WO2004002999A2 - Modified 2' and 3' -nucleoside produgs for treating flaviridae infections - Google Patents

Modified 2' and 3' -nucleoside produgs for treating flaviridae infections Download PDF

Info

Publication number
WO2004002999A2
WO2004002999A2 PCT/IB2003/003246 IB0303246W WO2004002999A2 WO 2004002999 A2 WO2004002999 A2 WO 2004002999A2 IB 0303246 W IB0303246 W IB 0303246W WO 2004002999 A2 WO2004002999 A2 WO 2004002999A2
Authority
WO
WIPO (PCT)
Prior art keywords
lower alkyl
alkyl
acyl
alkenyl
alkynyl
Prior art date
Application number
PCT/IB2003/003246
Other languages
French (fr)
Other versions
WO2004002999A8 (en
WO2004002999A3 (en
Inventor
Jean-Pierre Sommadossi
Poalo La Colla
Richard Storer
Gilles Gosselin
Original Assignee
Idenix (Cayman) Limited
Centre National De La Recherche Scientifique
Universita Degli Studi Di Cagliari
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=30003988&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2004002999(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to JP2004517158A priority Critical patent/JP2005533817A/en
Priority to CA2490191A priority patent/CA2490191C/en
Priority to ES03761744.6T priority patent/ES2469569T3/en
Priority to AP2005003212A priority patent/AP2005003212A0/en
Priority to AU2003247084A priority patent/AU2003247084B9/en
Priority to YUP-1140/04A priority patent/RS114004A/en
Priority to MXPA04012709A priority patent/MXPA04012709A/en
Priority to SI200332358T priority patent/SI1523489T1/en
Priority to BRPI0312286-7A priority patent/BR0312286A/en
Priority to DK03761744.6T priority patent/DK1523489T3/en
Priority to EP03761744.6A priority patent/EP1523489B1/en
Application filed by Idenix (Cayman) Limited, Centre National De La Recherche Scientifique, Universita Degli Studi Di Cagliari filed Critical Idenix (Cayman) Limited
Publication of WO2004002999A2 publication Critical patent/WO2004002999A2/en
Publication of WO2004002999A3 publication Critical patent/WO2004002999A3/en
Priority to IL16589304A priority patent/IL165893A0/en
Priority to NO20050465A priority patent/NO330755B1/en
Publication of WO2004002999A8 publication Critical patent/WO2004002999A8/en
Priority to NO20110343A priority patent/NO20110343A1/en
Priority to NO2014019C priority patent/NO2014019I1/en
Priority to CY2014031C priority patent/CY2014031I2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • 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
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P35/00Antineoplastic agents
    • 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
    • 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
    • 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
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids

Definitions

  • This invention is in the area of pharmaceutical chemistry, and is in particular, a 2' and/or 3' prodrug of 6-modified, V, 2', 3' or 4'-branched pyrimidine nucleoside or 8- modif ⁇ ed, 1', 2', 3' or 4 '-branched purine nucleoside for the treatment of a Flaviviridae infection, such as a hepatitis C virus infection.
  • a Flaviviridae infection such as a hepatitis C virus infection.
  • the Flaviviridae family of viruses comprises at least three distinct genera: pestiviruses, which cause disease in cattle and pigs; flaviviruses, which are the primary cause of diseases such as dengue fever and yellow fever; and hepaciviruses, whose sole member is HCV.
  • 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-
  • 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.
  • 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.
  • BVDV bovine viral diarrhea virus
  • CSFV classical swine fever virus
  • BDV border disease virus
  • Pestiviruses and hepaciviruses are closely related virus groups within the Flaviviridae family.
  • Other closely related viruses in this family include the GB virus A, GB virus A-like agents, GB virus-B and GB virus-C (also called hepatitis G virus, HGV).
  • the hepacivirus group (hepatitis C virus; HCV) consists of a number of closely related but genotypically distinguishable viruses that infect humans. There are approximately 6 HCV genotypes and more than 50 subtypes.
  • bovine viral diarrhea virus Due to the similarities between pestiviruses and hepaciviruses, combined with the poor ability of hepaciviruses to grow efficiently in cell culture, bovine viral diarrhea virus (BVDV) is often used as a sunogate to study the HCV virus.
  • BVDV bovine viral diarrhea virus
  • RNA viruses possess a single large open reading frame (ORF) encoding all the viral proteins necessary for virus replication. These proteins are expressed as a polyprotein that is co- and post-translationally processed by both cellular and virus-encoded proteinases to yield the mature viral proteins.
  • the viral proteins responsible for the replication of the viral genome RNA are located within approximately the carboxy-terminal. Two-thirds of the ORF are termed nonstructural (NS) proteins.
  • NS nonstructural
  • the mature nonstructural (NS) proteins in sequential order from the amino-terminus of the nonstructural protein coding region to the carboxy-terminus of the ORF, consist of p7, NS2 5 NS3, NS4A, NS4B, NS5A, and NS5B.
  • the NS proteins of pestiviruses and hepaciviruses share sequence domains that are characteristic of specific protein functions.
  • the NS3 proteins of viruses in both groups possess amino acid sequence motifs characteristic of serine proteinases and of helicases (Gorbalenya et al. (1988) Nature 333:22; Bazan and Fletterick (1989) Virology 111 -.631-639; Gorbalenya et al. (1989) Nucleic Acid Res. 17.3889-3897).
  • the NS5B proteins of pestiviruses and hepaciviruses have the motifs characteristic of RNA- directed RNA polymerases (Koonin, EN. and Dolja, VN. (1993) Crit. Rev. Biochem.
  • RNA-directed RNA polymerases activity (Behrens et al.(1996) EMBO J. 15:12-22;
  • HCV hepatitis C virus
  • HCV Hepatitis B Virus
  • HCV is an enveloped virus containing a positive-sense single-stranded RNA genome of approximately 9.4kb.
  • the viral genome consists of a 5' untranslated region (UTR), a long open reading frame encoding a polyprotein precursor of approximately 3011 amino acids, and a short 3' UTR.
  • the 5' UTR is the most highly conserved part of the HCV genome and is important for the initiation and control of polyprotein translation.
  • Translation of the HCV genome is initiated by a cap-independent mechanism known as internal ribosome entry. This mechanism involves the binding of ribosomes to an RNA sequence known as the internal ribosome entry site (IRES).
  • IRES internal ribosome entry site
  • Viral structural proteins include a nucleocapsid core protein (C) and two envelope glycoproteins, El and E2.
  • HCV also encodes two proteinases, a zinc-dependent metalloproteinase encoded by the NS2-NS3 region and a serine proteinase encoded in the NS3 region. These proteinases are required for cleavage of specific regions of the precursor polyprotein into mature peptides.
  • the carboxyl half of nonstructural protein 5, NS5B contains the RNA- dependent RNA polymerase.
  • the function of the remaining nonstructural proteins, NS4A and NS4B, and that of NS5A remain unknown.
  • IFNs Interferons
  • Flaviviridae including HCV, treatments, using interferon-based therapies.
  • U.S. Patent No. 5,980,884 to Blatt et al. discloses methods for retreatment of patients afflicted with HCV using consensus interferon.
  • U.S. Patent No. 5,980,884 to Blatt et al. discloses methods for retreatment of patients afflicted with HCV using consensus interferon.
  • U.S. Patent No. 5,980,884 to Blatt et al. discloses methods for retreatment of patients afflicted with HCV using consensus interferon.
  • Patent No. 5,942,223 to Bazer et al. discloses an anti-HCV therapy using ovine or bovine interferon-tau.
  • U.S. Patent No. 5,928,636 to Alber et al. discloses the combination therapy of interleukin- 12 and interferon alpha for the treatment of infectious diseases including HCV.
  • U.S. Patent No. 5,849,696 to Chretien et al. discloses the use of thymosins, alone or in combination with interferon, for treating HCV.
  • U.S. Patent No. 5,830,455 to Valtuena et al. discloses a combination HCV therapy employing interferon and a free radical scavenger.
  • 5,738,845 to Imakawa discloses the use of human interferon tau proteins for treating HCV.
  • Other interferon-based treatments for HCV are disclosed in U.S. Patent No. 5,676,942 to Testa et al, U.S. Patent No. 5,372,808 to Blatt et al, and U.S. Patent No. 5,849,696.
  • a number of patents also disclose pegylated forms of interferon, such as, U.S.
  • Interferon alpha-2a and interferon alpha-2b are cunently approved as monotherapy for the treatment of HCV.
  • ROFERON®-A (Roche) is the recombinant form of interferon alpha-2a.
  • PEGASYS® (Roche) is the pegylated (i.e. polyethylene glycol modified) form of interferon alpha-2a.
  • INTRON®A (Schering Corporation) is the recombinant form of Interferon alpha-2b
  • PEG-INTRON® Schering Corporation
  • Other forms of interferon alpha, as well as interferon beta, gamma, tau and omega are currently in clinical development for the treatment of HCV.
  • INFERGEN interferon alphacon-1 by InterMune
  • OMNIFERON natural interferon
  • ALBUFERON Human Genome Sciences
  • REBIF interferon beta- la
  • Ares-Serono Omega Interferon by BioMedicine
  • Oral Interferon Alpha by Amarillo Biosciences
  • interferon gamma interferon tau
  • interferon gamma- lb by InterMune
  • Ribavirin (l- ⁇ -D-ribofuranosyl-l-l,2,4-triazole-3-carboxamide) is a synthetic, non- interferon-inducing, broad spectrum antiviral nucleoside analog sold under the trade name, Virazole (The Merck Index, 11th edition, Editor: Budavari, S., Merck & Co., Inc., Rahway, NJ, pl304, 1989). United States Patent No. 3,798,209 and RE29,835 disclose and claim ribavirin. Ribavirin is structurally similar to guanosine, and has in vitro activity against several DNA and RNA viruses including Flaviviridae (Gary L. Davis. Gastroenterology 118:S104-S114, 2000).
  • Ribavirin reduces serum amino transferase levels to normal in 40% of patients, but it does not lower serum levels of HCV-RNA (Gary L. Davis. Gastroenterology 118:S104-
  • ribavirin alone is not effective in reducing viral RNA levels. Additionally, ribavirin has significant toxicity and is known to induce anemia.
  • Ribavirin is not approved fro monotherapy against HCV. It has been approved in combination with interferon alpha-2a or interferon alpha-2b for the treatment of HCV.
  • the current standard of care for chronic hepatitis C is combination therapy with an alpha interferon and ribavirin.
  • the combination of interferon and ribavirin for the treatment of HCV infection has been reported to be effective in the treatment of interferon naive patients (Battaglia, A.M. et al, Ann. Pharmacother. 34:487-494, 2000), as well as for treatment of patients when histological disease is present (Berenguer, M. et al. Antivir. Ther. 3(Suppl. 3): 125-136, 1998).
  • PEGASYS® pegylated interferon alpha-2a
  • COPEGUS® ribavirin
  • PCT Publication Nos. WO 99/59621, WO 00/37110, WO 01/81359, WO 02/32414 and WO 03/024461 by Schering Corporation disclose the use of pegylated interferon alpha and ribavirin combination therapy for the treatment of HCV.
  • PCT Publication Nos. WO 99/15194, WO 99/64016, and WO 00/24355 by Hoffmann-La Roche Inc also disclose the use of pegylated interferon alpha and ribavirin combination therapy for the treatment of HCV.
  • HCV-derived enzymes such as protease, helicase, and polymerase inhibitors
  • Drugs that inhibit other steps in HCV replication are also in development, for example, drugs that block production of HCV antigens from the RNA (IRES inhibitors), drugs that prevent the normal processing of HCV proteins (inhibitors of glycosylation), drugs that block entry of HCV into cells (by blocking its receptor) and nonspecific cytoprotective agents that block cell injury caused by the virus infection.
  • ribozymes which are enzymes that break down specific viral RNA molecules
  • antisense oligonucleotides which are small complementary segments of DNA that bind to viral RNA and inhibit viral replication
  • 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) are being investigated.
  • Non-substrate-based NS3 protease 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 /r ⁇ r ⁇ -phenoxyphenyl group are also being investigated.
  • Sch 68631 a phenanthrenequinone
  • HCV protease inhibitor Chom M. et al, Tetrahedron Letters 37:7229-7232, 1996.
  • Sch 351633 isolated from the fungus Penicillium griseofulvum, was identified as a protease inhibitor (Chu M. et al, Bioorganic and Medicinal Chemistry Letters 9:1949-1952).
