIRUS INFECTIONS
EP1282632A1 (en) 2000-04-20 2003-02-12 Schering Corporation Ribavirin-interferon alfa combination therapy for eradicating detectable hcv-rna in patients having chronic hepatitis c infection
US6787526B1 (en) * 2000-05-26 2004-09-07 Idenix Pharmaceuticals, Inc. Methods of treating hepatitis delta virus infection with β-L-2′-deoxy-nucleosides
IL153078A0 (en) 2000-05-26 2003-06-24 Idenix Cayman Ltd Methods for treating hepatitis delta virus infection with beta-l-2' deoxy nucleosides
US6875751B2 (en) 2000-06-15 2005-04-05 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
MY141594A (en) 2000-06-15 2010-05-14 Novirio Pharmaceuticals Ltd 3'-PRODRUGS OF 2'-DEOXY-ß-L-NUCLEOSIDES
US6815542B2 (en) * 2000-06-16 2004-11-09 Ribapharm, Inc. Nucleoside compounds and uses thereof
CA2409778A1 (en) 2000-06-20 2001-12-27 Incyte Genomics, Inc. Secreted proteins
JP3629187B2 (en) * 2000-06-28 2005-03-16 株式会社東芝 ELECTRIC FUSE, SEMICONDUCTOR DEVICE HAVING THE ELECTRIC FUSE, AND METHOD FOR MANUFACTURING THE SAME
UA72612C2 (en) 2000-07-06 2005-03-15 Pyrido[2.3-d]pyrimidine and pyrimido[4.5-d]pyrimidine nucleoside analogues, prodrugs and method for inhibiting growth of neoplastic cells
US6316174B1 (en) * 2000-08-24 2001-11-13 Eastman Kodak Company Color negative film
US20030008841A1 (en) 2000-08-30 2003-01-09 Rene Devos Anti-HCV nucleoside derivatives
SV2003000617A (en) 2000-08-31 2003-01-13 Lilly Co Eli INHIBITORS OF PROTEASA PEPTIDOMIMETICA REF. X-14912M
CA2421040C (en) 2000-09-01 2010-01-12 Ribozyme Pharmaceuticals, Incorporated Methods for synthesizing nucleosides, nucleoside derivatives and non-nucleoside derivatives
CN101862345B (en) 2000-10-18 2014-06-04 吉利德制药有限责任公司 Modified nucleosides for treatment of viral infections and abnormal cellular proliferation
WO2002033128A2 (en) 2000-10-18 2002-04-25 Pharmasset Limited Multiplex quantification of nucleic acids in diseased cells
AU2002213343A1 (en) 2000-10-18 2002-04-29 Schering Corporation Ribavirin-pegylated interferon alfa HCV combination therapy
EP1366055A2 (en) 2000-12-15 2003-12-03 Pharmasset Limited Antiviral agents for treatment of flaviviridae infections
WO2002057287A2 (en) 2001-01-22 2002-07-25 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
NZ540956A (en) 2001-03-01 2007-01-26 Pharmasset Inc Method for the synthesis of 2',3'-dideoxy-2',3'-didehydronucleosides
GB0112617D0 (en) 2001-05-23 2001-07-18 Hoffmann La Roche Antiviral nucleoside derivatives
GB0114286D0 (en) 2001-06-12 2001-08-01 Hoffmann La Roche Nucleoside Derivatives
WO2003000200A2 (en) 2001-06-22 2003-01-03 Pharmasset Ltd. β-2'-OR 3'-HALONUCLEOSIDES
WO2003024461A1 (en) 2001-09-20 2003-03-27 Schering Corporation Hcv combination therapy
US20030070752A1 (en) 2001-09-27 2003-04-17 Kevin Bergevin Method of manufacture for fluid handling barrier ribbon with polymeric tubes
US20040006002A1 (en) 2001-09-28 2004-01-08 Jean-Pierre Sommadossi Methods and compositions for treating flaviviruses and pestiviruses using 4'-modified nucleoside
AU2002330154A1 (en) 2001-09-28 2003-04-07 Centre National De La Recherche Scientifique Methods and compositions for treating hepatitis c virus using 4'-modified nucleosides