  • Nanomolar potency against the HCV NS3 protease enzyme has been achieved by the design of selective inhibitors based on the macromolecule eglin c.
  • Eglin c isolated from leech, is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, ⁇ - chymotrypsin, chymase and subtilisin. Qasim M.A. et al, Biochemistry 36:1598-1607,
  • U.S. patents disclose protease inhibitors for the treatment of HCV.
  • U.S. Patent No. 6,004,933 to Spruce et al. discloses a class of cysteine protease inhibitors for inhibiting HCV endopeptidase 2.
  • U.S. Patent No. 5,990,276 to Zhang et al. discloses synthetic inhibitors of hepatitis C virus NS3 protease. The inhibitor is a subsequence of a substrate of the NS3 protease or a substrate of the NS4A cofactor.
  • restriction enzymes to treat HCV is disclosed in U.S. Patent No. 5,538,865 to Reyes et al.
  • HCV inhibitor tripeptides are disclosed in US Patent Nos. 6,534,523, 6,410,531, and
  • Diaryl peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/48172 to Schering Corporation.
  • Imidazoleidinones as NS3 serine protease inhibitors of HCV are disclosed in WO 02/08198 to Schering Corporation and WO 02/48157 to Bristol Myers Squibb.
  • Squibb also disclose HCV protease inhibitors.
  • Helicase inhibitors for example 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, Piper idine 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,
  • nucleotides 326-348 comprising the 3' end of the NCR and nucleotides 371-388 located in the core coding region of the HCV RNA
  • Alt M. et al Archives of Virology, 1997, 142, 589-599; Galderisi U. et al, Journal of Cellular Physiology, 1999, 181, 251-257
  • 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);
  • Ribozymes such as nuclease-resistant ribozymes (Maccjak, D. J. et al, Hepatology 1999, 30, abstract 995) and those disclosed in U.S. Patent No. 6,043,077 to Barber et al, and U.S. Patent Nos. 5,869,253 and 5,610,054 to
  • Idenix Pharmaceuticals discloses the use of branched nucleosides in the treatment of flaviviruses (including HCV) and pestiviruses in International Publication Nos. WO
  • hepatitis C infection and flaviviruses and pestiviruses
  • a method for the treatment of hepatitis C infection (and flaviviruses and pestiviruses) in humans and other host animals is disclosed in the Idenix publications that includes administering an effective amount of a biologically active 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleosides or a pharmaceutically acceptable salt or derivative thereof, administered either alone or in combination with another antiviral agent, optionally in a pharmaceutically acceptable canier.
  • Patent 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
  • RNA replication inhibitors (VP50406) by ViroPharma/Wyeth, therapeutic vaccine by Intercell, therapeutic vaccine by Epimmune/Genencor, IRES inhibitor by Anadys, ANA 245 and ANA 246 by Anadys, immunotherapy (Therapore) by Avant, protease inhibitor by Corvas/SChering, helicase inhibitor by Vertex, fusion inhibitor by Trimeris, T cell therapy by CellExSys, polymerase inhibitor by Biocryst, targeted RNA chemistry by PTC Therapeutics, Dication by Immtech, Int., protease inhibitor by Agouron, protease inhibitor by Chiron/Medivir, anti
  • Nucleoside prodrugs have been previously described for the treatment of other forms of hepatitis.
  • WO 00/09531 (filed August 10, 1999) and WO 01/96353 (filed June 15, 2001) to Idenix Pharmaceuticals discloses 2'-deoxy- ⁇ -L-nucleosides and their 3 '-prodrugs for the treatment of HBV.
  • U.S. Patent No. 4,957,924 to Beauchamp discloses various therapeutic esters of acyclovir. In light of the fact that HCV infection has reached epidemic levels worldwide, and has tragic effects on the infected patient, there remains a strong need to provide new effective pharmaceutical agents to treat hepatitis C that have low toxicity to the host.
  • 2' and 3'-prodrugs of 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleosides, or their pharmaceutically acceptable salts or pharmaceutically acceptable formulations containing these compounds are useful in the prevention and treatment of Flaviviridae infections and other related conditions such as anti- Flaviviridae antibody positive and Flaviviridae - positive conditions, chronic liver inflammation caused by HCV, cinhosis, acute hepatitis, fulminant hepatitis, chronic persistent hepatitis, and fatigue.
  • These compounds or formulations can also be used prophylactically to prevent or retard the progression of clinical illness in individuals who are anti-Flaviviridae antibody or Flaviviridae-antigen positive or who have been exposed to a Flaviviridae.
  • a method for the treatment of a Flaviviridae viral infection in a host, including a human includes administering an effective amount of a 2' or 3'- prodrug of a biologically active 1', , 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside or a pharmaceutically acceptable salt thereof, administered either alone or in combination or alternation with another anti-Flaviviridae agent, optionally in a pharmaceutically acceptable canier.
  • 2 '-prodrug refers to a 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside that has a biologically cleavable moiety at the 2 '-position, including, but not limited to acyl, and in one embodiment, a natural or synthetic D or L amino acid, preferably an L-amino acid.
  • 3 '-prodrug refers to a 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside that has a biologically cleavable moiety at the 3 '-position, including, but not limited to acyl, and in one embodiment, a natural or synthetic D or L amino acid, preferably an L-amino acid.
  • Pharmaceutically acceptable salts include tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, ⁇ -ketoglutarate, and ⁇ - glycerophosphate, formate, fumarate, propionate, glycolate, lactate, pyruvate, oxalate, maleate, salicyate, sulfate, sulfonate, nitrate, bicarbonate, hydrobromate, hydrobromide, hydroiodide, carbonate, and phosphoric acid salts.
  • a particularly preferred embodiment is the mono or dihydrochloride salt.
  • the 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside includes biologically cleavable moieties at the 2' and/or 5' positions.
  • Prefened moieties are natural or synthetic D or L amino acid esters, including D or L-valyl, though preferably L-amino acid esters, such as L-valyl, and alkyl esters including acetyl.
  • this invention specifically includes 2'-D or L-amino acid ester and 2',5'-D or L-diamino acid ester, preferably L-amino acid ester, of V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC 50 of less than 15 micromolar, and even more preferably less than 10 micromolar; 2 '-(alkyl or aryl) ester or 2',5'-di(alkyl or aryl) ester of 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC 50 of less than 10 or 15 micromolar; and prodrugs of 2',5'-diesters of 1', 2', 3' or 4'-branched ⁇ - D or ⁇
  • prodrugs falling within the invention are 2',5'-L-divaline ester of ⁇ -D-2',6-dimethyl-cytidine (dival-2',6-diMe-L-dC); 2',5'-L-divaline ester of ⁇ -D- 2',6-dimethyl-thymidine; 2',5'-L-divaline ester of ⁇ -D-2',8-dirnethyl-adenosine; 2',5'-L- divaline ester of ⁇ -D-2',8-dimethyl-guanosine; 2',5'-L-divaline ester of ⁇ -D-2',6-dimethyl- 5-fluoro-cytidine; 2',5'-L-divaline ester of ⁇ -D-2',6-dimethyl-uridine; 2',5'-diacetyl ester of ⁇ -D-2',6-dimethyl
  • the 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside 3'- prodrug includes biologically cleavable moieties at the 3' and/or 5' positions.
  • Preferred moieties are natural or synthetic D or L amino acid esters, such as valyl, though preferably L-amino acids, such as L-valyl, and alkyl esters including acetyl.
  • this invention specifically includes 3 '-L-amino acid ester and 3',5'-L-diamino acid ester of 1', 2', 3' or 4'- branched ⁇ -D or ⁇ -L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC 50 of less than 15 micromolar, and even more preferably less than 10 micromolar; 3'-(alkyl or aryl) ester or 3',5'-L-di(alkyl or aryl) ester of 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC 5 o of less than 10 or 15 micromolar; and prodrugs of
  • 3 ',5 '-diesters of V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleosides wherein (i) the 3' ester is a natural or synthetic D or L amino acid ester and the 5 '-ester is an alkyl or aryl ester; (ii) both esters are natural or synthetic D or L-amino acid esters; (iii) both esters are independently alkyl or aryl esters; and (iv) the 3' ester is independently an alkyl or aryl ester and the 5 '-ester is a natural or synthetic D or L-amino acid ester, wherein the parent drug optionally has an EC50 of less than 10 or 15 micromolar.
  • prodrugs falling within the invention are 3'-L-valine ester of ⁇ -D-2',6- dimethyl-cytidine; 3'-L-valine ester of ⁇ -D-2',6-dimethyl-thymidine; 3 '-L- valine ester of ⁇ - D-2',8-dimethyl-adenosine; 3'-L-valine ester of ⁇ -D-2',8-dimethyl-guanosine; 3'-L-valine ester of ⁇ -D-2',6-dimethyl-5-fluorocytidine; 3'-L-valine ester of ⁇ -D-2',6-dimethyl-uridine; 3'-acetyl ester of ⁇ -D-2',6-dimethyl-cytidine; 3'-acetyl ester of ⁇ -D-2',6-dimethyl- thymidine; 3'-acetyl ester of ⁇ -D-2',8
  • prodrugs falling within the invention are 3',5'-L-divaline ester of ⁇ -D-2',6-dimethyl-cytidine (dival-2',6-diMe-L-dC); 3',5'-L-divaline ester of ⁇ -D-
  • the prodrug of V, 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside includes biologically cleavable moieties at the 2', 3' and/or 5' positions.
  • Prefened moieties are natural or synthetic D or L amino acid esters, including D or L-valyl, though preferably L-amino acid esters, such as L-valyl, and alkyl esters including acetyl.
  • this invention specifically includes 2',3'-L or D-diamino acid ester and 2',3',5'- L or D-triamino acid ester of 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleosides, prefenably L-amino acid, with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC50 of less than 15 micromolar, and even more preferably less than 10 micromolar; 2',3'-di(alkyl or aryl) ester or 2',3',5'-L-tri(alkyl or aryl) ester of 1', 2', 3' or
  • 2',3',5'- triesters of 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleosides wherein (i) all three esters are amino acid esters; (ii) all three esters are independently alkyl or aryl esters; (iii) the 2' ester is an amino acid ester, the 3' ester is an amino acid ester and the 5 '-ester is an alkyl or aryl ester; (iv) the 2' ester is an amino acid ester, the 3' ester is an alkyl or aryl ester and the 5'- ester is an alkyl or aryl ester; (v) the 2' ester is an alkyl or aryl ester, the 3' ester is an alkyl or aryl ester and the 5 '-ester is an amino acid ester; (vi) the 2' ester is an alkyl or aryl ester, the 3' ester is an
  • prodrugs falling within the invention include 2',3'-L-divaline ester of ⁇ -D-2',6-dimethyl-cytidine (dival-2',6-diMe-L-dC); 2',3'-L-divaline ester of ⁇ -D-2',6- dimethyl-thymidine; 2',3'-L-divaline ester of ⁇ -D-2',8-dimethyl-adenosine; 2',3'-L-divaline ester of ⁇ -D-2',8-dimethyl-guanosine; 2',3'-L-divaline ester of ⁇ -D-2',6-dimethyl-5-fluoro- cytidine; 2',3'-L-divaline ester of ⁇ -D-2',6-dimethyl-uridine; 2',3 '-diacetyl ester of ⁇ -D-D-
  • prodrugs falling within the invention include 2',3',5'-L- trivaline ester of ⁇ -D-2',6-dimethyl-cytidine (trival-2',6-diMe-L-dC); 2',3',5'-L-trivaline ester of ⁇ -D-2',6-dimethyl-thymidine; 2',3',5'-L-trivaline ester of ⁇ -D-2',8-dimethyl- adenosine; 2',3',5'-L-trivaline ester of ⁇ -D-2',8-dimethyl-guanosine; 2',3',5'-L-trivaline ester of ⁇ -D-2',6-dimethyl-5-fluoro-cytidine; 2',3',5'-L-trivaline ester of ⁇ -D-2',6- dimethyl-uridine; 2',3',5'-triacet
  • a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (I):
  • R 1 , R 2 and R 3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate); straight chained, branched or cyclic alkyl (including lower alkyl); acyl (including lower acyl); CO-alkyl, CO-aryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO- substituted aryl, 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; alkylsulfonyl, arylsulfonyl, aralkylsulfonyl, a lipid, including a phospholipid; an amino acid; and amino acid residue, a carbohydrate; a peptide; cholesterol; or other pharmaceutically acceptable leaving group
  • R 1 , R 2 and/or R 3 may be a pharmaceutically acceptable leaving group which is capable of providing a compound wherein R 1 , R 2 and/or
  • R 3 is independently H or phosphate (including mono-, di- or triphosphate), for example when administered in vivo.
  • a compound of Formula (II) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (II):
  • a compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric, or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV) or (V):
  • Base is selected from the group consisting of
  • R 1 , R 2 R 3 ' R 4 , and R 5 are as defined above; each W 1 , W 2 , W 3 and W 4 is independently N, CH, CF, CI, CBr, CCl, CCN, CCH 3 , CCF 3 ,
  • each W* is independently O, S, NH or NR 4 ;
  • X is O, S, SO 2 , CH 2 , CH 2 OH, CHF, CF 2 , C(Y 3 ) 2 , CHCN, C(CN) 2 , CHR 4 or C(R 4 ) 2 ;
  • X* is CH, CF, CY 3 or CR 4 ;
  • X 2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH 3 , CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , CH 2 OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR 4 , COO-alkyl, COO-aryl, CO- Oalkoxyalkyl, CONH 2 , CONHR 4 , CON(R 4 ) 2 , chloro, bromo, fluoro, iodo, CN, N 3 , OH,
  • each X 3 is independently a straight chained, branched or cyclic optionally substituted alkyl (including lower alkyl), CH 3 , CH 2 CN, CH 2 N 3 , CH 2 NH 2 , CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 OH, halogenated alkyl (including halogenated lower alkyl), CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , optionally substituted alkenyl, haloalkenyl,
  • Y 1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR 4 , NH 2 , NHR 4 , NR 4 R 5 , SH or SR 4 ; each Y 2 is independently O, S, NH or NR 4 ; each Y 3 is independently H, F, Cl, Br or I; wherein for Base (B), W 4 cannot be CH if W 1 , W 2 and W 3 are N; wherein for Base (E), (F), (K), (L), (W) and (X), W 4 cannot be CH if W 1 is N; each R 6 is independently an optionally substituted alkyl (including lower alkyl), CH 3 ,
  • CH 2 CN CH 2 N 3 , CH 2 NH 2 , CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 OH, halogenated alkyl (including halogenated lower alkyl), CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 ,
  • a compound of Formula (VI) or (VII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or (VII):
  • R 8 in Formula (VI) is -OH or -NH 2 only when X is carbon; and wherein; each R 8 and R 11 is independently hydrogen, an optionally substituted alkyl (including lower alkyl), CH 3 , CH 2 CN, CH 2 N 3 , CH 2 NH 2 , CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 OH, halogenated alkyl (including halogenated lower alkyl), CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH 2 C(O)OH, -CH 2 C(O)OR 4
  • a compound of Formula (VIII), (IX) or (X), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VIII), (DQ or (X):
  • each R 12 is independently a substituted alkyl (including lower alkyl), CH 2 CN, CH 2 N 3 , CH 2 NH 2 , CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 OH, halogenated alkyl (including halogenated lower alkyl), CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , substituted alkenyl, haloalkenyl
  • a compound of Formula (XI) or (XII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XI) or (XII):
  • Base Base*, R, R 1 , R 2 , R 3 , R 4 , R 5 , R 12 , R 13 , Y, Y 1 , Y 2 , Y 3 , W * , W 1 , W 2 , W 3 , W 4 , X, X*, X 1 ,
  • X 2 , and X 3 are as defined above; wherein, in one embodiment, R 8 in Formula (XI) is -OH or -NH 2 only when X is carbon; and wherein; 2004/002999
  • each R 8 and R 11 is independently hydrogen, an optionally substituted alkyl (including lower alkyl), CH 3 , CH 2 CN, CH 2 N 3 , CH 2 NH 2 , CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 OH, halogenated alkyl (including halogenated lower alkyl), CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH 2 C(0)OH, -CH 2 C(O)OR 4 , -CH 2 C(0)0(lower alkyl),
  • R 8 and R 13 , R 9 and R 13 , R 9 and R 11 or R 10 and R 12 can come together to form a bridged compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); or alternatively, R 12 and R 13 or R 9 and R 10 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably
  • a compound of Formula (XI) or (XII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XI) or (XII):
  • R 3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; acyl; a sulfonate ester; optionally substituted alkyl sulfonyl; optionally substituted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 3 is independently H, or mono-, di- or triphosphate; X" is selected from the group consisting of one or more O, S, SO, S0 2 , N, NH, NR and CH 2 wherein any of the aforementioned may be optionally substituted and may be variably positioned so as to form a 3-7 membered ring;
  • R is H, alkyl or acyl
  • B indicates a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or
  • Base is selected from the group consisting of:
  • each R', R", R'" and R" are independently selected from the group consisting of
  • H OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH 2 , NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* C0 2 -alkyl, CONH-alkyl, CON-dialkyl, OH, CF 3 , CH 2 OH, (CH 2 ) m OH, (CH 2
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Q and Q 2 independently are N or C-R;
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (XV), (XVI) or (XVII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XV), (XVI) or (XVII):
  • R is H, alkyl or acyl; and R', R", R'", R"", and R 3 and Base are as defined for Formula (XIII).
  • At most one of G and E can further be hydrogen.
  • a compound of Formula (XVIII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a
  • a compound of Formula (XIX), (XX), (XXI) (XXII) or (XXIII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XIX), (XX), (XXI) (XXII) or (XXIII):
  • A is selected from the group consisting of optionally substituted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF 2 CF 3 , CH 2 CO 2 R, (CH 2 ) m COOH, (CH 2 ) m COOR, (CH 2 ) m CO- NH* (CH 2 ) m CONR* and (CH 2 ) m CONHR;
  • Y is selected from the group consisting of H, optionally substituted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF 2 CF 3 , CH 2 CO 2 R, (CH 2 ) m COOH, (CH 2 ) m COOR, (CH 2 ) m CONH* (CH 2 ) m CONR 2 , and (CH 2 ) m CONHR;
  • X is selected from the group consisting of -OH, optionally substituted alkyl, cycloalkyl, alkenyl, alkynyl, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aryl, -O-aralkyl, -O-cycloal
  • R is H, alkyl or acyl
  • R 3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; substituted or unsubstituted alkyl; acyl; a sulfonate ester; optionally substituted alkyl sulfonyl; optionally substituted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 3 is independently H, or mono-, di- or triphosphate; and Base is a non-natural base selected from the group of:
  • each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH 2 , NH-alkyl, N-dialkyl,
  • W is C-R" orN; T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
  • Qi and Q 2 independently are N or C-R"";
  • Q 3 , Q 4 , Q 5 and Q 6 independently are N or CH; with the proviso that in bases (g) and (i), R', R"" are not H, OH, or NH 2 ; and Q,
  • T, V, Q* Q5 and Q 6 are not N.
  • the amino acid residue is of the formula C(0)C(R ⁇ )(R 12 )(NR 13 R 14 ), wherein:
  • R 11 is the side chain of an amino acid and wherein, as in proline, R 11 can optionally be attached to R 13 to form a ring structure; or alternatively, R n is an alkyl, aryl, heteroaryl or heterocyclic moiety;
  • R 12 is hydrogen, alkyl (including lower alkyl) or aryl
  • R 13 and R 14 are independently hydrogen, acyl (including an acyl derivative attached to R 11 ) or alkyl (including but not limited to methyl, ethyl, propyl, and cyclopropyl).
  • at least one of R 2 and R 3 is an amino acid residue, and is preferably L-valinyl.
  • the ⁇ -D- and ⁇ -L-nucleosides of this invention may inhibit Flaviviridae polymerase activity.
  • Nucleosides can be screened for their ability to inhibit Flaviviridae 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-Flaviviridae 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 ECso).
  • the parent of the prodrug compound exhibits an EC 50 of less than 25, 15, 10, 5, or 1 micromolar.
  • the compound exhibits an EC50 of less than 15 or 10 micromolar, when measured according to the polymerase assay described in Fenari et al, Jnl of Vir., 73:1649-1654, 1999; Ishii et al, Hepatology, 29:1227-1235,1999; Lohmann et al, Jnl. of Bio. Chem., 274:10807-10815, 1999; or Yamashita et al, Jnl. of Bio. Chem., 273:15479-15486, 1998.
  • combination and/or alternation therapy are provided.
  • an effective dosage 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.
  • the invention provides combinations of at least two of the herein described prodrugs.
  • the invention further provides at least one of the described 2' and 3'-prodrugs in combination or alternation with a second nucleoside that exhibits activity against a Flaviviridae, including but not limited to a parent drug of any of the prodrugs defined herein, i.e.
  • the 2' or 3'-prodrugs can be administered in combination or alternation with other anti-Flaviviridae agent exhibits an EC 5 0 of less than 10 or 15 micromolar, or their prodrugs or pharmaceutically acceptable salts.
  • Nonlimiting examples of antiviral agents that can be used in combination with the compounds disclosed herein include: 1) an interferon and/or ribavirin; (2) Substrate-based NS3 protease inhibitors;. (3) Non-substrate-based inhibitors; (4) Thiazolidine derivatives; (5) Thiazolidines and benzanilides; (6) A phenan-threnequinone; (7) NS3 inhibitors; (8) HCV helicase inhibitors; (9) polymerase inhibitors, including RNA-dependent RNA- polymerase inhibitors; (10) Antisense oligodeoxynucleotides; (11) Inhibitors of IRES- dependent translation; (12) Nuclease-resistant ribozymes; and (13) other compounds that exhibit activity against a flaviviridae.
  • the invention further includes administering the prodrug in combination or alternation with an immune modulator or other pharmaceutically active modifer of viral replication, including a biological material such as a protein, peptide, oligonucleotide, or gamma globulin, including but not limited to interfereon, interleukin, or an antisense oligonucleotides to genes which express or regulate Flaviviridae replication.
  • an immune modulator or other pharmaceutically active modifer of viral replication including a biological material such as a protein, peptide, oligonucleotide, or gamma globulin, including but not limited to interfereon, interleukin, or an antisense oligonucleotides to genes which express or regulate Flaviviridae replication.
  • the invention further includes administering the prodrug in combination or alternation with an immune modulator or other pharmaceutically active modifer of viral replication, including a biological material such as a protein, peptide, oligonucleotide, or gamma globulin, including but not limited to interfereon, interleukin, or an antisense oligonucleotides to genes which express or regulate Flaviviridae replication.
  • an immune modulator or other pharmaceutically active modifer of viral replication including a biological material such as a protein, peptide, oligonucleotide, or gamma globulin, including but not limited to interfereon, interleukin, or an antisense oligonucleotides to genes which express or regulate Flaviviridae replication.
  • an immune modulator or other pharmaceutically active modifer of viral replication including a biological material such as a protein, peptide, oligonucleotide, or gamma globulin, including but not limited to interfere
  • compositions comprising a compound of Formula (XIII) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable canier or dileuent;
  • compositions comprising a compound of Formula (XIII) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, with one or more other effective antiviral agent, optionally with a pharmaceutically acceptable canier or dileuent;
  • pharmaceutical compositions for the treatment of a Flaviviridae infection in a host comprising a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable canier or dileuent;
  • compositions for the treatment of a Flaviviridae infection in a host comprising a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, with one or more other effective antiviral agent, optionally with a pharmaceutically acceptable carrier or dileuent;
  • (j) uses for a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, optionally with a pharmaceutically acceptable canier or dileuent, in the manufacture of a medicament for the treatment of a Flaviviridae infection in a host; and (k) uses for a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, with one or more other effective antiviral agent, optionally with a pharmaceutically acceptable canier or dileuent, in the manufacture of a medicament for the treatment of a Flaviviridae infection in a host.
  • the parent nucleoside compound of any of the 2'- or 3'- prodrugs are provided for the treatment of a Flaviviridae infection and in particular a hepatitis C infection.
  • Figure 1 provides the structure of various non-limiting examples of nucleosides of the present invention, as well as other known nucleosides, in particular FIAU and ribavirin.
  • Figure 2 provides a non-limiting example of the steps involved in esterification of the V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside to obtain a 2'-prodrug.
  • the same general procedure can be used to obtain the 3 '-prodrug by selectively protecting the 2' and 5 '-hydroxyl groups or protecting the 2', 3' and 5 '-hydroxyl groups and selectively deprotecting the 3 ' -hydroxyl.
  • Figure 3 provides a non-limiting example of the steps involved in esterification of the 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside to obtain a 3 '-prodrug.