WO2003039523A2 (en) 2001-11-05 2003-05-15 Exiqon A/S OLIGONUCLEOTIDES MODIFIED WITH NOVEL α-L-RNA ANALOGUES
EP1569652A4 (en) 2001-12-14 2008-07-02 Pharmasset Inc N sp 4 /sp-acylcytosine nucleosides for treatment of viral iinfections
WO2003051899A1 (en) 2001-12-17 2003-06-26 Ribapharm Inc. Deazapurine nucleoside libraries and compounds
US6965066B1 (en) 2002-01-16 2005-11-15 Actodyne General, Inc. Elongated string support for a stringed musical instrument
WO2003061385A1 (en) 2002-01-17 2003-07-31 Ribapharm Inc. Tricyclic nucleoside library compounds, synthesis, and use as antiviral agents
AU2002341942A1 (en) 2002-01-17 2003-09-02 Ribapharm Inc. Sugar modified nucleosides as viral replication inhibitors
WO2003062256A1 (en) 2002-01-17 2003-07-31 Ribapharm Inc. 2'-beta-modified-6-substituted adenosine analogs and their use as antiviral agents
WO2003068244A1 (en) 2002-02-13 2003-08-21 Merck & Co., Inc. Methods of inhibiting orthopoxvirus replication with nucleoside compounds
CN1646534A (en) 2002-02-14 2005-07-27 法玛塞特有限公司 Modified fluorinated nucleoside analogues
WO2003072757A2 (en) 2002-02-28 2003-09-04 Biota, Inc. Nucleotide mimics and their prodrugs
AU2003214199A1 (en) 2002-03-18 2003-10-08 Massachusetts Institute Of Technology Event-driven charge-coupled device design and applications therefor
EP2306091A3 (en) * 2002-04-26 2012-12-26 Rolls-Royce Corporation Fuel premixing module for gas turbine engine combustor
US7247621B2 (en) 2002-04-30 2007-07-24 Valeant Research & Development Antiviral phosphonate compounds and methods therefor
EP1501850A2 (en) * 2002-05-06 2005-02-02 Genelabs Technologies, Inc. Nucleoside derivatives for treating hepatitis c virus infection
US20040014957A1 (en) 2002-05-24 2004-01-22 Anne Eldrup Oligonucleotides having modified nucleoside units
WO2003100017A2 (en) 2002-05-24 2003-12-04 Isis Pharmaceuticals, Inc. Oligonucleotides having modified nucleoside units
AU2003251524A1 (en) 2002-06-17 2003-12-31 Isis Pharmaceuticals, Inc. Carbocyclic nucleoside analogs as RNA-antivirals
DE10226932A1 (en) 2002-06-17 2003-12-24 Bayer Ag Radiation-curing coating agents
SE0201940D0 (en) 2002-06-20 2002-06-20 Astrazeneca Ab New combination II
US20070004669A1 (en) 2002-06-21 2007-01-04 Carroll Steven S Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
EP1572945A2 (en) 2002-06-27 2005-09-14 Merck & Co., Inc. Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
PT1523489E (en) 2002-06-28 2014-06-24 Centre Nat Rech Scient Modified 2' and 3' -nucleoside produgs for treating flaviridae infections
US7608600B2 (en) 2002-06-28 2009-10-27 Idenix Pharmaceuticals, Inc. Modified 2′ and 3′-nucleoside prodrugs for treating Flaviviridae infections
AU2003263412A1 (en) 2002-06-28 2004-01-19 Centre National De La Recherche Scientifique (Cnrs) 1'-, 2'- and 3'- modified nucleoside derivatives for treating flaviviridae infections
US20040003476A1 (en) * 2002-07-08 2004-01-08 Albert Bierria Portable power driven scarifying tool for pipe ends
US20060264389A1 (en) 2002-07-16 2006-11-23 Balkrishen Bhat Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
WO2004009020A2 (en) 2002-07-24 2004-01-29 Merck & Co., Inc. Pyrrolopyrimidine thionucleoside analogs as antivirals