  • Figure 4 provides a non-limiting example of the steps involved in esterification of the 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside to obtain a 2',3 '-prodrug.
  • the invention as disclosed herein is a compound, a method and composition for the treatment of a Flaviviridae infection in humans and other host animals.
  • the method includes the administration of an effective anti-Flaviviridae treatment amount of a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside as described herein or a pharmaceutically acceptable salt, derivative or prodrug thereof, optionally in a pharmaceutically acceptable canier.
  • the compounds of this invention either possesses antiviral (i.e., anti-HCV) activity, or are metabolized to a compound that exhibits such activity.
  • HCV is a member of the Flaviviridae family.
  • the Flaviviridae has been placed in a new monotypic genus, hepacivirus. Therefore, in one embodiment, the Flaviviridae is HCV. In an alternate embodiment, the Flaviviridae is a flavivirus or pestivirus.
  • the 2' and/or 3 '-prodrugs of a 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside are acyl derivates of a secondary or tertiary alcohol alpha to a secondary or tertiary carbon. Due to the steric hindrance of these prodrugs over the 5 '-prodrugs, an acyl derivative of a primary alcohol, these prodrugs differentially modulate the biological properties of the molecule in vivo.
  • the 2' and/or 3'-prodrugs of a V, , 3' or 4'- branched ⁇ -D or ⁇ -L nucleoside can provide a drug with increased half-life and improved pharmacokinetic profile.
  • the 2' or 3 '-prodrug in a preferred embodiment is a cleavable acyl group, and most particularly, an amino acid moiety, prepared from any naturally occurring and synthetic ⁇ , ⁇ ⁇ or ⁇ amino acid, including but is not limited to, glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine.
  • the amino acid is in the L-configuration.
  • the amino acid can be a derivative of alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, ⁇ -alanyl, ⁇ -valinyl 5 ⁇ - leucinyl, ⁇ -isoleuccinyl, ⁇ -prolinyl, ⁇ -phenylalaninyl, ⁇ -tryptophanyl, ⁇ -methioninyl, ⁇ - glycinyl, ⁇ -serinyl, ⁇ -threoninyl, ⁇ -cysteiny
  • the oral bio-availability of 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside as the neutral base and the HCl salt is low in rodents and non-human primates. It has been discovered that there is significant competition of V, , 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside with other nucleosides or nucleoside analogs for absorption, or transport, from the gastrointestinal tract and competition of other nucleosides or nucleoside analogs for the absorption with 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside.
  • 2' and 3 '-prodrugs of 1', 2 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside were obtained with higher oral bioavailability than the parent molecule and a reduced effect on the bioavailability of other nucleosides or nucleoside analogs used in combination.
  • the 2', 3', and/or 5'-mono, di or frivaline ester of a 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside have higher oral bioavailability than the parent 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside and reduced interaction with other nucleosides or nucleoside analogs when used in combination as compared to V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside.
  • the 2', 3', and/or 5'-mono, di or trivaline ester of a 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside can be converted to the parent V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside through de-esterification in the gastrointestinal mucosa, blood or liver.
  • the 2', 3', and/or 5'-mono, di or trivaline ester of a 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside can be converted to the parent V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside through de-esterification in the gastrointestinal mucosa, blood or liver.
  • 3', and/or 5 '-mono, di or trivaline ester of a 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside can be actively transported from the gastrointestinal lumen after oral delivery into the bloodstream by an amino acid transporter function in the mucosa of the gastrointestinal tract. This accounts for the increase in oral bioavailability compared to the parent 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside that is transported primarily by a nucleoside transporter function.
  • the present invention includes the following features:
  • compositions comprising a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4'- branched ⁇ -D or ⁇ -L nucleoside or a pharmaceutically acceptable salt thereof together with one or more other effective anti-HCV agents, optionally in a pharmaceutically acceptable canier or diluent;
  • compositions comprising a 2' and/or 3'-prodrug of a 1', 2', 3' or 4'- branched ⁇ -D or ⁇ -L nucleoside or a pharmaceutically acceptable salt thereof together with the parent of a different a V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside, optionally in a pharmaceutically acceptable carrier or diluent;
  • a method for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside, its pharmaceutically acceptable salt or composition in combination and/or alternation with one or more effective anti- HCV agent;
  • (k) a method for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of a 2' and/or 3 '-prodrug of a ⁇ -D-2'-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof; (1) a method for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of the 2'-valyl or acetyl ester of ⁇ -D-2'-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof; (m) use of a 2' and/or 3'-prodrug of a 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside, and pharmaceutically acceptable salts and compositions thereof for the treatment and/or prophylaxis of a Flaviviridae infection in a host; (n) use of a 2' and
  • Flaviviridae infection in a host (o) use of a 2' and/or 3'-prodrug of a V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside, or its pharmaceutically acceptable salt or composition with the parent of a different a 1 ', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside for the treatment and/or prophylaxis of a Flaviviridae infection in a host; (p) use of a 2' and/or 3 '-prodrug of a ⁇ -D-2 '-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof for the treatment and/or prophylaxis of a
  • Flaviviridae infection in a host (q) use of the 3'-valyl or acetyl ester of ⁇ -D-2 '-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof for the treatment and/or prophylaxis of a
  • Flaviviridae infection in a host (r) use of a 2' and/or 3'-prodrug of a V, 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleoside, and pharmaceutically acceptable salts and compositions thereof in the manufacture of a medicament for treatment and/or prophylaxis of a Flaviviridae infection;
  • Flaviviridae 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.
  • the Flaviviridae is HCV.
  • the Flaviviridae is a flavivirus or pestivirus. Specific flaviviruses include, without limitation:
  • 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 dianhea virus (“BVDV”), classical swine fever virus (“CSFV,” also called hog cholera virus), and border disease virus (“BDV”).
  • BVDV bovine viral dianhea virus
  • CSFV classical swine fever virus
  • BDV border disease virus
  • a compound of Formula (I), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (I):
  • R 1 , R 2 and R 3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate); straight chained, branched or cyclic alkyl (including lower alkyl); acyl (including lower acyl); CO-alkyl, CO-aryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO- substituted aryl, 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; alkylsulfonyl, arylsulfonyl, aralkylsulfonyl, a lipid, including a phospholipid; an amino acid; and amino acid residue, a carbohydrate; a peptide; cholesterol; or other pharmaceutically acceptable leaving group
  • Y 1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR 4 , NH* NHR 4 , NR 4 R 5 , SH or SR 4 ;
  • X 1 is a straight chained, branched or cyclic optionally substituted alkyl, CH 3 , CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , CH 2 OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR 4 , COO-alkyl, COO-aryl, CO- Oalkoxyalkyl, CONH* CONHR 4 , CON(R 4 )* chloro, bromo, fluoro, iodo, CN, N 3s OH,
  • X 2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH 3 , CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , CH 2 OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR 4 , COO-alkyl, COO-aryl, CO- Oalkoxyalkyl, CONH* CONHR 4 , CON(R 4 )* chloro, bromo, fluoro, iodo, CN, N 3 , OH,
  • each Y 3 is independently H, F, Cl, Br or I; each R 4 and R 5 is independently hydrogen, acyl (including lower acyl), alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl), lower alkyl, alkenyl, alkynyl or cycloalkyl.
  • a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (I) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein:
  • R 1 is H or phosphate (preferably H);
  • R 2 and R 3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R 2 and R 3 is acyl or an amino acid residue;
  • X 1 is CH 3 , CF 3 or CH 2 CH 3 ;
  • X 2 is H orNH 2 ;
  • Y is hydrogen, bromo, chloro, fluoro, iodo, NH or OH.
  • a compound of Formula (II) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (II):
  • a compound of Formula (II), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (II) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: R 1 is H or phosphate (preferably H);
  • R 2 and R 3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R 2 and R 3 is acyl or an amino acid residue;
  • X 1 is CH 3 , CF 3 or CH 2 CH 3 ; 2004/002
  • X 2 is H, F, Cl, Br, I or CH 3 ;
  • Y is hydrogen, bromo, chloro, fluoro, iodo, NH 2 or OH.
  • a compound of Formula (III), (IN) or (V) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV), or (V):
  • BAA BAB
  • BAC BAD
  • each W 1 , W 2 , W 3 and W 4 is independently N, CH, CF, CI, CBr, CCl, CCN, CCH 3 , CCF 3 , CCH 2 CH 3 , CC(O)NH 2 , CC(O)NHR 4 , CC(O)N(R 4 )* CC(O)OH, CC(0)OR 4 or CX 3 ; each W* is independently O, S, NH or NR 4 ; wherein for Base (B), W 4 cannot be CH if W 1 , W 2 and W 3 are N; wherein for Base (E), (F), (K), (L), (W) and (X), W 4 cannot be CH if W 1 is N; X is O, S, SO* CH* CH 2 OH, CHF, CF* C(Y 3 )* CHCN, C(CN) 2 , CHR 4 or C(R 4 ) 2 ; X* is CH, CF, CY 3 or
  • the compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof or the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV), or (V) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, wherein:
  • R 1 is H or phosphate (preferably H);
  • R 2 and R 3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R 2 and R 3 is acyl or an amino acid residue;
  • W 4 is CX 3 ;
  • X 3 is CH 3 , CF 3 or CH 2 CH 3 ;
  • R 6 is alkyl
  • X is O, S, SO 2 or CH*
  • the compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof or the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, wherein:
  • R 1 is H or phosphate (preferably H);
  • R 2 and R 3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R 2 and R 3 is an amino acid residue;
  • W 4 is CX 3 ;
  • X 3 is CH 3 , CF 3 or CH 2 CH 3 ;
  • R 6 is alkyl
  • X is O, S, SO 2 or CH*
  • the compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof or the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, wherein:
  • R 1 is H or phosphate (preferably H);
  • R 2 and R 3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R 2 and R 3 is acyl or an amino acid residue;
  • W 4 is CX 3 ;
  • X 3 is CH 3 , CF 3 or CH 2 CH 3 ;
  • R is alkyl; and X is O.
  • the compound of Formula (IV(a)), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof is provided, as well as the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (IV(a)):
  • Base is as defined herein; optionally substituted with an amine or cyclopropyl (e.g., 2- amino, 2,6-diamino or cyclopropyl guanosine);
  • an amine or cyclopropyl e.g., 2- amino, 2,6-diamino or cyclopropyl guanosine
  • R 7 is halo (F, Cl, Br or I), though preferably F;
  • 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; 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 2 is not phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); and R 2 is 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; 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 2 is not phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug).
  • a compound of Formula (VI) or (VII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof is provided, as well as a method for the freatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or (VII):
  • R 6 and R 7 or R 9 and R 10 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O 5 S and/or N).
  • a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, in which: • X is O, S, SO or SO 2 ; and/or • each R 6 is independently an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH 2 OH, CH2NH2, CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF
  • each R 7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, CO 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-alken
  • each R 9 is independently hydrogen, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl- 5 O-acyl, F, Cl, Br, I,
  • CN NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S-cycloalkyl, C0 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3> CF 2 CF 3 , CH 2 C0 2 R 4 ,
  • each R 10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substimted cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3 , CF 2 CF 3 , CH 2 C0 2 R 4 , (CH 2 ) m COOH, (CH 2 )
  • each R 8 and R 11 is independently H, CH 3 , CH 2 OH, CH 2 F, CH 2 N 3 , (CH 2 ) m COOH,
  • Base is selected from one of the following:
  • each R', R", R'" and R" is independently selected from the group consisting of
  • H substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Q and Q 2 independently are N or C-R; wherein R is H, alkyl or acyl;
  • Q 3j Q 4 , Q5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, in which:
  • X is O, S 5 SO or S0 2 ;
  • R 6 and R 7 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • each R 9 is independently hydrogen, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, C Br, I,
  • CN NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl 5 S-aralkyl, S-acyl, S-cycloalkyl, CO 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3s CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3 , CF 2 CF 3 , CH 2 CO 2 R 4 ,
  • each R 10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF 2 CF 3 , CH 2 C0 2 R 4 , (CH 2 ) m COOH, (CH 2 ) m COOR 4 ,
  • each m is independently 0 or 1 ; and/or Base is selected from one of the following:
  • each R', R", R'" and R" is independently selected from the group consisting of
  • H substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • W is C-R" or N
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Qj and Q 2 independently are N or C-R; wherein R is H, alkyl or acyl;
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VI), or its pharmaceutically acceptable salt or prodrug thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which:
  • X is O, S, SO or SO 2 ;
  • each R 6 is independently an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH 2 OH,
  • each R 7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, CO 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-alken
  • R 9 and R 10 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or • each R 8 and R 11 is independently H, CH 3 , CH 2 OH, CH 2 F, CH 2 N 3 , (CH 2 ) m COOH,
  • each m is independently 0 or 1 ;
  • Base is selected from one of the following: wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • W is C-R" orN
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Qi and Q 2 independently are N or C-R; wherein R is H, alkyl or acyl;
  • a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which:
  • R 6 and R 7 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • R 9 and R 10 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • each R 8 and R n is independently H, CH 3 , CH 2 OH, CH 2 F, CH 2 N 3 , (CH 2 ) m COOH, (CH 2 ) m COOR 4 , (CH 2 ) m CONH* (CH 2 ) m CON(R 4 )* (CH 2 ) m CONHR 4 andN-acyl; and/or
  • each m is independently 0 or 1 ; and/or • Base is selected from one of the following:
  • each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, 0-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- 2004/002999
  • W is C-R" orN; T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
  • Q ⁇ and Q 2 independently are N or C-R; wherein R is H, alkyl or acyl;
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VI), or its pharmaceutically acceptable salt or prodrug thereof, or a stereoisomeric, tautomeric or polymorphic form thereof as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, in which:
  • X is CH* CH 2 OH, CHF, CF* C(Y 3 )* CHCN, C(CN)* CHR 4 or C(R ) 2 ;
  • each R 6 is independently an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH 2 OH, CH 2 NH 2 , CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3,
  • each R 7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN,
  • NCO NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, C0 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3 , CF 2 CF 3 , CH 2 CO 2 R 4 , (CH 2 ) m COOH,
  • heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination;
  • each R 9 is independently hydrogen, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I,
  • CN NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S-cycloalkyl, C0 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF 2 CF 3 , CH 2 CO 2 R 4 ,
  • each R 10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF 2 CF 3 , CH 2 C0 2 R 4 , (CH 2 ) m COOH, (CH 2 ) m COOR 4 , (CH 2 ) m CONH* (CH 2 ) m CON(R 4 ) 2 , (CH 2 ) m CONHR 4 , an optionally substimted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or • each R 10 is independently hydrogen, an optionally substituted lower alkyl,
  • each m is independently 0 or 1 ;
  • Base is selected from one of the following:
  • each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • W is C-R" orN
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Qi and Q 2 independently are N or C-R;
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof as well as a method for the freatment of a host infected with a
  • Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which: 2004/002999
  • X is CH* CH 2 OH, CHF, CF* C(Y 3 )* CHCN, C(CN)* CHR 4 or C(R 4 ) 2 ;
  • R 6 and R 7 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • each R 9 is independently hydrogen, optionally substituted lower alkyl, optionally substimted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl,
  • each R 10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 ,
  • each R 8 and R 11 is independently H, CH 3 , CH 2 OH, CH 2 F, CH 2 N 3 , (CH 2 ) m COOH, (CH 2 ) m COOR 4 , (CH 2 ) m CONH* (CH 2 ) m CON(R 4 ) 2 , (CH 2 ) m CONHR 4 andN-acyl; and/or • each m is independently 0 or 1; and/or
  • Base is selected from one of the following:
  • each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • W is C-R" orN
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
  • O and Q 2 independently are N or C-R; wherein R is H, alkyl or acyl;
  • Q 3; Q 4s Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which:
  • X is CH* CH 2 OH, CHF, CF* C(Y 3 )* CHCN, C(CN) 2 , CHR 4 or C(R 4 ) 2 ;
  • each R 6 is independently an optionally substimted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substimted cycloalkyl, CH 2 OH,
  • each R 7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl 5 S- cycloalkyl, CO 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-alken
  • R 9 and R 10 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or • each R s and R 11 is independently H, CH 3 , CH 2 OH, CH 2 F, CH 2 N 3 , (CH 2 ) m COOH,
  • each m is independently 0 or 1;
  • Base is selected from one of the following: wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • W is C-R" orN
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
  • O and Q 2 independently are N or C-R; wherein R is H, alkyl or acyl;
  • Q 3j Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which:
  • X is CH* CH 2 OH, CHF, CF* C(Y 3 )* CHCN, C(CN)* CHR 4 or C(R 4 ) 2 ;
  • R 6 and R 7 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • R 9 and R 10 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • each R 8 and R n is independently H, CH 3 , CH 2 OH, CH 2 F, CH 2 N 3 , (CH 2 ) m COOH, (CH 2 ) m COOR 4 , (CH 2 ) m CONH* (CH 2 ) m CON(R 4 )* (CH 2 ) m CONHR 4 andN-acyl; and/or
  • each m is independently 0 or 1 ; and/or • Base is selected from one of the following:
  • each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, CL Br, I, CN, COOH, CONH* C0 2 -alkyl, CONH-alkyl,
  • W is C-R" orN; T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Q ⁇ and Q 2 independently are N or C-R; wherein R is H, alkyl or acyl;
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VII), or its pharmaceutically acceptable salt or prodrug thereof, or a stereoisomeric, tautomeric or polymorphic form thereof as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which:
  • X* is CH, CF, CY 3 or CR 4 ;
  • each R 6 is independently an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substimted alkynyl, optionally substimted cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3,
  • each R 7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN,
  • each R 9 is independently hydrogen, optionally substimted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I,
  • CN NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S-cycloalkyl, CO 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF 2 CF 3 , CH 2 CO 2 R 4 ,
  • each R 10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substimted alkynyl, optionally substimted cycloalkyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF 2 CF 3 , CH 2 C0 2 R 4 , (CH 2 ) m COOH, (CH 2 )
  • each m is independently 0 or 1 ;
  • Base is selected from one of the following:
  • each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • W is C-R" orN
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
  • Qi and Q 2 independently are N or C-R; wherein R is H, alkyl or acyl;
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof as well as a method for the freatment of a host infected with a
  • Flaviviridae comprising administering an effective freatment amount of compound of Formula (Nil) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which:
  • X* is CH, CF, CY 3 or CR 4 ;
  • R 6 and R 7 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more ⁇ , O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • each R 9 is independently hydrogen, optionally substituted lower alkyl, optionally substimted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I,
  • each R 10 is independently hydrogen, an optionally substituted lower alkyl, optionally substimted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH 2 OH, CH 2 NH 2 , CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3 , CF 2 CF 3 , CH 2 C0 2 R 4 , (CH 2 ) m COOH,
  • each m is independently 0 or 1;
  • Base is selected from one of the following: wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • W is C-R" orN
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
  • Qi and Q 2 independently are N or C-R;
  • R is H, alkyl or acyl
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which:
  • X* is CH, CF, CY 3 or CR 4 ;
  • each R 6 is independently an optionally substituted lower alkyl, optionally substimted alkenyl, optionally substimted alkynyl, optionally substimted cycloalkyl, CH 2 OH,
  • each R 7 is independently -OH, optionally substituted lower alkyl, optionally substimted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, CO 2 -alkyl, CONH-alkyl, CON-dialkyl, CONH-
  • R 9 and R 10 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or • each R 8 and R 11 is independently H, CH 3 , CH 2 OH, CH 2 F, CH 2 N 3 , (CH 2 ) m COOH,
  • each m is independently 0 or 1;
  • Base is selected from one of the following: wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO 2 -alkyl, CONH-alkyl
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
  • Qi and Q 2 independently are N or C-R;
  • R is H, alkyl or acyl
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (VII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which:
  • X* is CH, CF, CY 3 or CR 4 ;
  • R 6 and R 7 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • R 9 and R 10 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
  • each R 8 and R u is independently H, CH 3 , CH 2 OH, CH 2 F, CH 2 N 3 , (CH 2 ) m COOH, (CH 2 ) m COOR 4 , (CH 2 ) m CONH* (CH 2 ) m CON(R 4 )* (CH 2 ) m CONHR 4 and N-acyl; and/or
  • each m is independently 0 or 1; and/or • Base is selected from one of the following:
  • each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO 2 -alkyl, CONH-alkyl,
  • W is C-R" or N; T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Qi and Q 2 independently are N or C-R;
  • R is H, alkyl or acyl
  • Q 3; Q 4 , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • 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 substimted with one or more substituents as described in the
  • R 8 and R 10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; (5) X is O, S, S0 2 or CH 2 ; (6) W 4 is CX 3 ; and (7) X 3 is CH 3 , CF 3 or CH 2 CH 3 .
  • 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 substimted with one or more substituents as described
  • 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
  • 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
  • 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
  • R 1 is independently H or phosphate (including monophosphate, diphosphate, friphosphate, 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
  • the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof and the method for the freatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, in which: (1) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl
  • R 1 is independently H or phosphate
  • R 6 is alkyl
  • R 7 and R 9 are independently OR 2 , alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino or di(loweralkyl)-amino; (4) R 8 and R 10 are independently
  • 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 ary
  • the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which: (1) R 1 is independently H or phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); acyl
  • R 1 is independently H or phosphate
  • R 6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino or di(loweralkyl)amino
  • R 7 and R 9 are independently OR 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 substimted with one or more substituents
  • R 6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino or di(loweralkyl)amino; (3) R 7 and R 9 are independently OR 2 , alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O- alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino, or di(loweralkyl)amino; (4) R 8 and R 10 are hydrogen; (5) X is O; (6) W 4 is CX 3 ; and (7) X 3 is CH 3 , CF 3 or CH 2 CH 3 .
  • the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which: (1) R 1 is independently H or phosphate; (2) R 6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino or di(loweralkyl)amino; (3) R 7 and R 9 are independently OR 2 ; (4) R 8 and R 10 are hydrogen; (5) X is O
  • the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which: (1) R 1 is independently H or phosphate; (2) R 6 is alkyl; (3) R 7 and R 9 are independently OR 2 ; (4) R 8 and R 10 are hydrogen; (5) X is O, S, SO* or CH 2 ; (6) W 4 is CX 3 ; and (7) X 3 is CH 3 , CF 3 or CH 2 CH 3 .
  • the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which: (1) R 1 is independently H or phosphate; (2) R 6 is alkyl; (3) R 7 and R 9 are independently OR 2 ; (4) R 8 and R 10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; (5) X is O; (6) W 4 is CX 3 ; and (7) X 3 is CH 3 , CF 3 or CH 2 CH 3 .
  • the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof, is provided, in which: (1) R 1 is independently H or phosphate; (2) R 6 is alkyl;
  • R 7 and R 9 are independently OR 2 , alkyl (including lower alkyl), alkenyl, alkynyl, Br- vinyl, O-alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino or di(loweralkyl)amino; (4) R 8 and R 10 are hydrogen; (5) X is O; (6) W 4 is CX 3 ; and (7) X 3 is CH 3 , CF 3 or CH 2 CH 3 .
  • the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, in which:
  • Base 8-methyladenine; (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 8-methylguanine; (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; (1) Base is 6-methylcytosine; (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 6-methylthymidine; (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; 2004/002999
  • Base 6-methyluracil; (2) R 1 is hydrogen; (3) R 6 is methyl; (4) R 7 and R 9 are hydroxyl; (5) R and R ,10 are hydrogen; and (6) X is O;
  • Base 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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*
  • Base 8-methyladenine; (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*
  • a compound of Formula (VIII), (IX) or (X) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric, or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (VIII), (IX), or (X):
  • each R 12 is independently a substimted alkyl (including lower alkyl), CH 2 CN, CH 2 N 3 , CH 2 NH 2 , CH 2 NHCH 3 , CH 2 N(CH 3 ) 2 , CH 2 OH, halogenated alkyl (including halogenated lower alkyl), CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , substimted alkenyl, halo
  • R 12 and R 13 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); and each m is independently 0, 1 or 2.