US20040067877A1 (en) 2002-08-01 2004-04-08 Schinazi Raymond F. 2', 3'-Dideoxynucleoside analogues for the treatment or prevention of Flaviviridae infections
JP4085742B2 (en) * 2002-08-22 2008-05-14 株式会社ニコン Lens barrel and camera system provided with the lens barrel
WO2004023921A1 (en) 2002-09-16 2004-03-25 Kyeong Ho Kim Decoration band
US7094768B2 (en) 2002-09-30 2006-08-22 Genelabs Technologies, Inc. Nucleoside derivatives for treating hepatitis C virus infection
JP2006505537A (en) 2002-09-30 2006-02-16 ジェネラブス テクノロジーズ,インコーポレイテッド Nucleoside derivatives for treating hepatitis C virus infection
US20040229840A1 (en) 2002-10-29 2004-11-18 Balkrishen Bhat Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
AU2003291726A1 (en) 2002-11-04 2004-06-07 Xenoport, Inc. Gemcitabine prodrugs, pharmaceutical compositions and uses thereof
AU2003295388A1 (en) 2002-11-05 2004-06-03 Isis Pharmaceuticals, Inc. 2'-substituted oligomeric compounds and compositions for use in gene modulations
WO2004044132A2 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Modified oligonucleotides for use in rna interference
TWI332507B (en) 2002-11-19 2010-11-01 Hoffmann La Roche Antiviral nucleoside derivatives
CA2509687C (en) 2002-12-12 2012-08-14 Idenix (Cayman) Limited Process for the production of 2'-branched nucleosides
PL377608A1 (en) 2002-12-23 2006-02-06 Idenix (Cayman) Limited Process for the production of 3'-nucleoside prodrugs
WO2004065398A2 (en) 2003-01-15 2004-08-05 Ribapharm Inc. Synthesis and use of 2'-substituted-n6-modified nucleosides
AR043006A1 (en) 2003-02-12 2005-07-13 Merck & Co Inc PROCESS TO PREPARE RAMIFIED RIBONUCLEOSIDS
AU2003225705A1 (en) 2003-03-07 2004-09-30 Ribapharm Inc. Cytidine analogs and methods of use
WO2004084453A2 (en) 2003-03-20 2004-09-30 Microbiologica Quimica E Farmaceutica Ltd. METHODS OF MANUFACTURE OF 2'-DEOXY-β-L-NUCLEOSIDES
MXPA05010419A (en) 2003-03-28 2006-05-31 Pharmasset Inc Compounds for the treatment of flaviviridae infections.
US7452901B2 (en) 2003-04-25 2008-11-18 Gilead Sciences, Inc. Anti-cancer phosphonate analogs
US7595390B2 (en) 2003-04-28 2009-09-29 Novartis Ag Industrially scalable nucleoside synthesis
US20040259934A1 (en) 2003-05-01 2004-12-23 Olsen David B. Inhibiting Coronaviridae viral replication and treating Coronaviridae viral infection with nucleoside compounds
EP1656093A2 (en) 2003-05-14 2006-05-17 Idenix (Cayman) Limited Nucleosides for treatment of infection by corona viruses, toga viruses and picorna viruses
US20040229839A1 (en) 2003-05-14 2004-11-18 Biocryst Pharmaceuticals, Inc. Substituted nucleosides, preparation thereof and use as inhibitors of RNA viral polymerases
WO2005020885A2 (en) 2003-05-21 2005-03-10 Isis Pharmaceuticals, Inc. Compositions and methods for the treatment of severe acute respiratory syndrome (sars)
WO2004106356A1 (en) 2003-05-27 2004-12-09 Syddansk Universitet Functionalized nucleotide derivatives
CN100503628C (en) 2003-05-30 2009-06-24 法莫赛特股份有限公司 Modified fluorinated nucleoside analogues
ES2276311T3 (en) 2003-06-19 2007-06-16 F. Hoffmann-La Roche Ag PROCESS TO PRADERATE DERIVATIVES OF 4 '-AZIDONUCLEOSIDO.