  • a compound of Formula (XI) or (XII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric, or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (XI) or (XII):
  • R 8 in Formula (XI) is -OH or -NH 2 only when X is carbon; and wherein; each R 8 and R ⁇ is independently hydrogen, an optionally substimted alkyl (including lower alkyl), CH 3 , CH 2 CN, CH 2 N 3 , CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 OH, halogenated alkyl (including halogenated lower alkyl), CF 3 , C(Y 3 ) 3 , 2-Br-ethyl, CH 2 F, CH 2 C1, CH 2 CF 3 , CF 2 CF 3 , C(Y 3 ) 2 C(Y 3 ) 3 , optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH 2 C(0)OH, -CH 2 C(0)OR 4 ,
  • compounds of the Formula (XIII) or (XIV) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (XIII) or (XIV):
  • R 3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; acyl; a sulfonate ester; optionally substimted alkyl sulfonyl; optionally substituted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 3 is independently H, or mono-, di- or triphosphate; X" is selected from the group consisting of one or more O, S, SO, SO* N, NH, NR and CH 2 wherein any of the aforementioned may be optionally substimted and may be variably positioned so as to form a 3-7 membered ring
  • R is H, alkyl or acyl
  • B indicates a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substimted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or
  • Base is selected from the group consisting of:
  • each R', R", R'" and R" are independently selected from the group consisting of
  • H OH, substimted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
  • W is C-R" orN; T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Qi and Q 2 independently are N or C-R;
  • R is H, alkyl or acyl
  • Q 3 ⁇ Q , Q 5 and Q 6 independently are N or CH; and tautomeric forms thereof.
  • a compound of Formula (XV), (XVI) or (XVII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XV), (XVI), or (XVII):
  • G and E independently are selected from the group consisting of H, CH 3 , CH 2 OH 5 CH F, CH 2 N 3 , CH 2 CN, (CH 2 ) m COOH, (CH 2 ) m COOR, (CH 2 ) m CONH* (CH 2 ) m CONR* (CH 2 ) m CONHR and N-acyl; R is H, alkyl or acyl; m is O or 1; and R 3 and Base are as defined for Formula (XIII).
  • At most one of G and E can further be hydrogen.
  • a compound of Formula (XVIII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof is provided, as well as a method for the freatment of a host infected with a
  • Flaviviridae comprising administering an effective treatment amount of compound of Formula (XVIII):
  • R 3 and Base are as defined for Formula (XIII).
  • a compound of Formula (XIX), (XX), (XXI) (XXII) or (XX ⁇ i) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymo ⁇ hic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XIX), (XXI), (XXII), or (XXIII):
  • A is selected from the group consisting of optionally substituted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1 5 CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3 , CF 2 CF 3 , CH 2 CO 2 R, (CH 2 ) m COOH, (CH 2 ) m COOR, (CH 2 ) m CO- NH* (CH 2 ) m CONR* and (CH 2 ) m CONHR;
  • Y is selected from the group consisting of H, optionally substimted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH 2 OH, CH 2 NH* CH 2 NHCH 3 , CH 2 N(CH 3 )* CH 2 F, CH 2 C1, CH 2 N 3 , CH 2 CN, CH 2 CF 3 , CF 3, CF 2 CF 3 , CH 2 CO 2 R, (CH 2 ) m COOH, (CH 2 ) m COOR, (CH 2 ) m CONH* (CH 2 ) m CONR 2 , and (CH 2 ) m CONHR;
  • R is H, alkyl or acyl;
  • X is selected from the group consisting of -OH, optionally substituted alkyl, cycloalkyl, alkenyl, alkynyl, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aryl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N 3 , NH-acyl, NH-alkyl, N- dialkyl, NH-alkenyl, NH-alkynyl, NH-aryl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S- alkenyl, S-alkynyl, S-aryl, S-aralkyl, S-acyl, S-cycloalkyl, C0 2 -alkyl, CONH-alkyl,
  • Base is a non-natural base selected from the group of:
  • each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO 2 -alkyl, CONH-alkyl, CON-dialkyl, OH,
  • T and V independently are CH or N;
  • Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
  • Qi and Q 2 independently are N or C-R"";
  • Q 3 , Q4, Q5 and Q 6 independently are N or CH; with the proviso that in bases (g) and (i), R', R"" are not H, OH, or NH* and Q,
  • T, V, Q* Q 5 and Q 6 are not N.
  • a compound of Formula (IX), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (IX):
  • R 1 , R 2 and R 3 are independently H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl; CO-aryl; CO-alkoxyalkyl; CO-aryloxyalkyl; CO-substituted aryl; sulfonate ester; benzyl, wherein the phenyl group is optionally substituted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; or a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R 1 , R 2 and/or R 3 is independently H or phosphate;
  • X is O, S, SO 2 or CH 2 ;
  • Base* is a purine or pyrimidine base
  • R 12 is C(Y 3 ) 3 ;
  • Y 3 is independently H, F, Cl, Br or I;
  • R 13 is fluoro
  • X is O
  • Y 3 is H
  • R 1 , R 2 and R 3 are also H.
  • nucleosides of the present invention have several chiral centers and may exist in and be isolated in optically active and racemic forms. Some compounds may exhibit polymo ⁇ hism. It is to be understood that the present invention encompasses any racemic, optically-active, diastereomeric, polymorphic, or stereoisomeric form, or mixtures thereof, of a compound of the invention, which possess the useful properties described herein. It being well known in the art how to prepare optically active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).
  • the 1' and 4' carbons of the nucleoside are chiral, their nonhydrogen substituents (the base and the CHOR groups, respectively) can be either cis (on the same side) or trans (on opposite sides) with respect to the sugar ring system.
  • the four optical isomers therefore are represented by the following configurations (when orienting the sugar moiety in a horizontal plane such that the oxygen atom is in the back): cis (with both groups “up”, which corresponds to the configuration of naturally occuning ⁇ - D nucleosides), cis (with both groups “down”, which is a nonnaturally occuning ⁇ -L configuration), trans (with the C2' substituent "up” and the C4' substituent "down”), and trans (with the C2' substituent "down” and the C4' substituent "up”).
  • the "D-nucleosides” are cis nucleosides in a natural configuration and the "L-nucleosides” are cis nucleosides in the nonnaturally occuning configuration.
  • most amino acids are chiral (designated as L or D, wherein the L enantiomer is the naturally occuning configuration) and can exist as separate enantiomers.
  • methods to obtain optically active materials include at least the following. i) physical separation of crystals - a technique whereby macroscopic crystals of the individual enantiomers are manually separated.
  • This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct; ii) simultaneous crystallization - a technique whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state; iii) enzymatic resolutions - a technique whereby partial or complete separation of a racemate by virtue of differing rates of reaction for the enantiomers with an enzyme; iv) enzymatic asymmetric synthesis - a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer; v) chemical asymmetric synthesis - a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the
  • the resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer; vii) first- and second-order asymmetric transformations - a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer.
  • kinetic resolutions this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions; ix) enantiospecific synthesis from non-racemic precursors - a synthetic technique whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis; x) chiral liquid chromatography - a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase.
  • the stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions; xi) chiral gas chromatography - a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase; xii) extraction with chiral solvents - a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent; xiii) transport across chiral membranes - a technique whereby a racemate is placed in contact with a thin membrane barrier.
  • the barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane banier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
  • alkyl refers to a saturated straight, branched, or cyclic, primary, secondary, or tertiary hydrocarbon of typically C ⁇ to
  • Cio and specifically includes methyl, CF 3 , CC1 3 , CFC1* CF 2 C1, ethyl, CH 2 CF 3 , CF 2 CF 3 , propyl, isopropyl, cyclopropyl, butyl, isobutyl, secbutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 3-methylpentyl, 2,2- dimethylbutyl, and 2,3-dimethylbutyl.
  • the term includes both substimted and unsubstituted alkyl groups, and particularly includes halogenated alkyl groups, and even more particularly fluorinated alkyl groups.
  • moieties with which the alkyl group can be substimted are selected from the group consisting of halogen (fluoro, chloro, bromo or iodo), hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nifro, 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,
  • lower alkyl refers to a Ci to C 4 saturated straight, branched, or if appropriate, a cyclic (for example, cyclopropyl) alkyl group, including both substituted and unsubstituted moieties.
  • alkylamino or "arylamino” refers to an amino group that has one or two alkyl or aryl substituents, respectively. Unless otherwise specifically stated in this application, when alkyl is a suitable moiety, lower alkyl is prefened. Similarly, when alkyl or lower alkyl is a suitable moiety, unsubstituted alkyl or lower alkyl is prefened.
  • protected refers to a group that is added to an oxygen, nitrogen, or phosphorus atom to prevent its further reaction or for other pu ⁇ oses.
  • 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 unsubstimted moieties.
  • the aryl group can be substimted with any described moiety, including, but not limited to,one or more moieties selected from the group consisting of halogen (fluoro, chloro, bromo or iodo), 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.
  • 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 -alkylaminopurine, N 6 -thioalkyl purine, N 2 -alkylpurines, N 2 - alkyl-6-thiopurines, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6- azapyrimidine, including 6-azacytosine,
  • 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 frimethylsilyl, dimethylhexylsilyl, t- butyldimethylsilyl, and t-butyldiphenylsilyl, frityl, alkyl groups, and acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl.
  • acyl refers to a group of the formula C(O)R', wherein R' is an straight, branched, or cyclic alkyl (including lower alkyl), amino acid, aryl including phenyl, alkaryl, aralkyl including benzyl, alkoxyalkyl including methoxymethyl, aryloxyalkyl such as phenoxymethyl; or substimted alkyl (including lower alkyl), aryl including phenyl optionally substituted with chloro, bromo, fluoro, iodo, to C 4 alkyl or d to C 4 alkoxy, sulfonate esters such as alkyl or aralkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxy-trityl, substituted benzyl, alkaryl, aralkyl including benzyl,
  • Aryl groups in the esters optimally comprise a phenyl group.
  • acyl groups include acetyl, trifluoroacetyl, methylacetyl, cyclopropylacetyl, cyclopropyl carboxy, propionyl, butyryl, hexanoyl, heptanoyl, octanoyl, neo-heptanoyl, phenylacetyl, 2-acetoxy-2-phenylacetyl, diphenylacetyl, ⁇ -methoxy- ⁇ - trifluoromethyl-phenylacetyl, bromoacetyl, 2-nitro-benzeneacetyl, 4-chloro-benzeneacetyl, 2-chloro-2,2-diphenylacetyl, 2-chloro-2-phenylacetyl, frimethylacetyl, chlorodifluoroacetyl, perfluoroacetyl, fluoro
  • acyl when the term acyl is used, it is meant to be a specific and independent disclosure of acetyl, trifluoroacetyl, methylacetyl, cyclopropylacetyl, propionyl, butyryl, hexanoyl, heptanoyl, octanoyl, neo-heptanoyl, phenylacetyl, diphenylacetyl, ⁇ -frifluoromethyl-phenylacetyl, bromoacetyl, 4-chloro- benzeneacetyl, 2-chloro-2,2-diphenylacetyl, 2-chloro-2-phenylacetyl, frimethylacetyl, chlorodifluoroacetyl, perfluoroacetyl, fluoroacetyl, bromodifluoroacetyl, 2-thiopheneacetyl, tert-butylacetyl,
  • amino acid includes naturally occuning and synthetic , ⁇ ⁇ or ⁇ amino acids, and includes but is not limited to, amino acids found in proteins, i.e. glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine.
  • the amino acid is in the L-configuration.
  • the amino acid can be a derivative of alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, ⁇ - alanyl, ⁇ -valinyl, ⁇ -leucinyl, ⁇ -isoleuccinyl, ⁇ -prolinyl, ⁇ -phenylalaninyl, ⁇ -tryptophanyl, ⁇ -methioninyl, ⁇ -glycinyl, ⁇ -serinyl, ⁇ -threoninyl, ⁇ -cysteinyl
  • Tables 1-24 set out examples of species within the present invention.
  • amino acid it is considered to be a specific and independent disclosure of each of the esters of ⁇ , ⁇ ⁇ or ⁇ glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine in the D and L-configurations.
  • nucleoside composition that includes at least 85 or 90% by weight, preferably
  • the compounds are substantially free of enantiomers.
  • isolated refers to a nucleoside composition that includes at least 85%, 90%, 95%, 98%, 99%, or 100% by weight, of the nucleoside, the remainder comprising other chemical species or enantiomers.
  • 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 Flaviviridae viral 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 Flaviviridae genome and animals, in particular, primates (including chimpanzees) and humans. In most animal applications of the present invention, the host is a human patient. Veterinary applications, in certain indications, however, are clearly anticipated by the present invention (such as chimpanzees).
  • 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 a Flaviviridae, or are metabolized to a compound that exhibits such activity.
  • 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 refened to as "physiologically acceptable salts"), and a compound, which has been alkylated, acylated, or otherwise modified at the 5 '-position, or on the purine or pyrimidine base (a type of "pharmaceutically acceptable prodrug").