GB0317009D0 (en) 2003-07-21 2003-08-27 Univ Cardiff Chemical compounds
WO2005021588A1 (en) 2003-08-29 2005-03-10 The Nottingham Trent University Gastric and prostate cancer associated antigens
AU2004275770A1 (en) 2003-09-22 2005-04-07 Acidophil Llc Small molecule compositions and methods for increasing drug efficiency using compositions thereof
US7144868B2 (en) * 2003-10-27 2006-12-05 Genelabs Technologies, Inc. Nucleoside compounds for treating viral infections
US20050137141A1 (en) 2003-10-24 2005-06-23 John Hilfinger Prodrug composition
US7157434B2 (en) 2003-10-27 2007-01-02 Genelabs Technologies, Inc. Nucleoside compounds for treating viral infections
CA2543090A1 (en) 2003-10-27 2005-06-16 Genelabs Technologies, Inc. Nucleoside compounds for treating viral infections
US6908824B2 (en) * 2003-11-06 2005-06-21 Chartered Semiconductor Manufacturing Ltd. Self-aligned lateral heterojunction bipolar transistor
JP2005302670A (en) 2004-04-16 2005-10-27 Matsushita Electric Ind Co Ltd Electronic device
WO2006016930A2 (en) 2004-05-14 2006-02-16 Intermune, Inc. Methods for treating hcv infection
EP1758453B1 (en) 2004-06-15 2014-07-16 Merck Sharp & Dohme Corp. C-purine nucleoside analogs as inhibitors of rna-dependent rna viral polymerase
US7560434B2 (en) 2004-06-22 2009-07-14 Biocryst Pharmaceuticals, Inc. AZA nucleosides, preparation thereof and use as inhibitors of RNA viral polymerases
RU2007102281A (en) 2004-06-23 2008-07-27 Айденикс (Кайман) Лимитед (Ky) 5-AZA-7-DEAZAPURINE DERIVATIVES FOR THE TREATMENT OF DISEASES ASSOCIATED WITH FLAVIVIRIDAE
CN1972696B (en) 2004-06-24 2010-08-11 默沙东公司 Nucleoside aryl phosphoramidates for the treatment of RNA-dependent RNA viral infection
CN101023094B (en) 2004-07-21 2011-05-18 法莫赛特股份有限公司 Preparation of alkyl-substituted 2-deoxy-2-fluoro-d-ribofuranosyl pyrimidines and purines and their derivatives
ATE543789T1 (en) 2004-07-21 2012-02-15 Pharmasset Inc PRODUCTION OF ALKYL-SUBSTITUTED 2-DESOXY-2-FLUOROD-RIBOFURANOSYLPYRIMIDINES AND PURINES AND DERIVATIVES THEREOF
DE602005015466D1 (en) 2004-08-23 2009-08-27 Hoffmann La Roche ANTIVIRAL 4'-AZIDONUCLEOSIDE
RU2007115419A (en) 2004-09-24 2008-10-27 Айденикс (Кайман) Лимитед (Ky) METHODS AND COMPOSITIONS FOR TREATMENT OF FLAVIVIRUSES, PESTIVIRUSES AND HEPACIVIRUS
CA2583351A1 (en) 2004-10-06 2006-04-13 Migenix Inc. Combination anti-viral compositions comprising castanospermine and methods of use
EP1804812A4 (en) 2004-10-21 2009-09-02 Merck & Co Inc Fluorinated pyrroloý2,3-d¨pyrimidine nucleosides for the treatment of rna-dependent rna viral infection
US7414031B2 (en) 2004-11-22 2008-08-19 Genelabs Technologies, Inc. 5-nitro-nucleoside compounds for treating viral infections
EP1828217A2 (en) 2004-12-16 2007-09-05 Febit Biotech GmbH Polymerase-independent analysis of the sequence of polynucleotides
EP1853317A2 (en) 2005-02-09 2007-11-14 Migenix Inc. Compositions and methods for treating or preventing flaviviridae infections
JP2008535932A (en) 2005-03-09 2008-09-04 イデニクス(ケイマン)リミテツド Nucleosides with unnatural bases as antiviral agents
DE102005012681A1 (en) 2005-03-18 2006-09-21 Weber, Lutz, Dr. New 1,5-dihydro-pyrrol-2-one compounds are HDM2 inhibitors, useful for treating e.g. stroke, heart infarct, ischemia, multiple sclerosis, Alzheimer's disease, degenerative disease, viral infection and cancer
US8163744B2 (en) 2005-03-18 2012-04-24 Nexuspharma, Inc. Tetrahydro-isoquinolin-1-ones for the treatment of cancer
GT200600119A (en) 2005-03-24 2006-10-25 PHARMACEUTICAL COMPOSITIONS
AR056327A1 (en) 2005-04-25 2007-10-03 Genelabs Tech Inc NUCLEOSID COMPOUNDS FOR THE TREATMENT OF VIRAL INFECTIONS
WO2006121820A1 (en) 2005-05-05 2006-11-16 Valeant Research & Development Phosphoramidate prodrugs for treatment of viral infection
EP1893198A2 (en) 2005-05-31 2008-03-05 Novartis AG Treatment of liver diseases in which iron plays a role in pathogenesis
EP2614709A1 (en) 2005-07-18 2013-07-17 Novartis AG Small animal model for HCV replication
AU2006279720A1 (en) 2005-08-12 2007-02-22 Merck & Co., Inc. Novel 2'-C-methyl and 4'-C-methyl nucleoside derivatives
AR057096A1 (en) 2005-08-26 2007-11-14 Chancellors Masters And Schola PROCESS TO PREPARE SACARINIC ACIDS AND LACTONS
WO2007065829A1 (en) 2005-12-09 2007-06-14 F. Hoffmann-La Roche Ag Antiviral nucleosides
CA2634749C (en) 2005-12-23 2014-08-19 Idenix Pharmaceuticals, Inc. Process for preparing a synthetic intermediate for preparation of branched nucleosides
AU2007215114A1 (en) 2006-02-14 2007-08-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Nucleoside aryl phosphoramidates for the treatment of RNA-dependent RNA viral infection
US7951789B2 (en) 2006-12-28 2011-05-31 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US7964580B2 (en) 2007-03-30 2011-06-21 Pharmasset, Inc. Nucleoside phosphoramidate prodrugs
EP2155771A4 (en) 2007-05-14 2012-09-05 Rfs Pharma Llc Azido purine nucleosides for treatment of viral infections
GB0718575D0 (en) 2007-09-24 2007-10-31 Angeletti P Ist Richerche Bio Nucleoside derivatives as inhibitors of viral polymerases
US20090318380A1 (en) 2007-11-20 2009-12-24 Pharmasset, Inc. 2',4'-substituted nucleosides as antiviral agents
CN104262345B (en) 2008-04-23 2017-06-23 吉利德科学公司 For the CARBA nucleoside analogs of 1 ' substitution of antiviral therapy
EP2307434B1 (en) 2008-07-02 2014-02-12 IDENIX Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
GB0815968D0 (en) 2008-09-03 2008-10-08 Angeletti P Ist Richerche Bio Antiviral agents
GB0900914D0 (en) 2009-01-20 2009-03-04 Angeletti P Ist Richerche Bio Antiviral agents
US7973013B2 (en) 2009-09-21 2011-07-05 Gilead Sciences, Inc. 2'-fluoro substituted carba-nucleoside analogs for antiviral treatment
CA2795054A1 (en) 2010-04-01 2011-10-06 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US20130273005A1 (en) 2010-12-20 2013-10-17 Gilead Sciences, Inc. Methods for treating hcv
WO2012154321A1 (en) 2011-03-31 2012-11-15 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
KR20130138840A (en) 2011-04-13 2013-12-19 머크 샤프 앤드 돔 코포레이션 2'-substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
EP2696679B1 (en) 2011-04-13 2017-08-02 Merck Sharp & Dohme Corp. 2'-cyano substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
US9156872B2 (en) 2011-04-13 2015-10-13 Merck Sharp & Dohme Corp. 2′-azido substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
KR20140033446A (en) 2011-05-19 2014-03-18 알에프에스 파마 엘엘씨 Purine monophosphate prodrugs for treatment of viral infections
AR088441A1 (en) 2011-09-12 2014-06-11 Idenix Pharmaceuticals Inc SUBSTITUTED CARBONYLOXYMETHYLPHOSPHORAMIDATE COMPOUNDS AND PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF VIRAL INFECTIONS