  • physiologically acceptable salts alternatively refened to as "physiologically acceptable salts”
  • a compound, which has been alkylated, acylated, or otherwise modified at the 5 '-position, or on the purine or pyrimidine base a type of "pharmaceutically acceptable prodrug”
  • 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
  • pharmaceutically acceptable salts are organic acid addition salts formed by addition of acids, which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorate, ⁇ -ketoglutarate, ⁇ -glycerophosphate, formate, fumarate, propionate, glycolate, lactate, pyruvate, oxalate, maleate, and salicylate.
  • acids which form a physiological acceptable anion
  • Suitable inorganic salts may also be formed, including, sulfate, nitrate, bicarbonate, carbonate salts, hydrobromate and phosphoric acid.
  • the salt is a mono- or di- hydrochloride salt.
  • 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.
  • the salt is a hydrochloride salt of the compound.
  • the pharmaceutically acceptable salt is a dihydrochloride salt.
  • 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 reduces polarity and allows passage into cells.
  • 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. Bischoferger, Antiviral Research, 1995, 27:1-17. Any of these can be used in combination with the disclosed nucleosides to achieve a desired effect.
  • the nucleoside is delivered as a phosphonate or a
  • the active nucleoside can also be provided as a 2', 3' and/or 5'-phosphoether lipid or a 2', 3' and/or 5'-ether lipid.
  • Non-limiting examples are described include 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. 1990. "Novel membrane-interactive ether lipid analogs that inhibit infectious HJN-1 production and induce defective virus formation.” AIDS Res. Hum. Retro Viruses. 6:491-501; Piantadosi, C, J. Marasco C.J., S.L.
  • Nonlimiting examples of U.S. patents that disclose suitable lipophilic substituents that can be covalently incorporated into the nucleoside, preferably at the 2', 3' and/or 5'-OH position of the nucleoside or lipophilic preparations include U.S. Patent Nos. 5,149,794 (Sep. 22, 1992, Yatvin et al); 5,194,654 (Mar. 16, 1993, Hostetler et al, 5,223,263 (June 29, 1993, Hostetler et al); 5,256,641 (Oct. 26, 1993, Yatvin et al); 5,411,947 (May 2, 1995, Hostetler et al); 5,463,092 (Oct.
  • lipophilic substituents that can be attached to the nucleosides of the present invention, or lipophilic preparations, include WO 89/02733, W0 90/00555, W0 91/16920, W0 91/18914, W0 93/00910, W0 94/26273, W0 96/15132, EP 0350287, EP 93917054.4, and W0 91/19721.
  • Aryl esters especially phenyl esters, are also provided. Nonlimiting examples are disclosed in DeLambert et al., J. Med. Chem. 37: 498 (1994). Phenyl esters containing a carboxylic ester ortho to the phosphate are also provided. Khamnei and Tonence, J. Med. Chem.; 39:4109-4115 (1996). In particular, benzyl esters, which generate the parent compound, in some cases using substituents at the ortho- or para-position to accelerate hydrolysis, are provided. Examples of this class of prodrugs are described by Mitchell et al., J. Chem. Soc. Perkin Trans. I 2345 (1992); Brook, et al. WO 91/19721; and Glazier et al. WO 91/19721.
  • Cyclic and noncyclic phosphonate esters are also provided. Nonlimiting examples are disclosed in Hunston et al., J. Med. Chem. 27: 440-444 (1984) and Starrett et al. J. Med.
  • cyclic 3',5'-phosphate esters are provided. Nonlimiting examples are disclosed in Meier et al. J. Med. Chem. 22: 811-815 (1979). Cyclic l',3'-propanyl phosphonate and phosphate esters, such as ones containing a fused aryl ring, i.e. the cyclosaligenyl ester, are also provided (Meier et al., Bioorg. Med. Chem. Lett. 7: 99-104 (1997)). Unsubstituted cyclic l',3'- ⁇ ropanyl esters of the monophosphates are also provided (Farquhar et al., J.
  • Cyclic phosphoramidates are known to cleave in vivo by an oxidative mechanism. Therefore, in one embodiment of the present invention, a variety of substituted l',3' propanyl cyclic phosphoramidates are provided. Non-limiting examples are disclosed by Zon, Progress in Med. Chem. 19, 1205 (1982). Additionally, a number of 2'- and 3'- substimted proesters are provided.
  • 2'-Substituents include methyl, dimethyl, bromo, trifluoromethyl, chloro, hydroxy, and methoxy; 3 '-substituents including phenyl, methyl, trifluoromethyl, ethyl, propyl, i-propyl, and cyclohexyl. A variety of l'-substituted analogs are also provided.
  • Cyclic esters of phosphorus-containing compounds are also provided. Non-limiting examples are described in the following:
  • V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, optionally 1 heteroatom, substimted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from both O groups attached to the phosphorus; or
  • V and Z are connected via an additional 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryl group at the beta, and gamma position to the O attached to the phosphorus;
  • V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said carbon atoms that is three atoms from an O attached to the phosphorus;
  • Z and W are connected via an additional 3-5 atoms to form a cyclic group, optionally containing one heteroatom, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
  • W and W are connected via an additional 2-5 atoms to form a cyclic group, optionally containing 0-2 heteroatoms, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
  • Z is selected from the group consisting of -CHR 2 OH, -CHR 2 OC(O)R 3 , -CHR 2
  • R 2 is selected from the group consisting of R 3 and -H;
  • R 3 is selected from the group consisting of alkyl, aryl, alicyclic, and aralkyl; • R 12 is selected from the group consisting of -H, and lower acyl;
  • M is the biologically active agent, and that is attached to the phosphorus in formula I via the 2', 3' and/or 5'-hydroxyl.
  • the active compounds of the present invention can be administered in combination or alternation with another anti-flavivirus or pestivirus agent, or in particular an anti-HCV agent to treat any of the conditions described herein.
  • effective dosages of two or more agents are administered together, whereas in alternation or sequential-step therapy, an effective dosage of each agent is administered serially or sequentially.
  • the dosages given will depend on abso ⁇ tion, 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.
  • an anti-HCV (or anti-pestivirus or anti-flavivirus) compound that exhibits an EC 50 of 10-15 ⁇ M, or preferably less than 1-5 ⁇ M, is desirable.
  • the efficacy of a drug against the viral 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 prefened over alternation therapy because it induces multiple simultaneous stresses on the virus.
  • Any of the viral treatments described in the Background of the Invention can be used in combination or alternation with the compounds described in this specification. Nonlimiting examples include:
  • Protease inhibitors examples include substrate-based NS3 protease inhibitors (Attwood et al., Antiviral peptide derivatives, PCT WO 98/22496, 1998; Attwood et al., Antiviral Chemistry and Chemotherapy 1999, 10, 259-273; Attwood et al., Preparation and use of amino acid derivatives as anti-viral agents, German Patent Pub. 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); Non-substrate-based NS3 protease 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.
  • Eglin c isolated from leech, is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, ⁇ -chymotrypsin, chymase and subtilisin. Qasim M.A. et al., Biochemistry 36:1598-1607, 1997.
  • U.S. patents disclosing protease inhibitors for the treatment of HCV include, for example, U.S. Patent No. 6,004,933 to Spruce et al. which discloses a class of cysteine protease inhibitors for inhibiting HCV endopeptidase 2; U.S. Patent No. 5,990,276 to Zhang et al. which discloses synthetic inhibitors of hepatitis C virus NS3 protease; U.S. Patent No. 5,538,865 to Reyes et a; WO 02/008251 to Corvas International, Inc, and WO 02/08187 and WO 02/008256 to Schering Corporation.
  • HCV inhibitor tripeptides are disclosed in US Patent Nos.
  • Diaryl peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/48172 to Schering Co ⁇ oration.
  • Imidazoleidinones as NS3 serine protease inhibitors of HCV are disclosed in WO 02/08198 to Schering Co ⁇ oration and WO 02/48157 to Bristol Myers Squibb.
  • Vertex Pharmaceuticals and WO 02/48116 to Bristol Myers Squibb also disclose HCV protease inhibitors.
  • S-ODN Antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5' non-coding region (NCR) of the virus (Alt M. et al., Hepatology, 1995, 22, 707-717), or nucleotides 326-348 comprising the 3' end of the NCR and nucleotides 371-388 located in the core coding region of the HCV RNA (Alt M. et al., Archives of Virology, 1997, 142, 589-599; Galderisi U.
  • Patent 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.
  • RNA replication inhibitors (VP50406) by ViroPharma/Wyeth, therapeutic vaccine by Intercell, therapeutic vaccine by Epimmune/Genencor, IRES inhibitor by Anadys, ANA 245 and ANA 246 by Anadys, immunotherapy (Therapore) by Avant, protease inhibitor by Corvas/SChering, helicase inhibitor by Vertex, fusion inhibitor by Trimeris, T cell therapy by CellExSys, polymerase inhibitor by Biocryst, targeted RNA chemistry by PTC Therapeutics, Dication by Immtech, Int., protease inhibitor by Agouron, protease inhibitor by Chiron/Medivir, antisense therapy by AVI BioPharma, antisense therapy by Hybridon, hemopurifier by
  • Hosts including humans, infected with pestivirus, flavivirus, HCV infection, or any other condition described herein, or another organism replicating through a RNA-dependent RNA viral polymerase, or for treating any other disorder described herein, 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 canier or dilutent.
  • 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 pestivirus, flavivirus or HCV 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. Lower doses may be preferable, for example doses of 0.5-100 mg, 0.5-50 mg, 0.5-10 mg, or 0.5-5 mg per kilogram body weight per day. Even lower doses may be useful, and thus ranges can include from 0.1-0.5 mg per kilogram body weight 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.
  • An oral dosage of 50-1000 mg is usually convenient, including in one or multiple dosage forms of 50, 100, 200, 250, 300, 400, 500, 600, 700, 800, 900 or 1000 mgs.
  • Lower doses may be preferable, for example from 10-100 or 1-50 mg.
  • Also contemplated are doses of 0.1-50 mg, or 0.1-20 mg or 0.1-10.0 mg.
  • lower doses may be utilized in the case of administration by a non-oral route, as, for example, by injection or inhalation.
  • 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
  • compositions 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 prefened mode of administration of the active compound is oral.
  • Oral compositions will generally include an inert diluent or an edible canier. They may be enclosed in gelatin capsules or compressed into tablets.
  • the active compound can be inco ⁇ orated with excipients and used in the form of tablets, troches, or capsules.
  • Pharmaceutically compatible binding agents, and/or adjuvant materials can e 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, sucutaneous, 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 ethylenediaminetefraacetic 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.
  • prefened caniers are physiological saline or phosphate buffered saline (PBS).
  • the active compounds are prepared with caniers 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 Co ⁇ oration.
  • Liposomal suspensions including liposomes targeted to infected cells with monoclonal antibodies to viral antigens are also prefened 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. Patent No. 4,522,811 (which is inco ⁇ orated herein by reference in its entirety).
  • 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.
  • 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.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , Y, W 1 , W 2 , W 3 , X, X 1 , X 2 and X 3 are as defined herein can be prepared by one of the following general methods.
  • 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
  • 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 Lewis acid such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate
  • 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.
  • the l'-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), substimtion 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.
  • 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 l'-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 conesponding to the compounds of the invention can be prepared following the same general methods (1 or 2), beginning with the conesponding L-sugar or nucleoside L-enantiomer as starting material.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 9 , R 10 , Y, W 1 , W 2 , W 3 , X, X 1 , X 2 and X 3 are as defined herein 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.
  • 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 substimted 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* ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 O 2 - ammonium molybdate, NaBr0 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
  • 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 substimted 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, MhO* ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 0 2 -ammonium molybdate, NaBr0 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 conesponding to the compounds of the mvention can be prepared following the same foregoing general methods, beginning with the conesponding L-sugar or nucleoside L-enantiomer as starting material.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , Y, W 1 , W 2 , W 3 , X, X 1 , X 2 and X 3 are as defined herein can be prepared by one of the following general methods.
  • 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, substimtion, 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* ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 0 2 - ammonium molybdate, NaBr0 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,
  • 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 trimethylsilylfriflate 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 substimted 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 temperamre 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* rathenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 0 2 -ammonium molybdate, NaBr0 2 -CAN, NaOCl in HO Ac, 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 conesponding to the compounds of the invention can be prepared following the same foregoing general methods, beginning with the conesponding L-sugar or nucleoside L-enantiomer as starting material. D. General Synthesis of 4 '-C-Branched Nucleosides
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R s , R 9 , R 10 , Y, W 1 , W 2 , W 3 , X, X 1 , X 2 and X 3 are as defined herein can be prepared by one of the following general methods. 1. Modification from the pentodialdo-furanose
  • the key starting material for this process is an appropriately substituted pentodialdo- furanose.