US8507460B2 (en) 2011-10-14 2013-08-13 Idenix Pharmaceuticals, Inc. Substituted 3′,5′-cyclic phosphates of purine nucleotide compounds and pharmaceutical compositions for the treatment of viral infections
WO2013177195A1 (en) 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. 3',5'-cyclic phosphate prodrugs for hcv infection
US9109001B2 (en) 2012-05-22 2015-08-18 Idenix Pharmaceuticals, Inc. 3′,5′-cyclic phosphoramidate prodrugs for HCV infection
WO2013177219A1 (en) 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. D-amino acid compounds for liver disease
WO2014052638A1 (en) 2012-09-27 2014-04-03 Idenix Pharmaceuticals, Inc. Esters and malonates of sate prodrugs
US10513534B2 (en) 2012-10-08 2019-12-24 Idenix Pharmaceuticals Llc 2′-chloro nucleoside analogs for HCV infection
WO2014059901A1 (en) 2012-10-17 2014-04-24 Merck Sharp & Dohme Corp. 2'-cyano substituted nucleoside derivatives and methods of use thereof for treatment of viral diseases
WO2014059902A1 (en) 2012-10-17 2014-04-24 Merck Sharp & Dohme Corp. 2'-disubstituted substituted nucleoside derivatives and methods of use thereof for treatment of viral diseases
AR092959A1 (en) 2012-10-17 2015-05-06 Merck Sharp & Dohme DERIVATIVES OF NUCLEOSIDS 2-METHYL SUBSTITUTED AND METHODS OF USE OF THE SAME FOR THE TREATMENT OF VIRAL DISEASES
EP2909223B1 (en) 2012-10-19 2017-03-22 Idenix Pharmaceuticals LLC Dinucleotide compounds for hcv infection
WO2014066239A1 (en) 2012-10-22 2014-05-01 Idenix Pharmaceuticals, Inc. 2',4'-bridged nucleosides for hcv infection
US20140140951A1 (en) 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. D-Alanine Ester of Rp-Nucleoside Analog
WO2014078436A1 (en) 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. D-alanine ester of sp-nucleoside analog
EP2935304A1 (en) 2012-12-19 2015-10-28 IDENIX Pharmaceuticals, Inc. 4'-fluoro nucleosides for the treatment of hcv
WO2014137930A1 (en) 2013-03-04 2014-09-12 Idenix Pharmaceuticals, Inc. Thiophosphate nucleosides for the treatment of hcv
EP2970357A1 (en) 2013-03-13 2016-01-20 IDENIX Pharmaceuticals, Inc. Amino acid phosphoramidate pronucleotides of 2'-cyano, azido and amino nucleosides for the treatment of hcv
US9187515B2 (en) 2013-04-01 2015-11-17 Idenix Pharmaceuticals Llc 2′,4′-fluoro nucleosides for the treatment of HCV
EP3004130B1 (en) 2013-06-05 2019-08-07 Idenix Pharmaceuticals LLC. 1',4'-thio nucleosides for the treatment of hcv
EP3010512B1 (en) 2013-06-18 2017-12-27 Merck Sharp & Dohme Corp. Cyclic phosphonate substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
EP3027636B1 (en) 2013-08-01 2022-01-05 Idenix Pharmaceuticals LLC D-amino acid phosphoramidate pronucleotides of halogeno pyrimidine compounds for liver disease

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7169766B2 (en) * 2000-05-23 2007-01-30 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus
US6914054B2 (en) * 2000-05-23 2005-07-05 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus
US10363265B2 (en) * 2000-05-23 2019-07-30 Idenix Pharmaceuticals Llc Methods and compositions for treating hepatitis C virus
US8299038B2 (en) * 2000-05-23 2012-10-30 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus
US7608597B2 (en) * 2000-05-23 2009-10-27 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus
US7157441B2 (en) * 2000-05-23 2007-01-02 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus
US7105493B2 (en) * 2000-05-26 2006-09-12 Idenix Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
US7148206B2 (en) * 2000-05-26 2006-12-12 Idenix Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
US6812219B2 (en) * 2000-05-26 2004-11-02 Idenix Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
US8343937B2 (en) * 2000-05-26 2013-01-01 Idenix Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
US7101861B2 (en) * 2000-05-26 2006-09-05 Indenix Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
US7582618B2 (en) * 2002-06-28 2009-09-01 Idenix Pharmaceuticals, Inc. 2′-C-methyl-3′-O-L-valine ester ribofuranosyl cytidine for treatment of flaviviridae infections
US7625875B2 (en) * 2002-06-28 2009-12-01 Idenix Pharmaceuticals, Inc. 2′ and 3′-nucleoside prodrugs for treating Flaviviridae infections
US7662798B2 (en) * 2002-06-28 2010-02-16 Idenix Pharmaceuticals, Inc. 2′ and 3′-nucleoside prodrugs for treating Flaviviridae infections
US7824851B2 (en) * 2002-11-15 2010-11-02 Idenix Pharmaceuticals, Inc. 2′-branched nucleosides and Flaviviridae mutation

Also Published As

Publication number Publication date
CZ20024225A3 (en) 2003-10-15
WO2001092282A3 (en) 2002-05-02
NO20025600L (en) 2003-01-17
AU2001272923A1 (en) 2001-12-11
CA2410579A1 (en) 2001-12-06
CA2410579C (en) 2010-04-20
ZA200404307B (en) 2008-04-30
US20060166865A1 (en) 2006-07-27
US6812219B2 (en) 2004-11-02
EP1294735A2 (en) 2003-03-26
JP2004510698A (en) 2004-04-08
JP5230052B2 (en) 2013-07-10
NO327249B1 (en) 2009-05-18
WO2001092282A2 (en) 2001-12-06
AU2007202602A1 (en) 2007-07-19
CN1468249A (en) 2004-01-14
EA200201262A1 (en) 2003-08-28
US20130315863A1 (en) 2013-11-28
MA26916A1 (en) 2004-12-20
TW200425898A (en) 2004-12-01
YU92202A (en) 2006-01-16
US7101861B2 (en) 2006-09-05
US20030060400A1 (en) 2003-03-27
NZ536570A (en) 2006-08-31
US20130310336A1 (en) 2013-11-21
KR20030036189A (en) 2003-05-09
EA007867B1 (en) 2007-02-27
US7105493B2 (en) 2006-09-12
AP2002002705A0 (en) 2002-12-31
US9968628B2 (en) 2018-05-15
OA12382A (en) 2006-04-17
ZA200210112B (en) 2004-06-23
US20040102414A1 (en) 2004-05-27
PE20020051A1 (en) 2002-02-16
US8343937B2 (en) 2013-01-01
EA200601591A1 (en) 2007-02-27
IL153020A0 (en) 2003-06-24
PL359169A1 (en) 2004-08-23
US7148206B2 (en) 2006-12-12
EP1736478A1 (en) 2006-12-27
AP1727A (en) 2007-03-06
EP1736478B1 (en) 2015-07-22
AR032883A1 (en) 2003-12-03
US20070037773A1 (en) 2007-02-15
US20040063622A1 (en) 2004-04-01
US7163929B2 (en) 2007-01-16
US20040097462A1 (en) 2004-05-20
CZ301182B6 (en) 2009-12-02
BR0111196A (en) 2004-04-06
NZ547204A (en) 2008-01-31
AP2006003708A0 (en) 2006-08-31
JP2013079257A (en) 2013-05-02
MXPA02011691A (en) 2004-05-17
NO20025600D0 (en) 2002-11-21
KR20080021797A (en) 2008-03-07
CN101099745A (en) 2008-01-09
CN1315862C (en) 2007-05-16

Similar Documents

Publication Publication Date Title
US20180235993A1 (en) Methods and compositions for treating flaviviruses and pestiviruses
US20040006002A1 (en) Methods and compositions for treating flaviviruses and pestiviruses using 4&#39;-modified nucleoside
US10363265B2 (en) Methods and compositions for treating hepatitis C virus
US20080280850A1 (en) Methods and Compositions for Treating Flaviviruses, Pestiviruses and Hepacivirus
WO2004096197A2 (en) 5-aza-7-deazapurine nucleosides for treating flaviviridae

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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