  • the pentodialdo-furanose can be purchased or can be prepared by any known means including standard epimerization, substimtion and cyclization techniques.
  • the pentodialdo-furanose is prepared from the appropriately substimted hexose.
  • the hexose can be purchased or can be prepared by any known means including standard epimerization (e.g. via alkaline treatment), substimtion and coupling techniques.
  • the hexose can be either in the furanose form, or cyclized via any means known in the art, such as methodology taught by Townsend Chemistry of Nucleosides and Nucleotides, Plenum Press, 1994, preferably by selectively protecting the hexose, to give the appropriate hexafuranose.
  • the 4'-hydroxymethylene of the hexafuranose then can be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperamre to yield the 4'- aldo-modified sugar.
  • Possible oxidizing agents are Swern reagents, 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* ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl 2 -pyridine, H 2 0 2 -ammonium molybdate, NaBrO 2 -CAN, NaOCl in HOAc, copper chromite, copper oxide, Raney nickel, palladium acetate, Meerwin-Pondorf-Verley reagent (alumin
  • the pentodialdo-furanose 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.
  • a suitable protecting group preferably with an acyl or silyl group
  • the protected pentodialdo-furanose can then be coupled with a suitable electrophilic alkyl, halogeno-alkyl (i.e. CF 3 ), alkenyl or alkynyl (i.e. allyl), to obtain the 4'-alkylated sugar.
  • the protected pentodialdo-furanose can be coupled with the conesponding carbonyl, such as formaldehyde, in the presence of a base, such as sodium hydroxide, with the appropriate polar solvent, such as dioxane, at a suitable temperamre, which can then be reduced with an appropriate reducing agent to give the 4 '-alkylated sugar.
  • the reduction is canied out using PhOC(S)Cl, DMAP, preferably in acetonitrile at room temperamre, followed by treatment of ACC ⁇ and TMSS refluxed in toluene.
  • 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 temperamre.
  • a Lewis acid such as tin tetrachloride, titanium tetrachloride or trimethylsilyltriflate in the appropriate solvent at a suitable temperamre.
  • 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.
  • the 4 '-C-branched ribonucleoside is desired.
  • deoxyribonucleoside is desired.
  • a formed ribo-nucleoside 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 conesponding to the compounds of the invention can be prepared following the same foregoing general methods, beginning with the conesponding L- pentodialdo-furanose as starting material.
  • the key starting material for this process is an appropriately substituted 1', 2', 3' or 4'-branched ⁇ -D or ⁇ -L nucleosides.
  • the branched nucleoside can be purchased or can be prepared by any known means including the techniques disclosed herein.
  • the branched nucleoside can be optionally protected with a suitable protecting group, preferably with a silyl group, by methods well known to those skilled in the art, as taught by Greene et al.
  • the protected branched nucleoside can then be coupled with a suitable acyl doner, such as an acyl chloride and/or an acyl anhydride with the appropriate protic or aprotic solvent at a suitable temperamre, to give the 2' and/or 3' prodrug of 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside.
  • a suitable acyl doner such as an acyl chloride and/or an acyl anhydride with the appropriate protic or aprotic solvent at a suitable temperamre
  • the protected branched nucleoside can then be coupled with a suitable acyl, such as a carboxylic acid, such as alkanoic acid and/or amino acid residue, optionally with a suitable coupling agent, with the appropriate aprotic solvent at a suitable temperature, to give the 2' and/or 3' prodrug of 1', 2', 3' or 4 '-branched ⁇ -D or ⁇ -L nucleoside.
  • a suitable acyl such as a carboxylic acid, such as alkanoic acid and/or amino acid residue
  • a suitable coupling agent with the appropriate aprotic solvent at a suitable temperature
  • Possible coupling reagents are any reagents that promote coupling, including but are not limiting to, Mitsunobu reagents (e.g. diisopropyl azodicarboxylate and diethyl azodicarboxylate) with friphenylphosphine or various carbodiimides.
  • simple amino-alcohols can be esterified using acid chlorides in refluxing acetonitrile-benzene mixture (See Scheme 7 below: Synthetic Communications, 1978, 8(5), 327-333; hereby incorporated by reference).
  • esterification can be achieved using an anhydride, as described in J Am. Chem. Soc, 1999, 727(24), 5661-5664, which is hereby inco ⁇ ortated by reference. See Figures 2, 3 and 4.
  • ACN acetonitrile
  • IR spectra were measured on a Nicolet 51 OP FT-IR spectrometer. Mass spectra were recorded on a Micromass Autospec high-resolution mass spectrometer. TLC were performed on Uniplates (silica gel) purchased from Analtech Co. Column chromatography was performed using either silica gel-60 (220-440 mesh) for flash chromatography or silica gel G (TLC grade, > 440 mesh) for vacuum flash column chromatography. UV spectra were obtained on a Beckman DU 650 spectrophotometer.
  • Elemental analysis was performed by Atlantic Microlab, Inc., Norcross, GA, or Galbraith Laboratories, Inc., Knoxville, TN.
  • HPLC was performed with a Waters HPLC system (Millipore Corporation, Milford, MA) equipped with a Model 600 controller, a Model 996 photodiode anay detector and a Model 717 plus autosampler. Millennium 2010 software was used for system control, data acquisition and processing.
  • nucleosides of Formula XXIV are prepared.
  • nucleosides of Formula XXVI are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXVII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXVIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXIX are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • Carbonyldiimidazole (377 mg, 2.33 mmol) was added to a solution of N-(tert-butoxycarbonyl)-L-valine (507 mg, 2.33 mmol) in 15 mL of anhydrous tetrahydrofuran.
  • nucleosides of Formula XXX are prepared.
  • R , 1 1 , ⁇ R>2 , X v l, v X2, and Y are defined herein.
  • nucleosides of Formula XXXI are prepared, using the appropriate sugar and pyrimidine bases.
  • nucleosides of Formula XXXII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXXIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXXIV are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXXV are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXXVII are prepared, using the appropriate sugar and pyrimidine bases.
  • nucleosides of Formula XXXVIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXXX are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • nucleosides of Formula XXXXI are prepared, using the appropriate sugar and pyrimidine or purine bases.
  • the title compound can be prepared according to a published procedure (Jones, G. H.; Moffatt, J. G. Oxidation of carbohydrates by the sulfoxide-carbodiimide and related methods. Oxidation with dicyclohexylcarbodiimide-DMSO, diisopropylcarbodiimide- DMSO, acetic anhydride-DMSO, and phosphorus pentoxide-DMSO: in Methods in
  • the title compound can be prepared according to a published procedure (Gunic, E.; Girardet, J.-L.; Pietrzkowski, Z.; Esler, C; Wang, G. "Synthesis and cytotoxicity of 4'-C- and 5"-C-substituted Toyocamycins" Bioorg. Med. Chem. 2001, 9, 163-170).
  • EXAMPLE 11 PREPARATION OF O-6-DlPHENYLCARBAMOYL-N 2 -ISOBUTYRYL-9-(2,3- DI-O-ACETYL-5-O-BENZOYL-4-C-METHYL- ⁇ -D-RIBOFURANOSYL)-8-METHYLGUANINE -

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Virology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

2' and/or 3' prodrugs of l', 2', 3' or 4'-branchednucleosides, and their pharmaceutically acceptable salts and derivatives are described. These prodrugs are useful in the prevention and treatment of Flaviviridae infections, including HCV infection, and other related conditions. Compounds and compositions of the prodrugs of the present invention are described. Methods and uses are also provided that include the administration of an effective amount of the prodrugs of the present invention, or their pharmaceutically acceptable salts or derivatives. These drugs may optionally be administered in combination or alteration with further anti-viral agents to prevent or treat Flaviviridae infections and other related conditions.

Description

MODIFIED V AND 3'-NUCLEOSIDE PRODRUGS FOR TREATING FLA VIVIRIDAE INFECTIONS
CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of priority to U.S. Provisional application No.
60/392,350, filed June 28, 2002; U.S. Provisional Application No. 60/466,194, filed April 28, 2003; and U.S. Provisional Application No. 60/470,949, filed May 14, 2003, the disclosures of each of which are incorporated herein by reference.
FIELD OF THE INVENTION This invention is in the area of pharmaceutical chemistry, and is in particular, a 2' and/or 3' prodrug of 6-modified, V, 2', 3' or 4'-branched pyrimidine nucleoside or 8- modifϊed, 1', 2', 3' or 4 '-branched purine nucleoside for the treatment of a Flaviviridae infection, such as a hepatitis C virus infection.
BACKGROUND OF THE INVENTION Flaviviridae Viruses
The Flaviviridae family of viruses comprises at least three distinct genera: pestiviruses, which cause disease in cattle and pigs; flaviviruses, which are the primary cause of diseases such as dengue fever and yellow fever; and hepaciviruses, whose sole member is HCV. 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).
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.
Pestiviruses and hepaciviruses are closely related virus groups within the Flaviviridae family. Other closely related viruses in this family include the GB virus A, GB virus A-like agents, GB virus-B and GB virus-C (also called hepatitis G virus, HGV). The hepacivirus group (hepatitis C virus; HCV) consists of a number of closely related but genotypically distinguishable viruses that infect humans. There are approximately 6 HCV genotypes and more than 50 subtypes. Due to the similarities between pestiviruses and hepaciviruses, combined with the poor ability of hepaciviruses to grow efficiently in cell culture, bovine viral diarrhea virus (BVDV) is often used as a sunogate to study the HCV virus.
The genetic organization of pestiviruses and hepaciviruses is very similar. These positive stranded RNA viruses possess a single large open reading frame (ORF) encoding all the viral proteins necessary for virus replication. These proteins are expressed as a polyprotein that is co- and post-translationally processed by both cellular and virus-encoded proteinases to yield the mature viral proteins. The viral proteins responsible for the replication of the viral genome RNA are located within approximately the carboxy-terminal. Two-thirds of the ORF are termed nonstructural (NS) proteins. The genetic organization and polyprotein processing of the nonstructural protein portion of the ORF for pestiviruses and hepaciviruses is very similar. For both the pestiviruses and hepaciviruses, the mature nonstructural (NS) proteins, in sequential order from the amino-terminus of the nonstructural protein coding region to the carboxy-terminus of the ORF, consist of p7, NS25 NS3, NS4A, NS4B, NS5A, and NS5B.
The NS proteins of pestiviruses and hepaciviruses share sequence domains that are characteristic of specific protein functions. For example, the NS3 proteins of viruses in both groups possess amino acid sequence motifs characteristic of serine proteinases and of helicases (Gorbalenya et al. (1988) Nature 333:22; Bazan and Fletterick (1989) Virology 111 -.631-639; Gorbalenya et al. (1989) Nucleic Acid Res. 17.3889-3897). Similarly, the NS5B proteins of pestiviruses and hepaciviruses have the motifs characteristic of RNA- directed RNA polymerases (Koonin, EN. and Dolja, VN. (1993) Crit. Rev. Biochem.
Molec. Biol 28:375-430).
The actual roles and functions of the ΝS proteins of pestiviruses and hepaciviruses in the lifecycle of the viruses are directly analogous. In both cases, the ΝS3 serine proteinase is responsible for all proteolytic processing of polyprotein precursors downstream of its position in the ORF (Wiskerchen and Collett (1991) Virology 184:341-
350; Bartenschlager et al. (1993) J. Virol. 67:3835-3844; Eckart et al. (1993) Biochem.
Biophys. Res. Comm. 192:399-406; Grakoui et al. (1993) J. Virol. 67:2832-2843; Grakoui et al. (1993) Proc. Natl. Acad. Sci. USA 90:10583-10587; Hijikata et al. (1993) J. Virol. 67:4665-4675; Tome et al. (1993) J. Virol. 67:4017-4026). The NS4A protein, in both cases, acts as a cofactor with the NS3 serine protease (Bartenschlager et al. (1994) J. Virol.
68:5045-5055; Failla et al. (1994) J. Virol. 68: 3753-3760; Lin et al. (1994) 68:8147-8157;
Xu et al. (1997) J. Virol. 71:5312-5322). The NS3 protein of both viruses also functions as a helicase (Kim et al. (1995) Biochem. Biophys. Res. Comm. 215: 160-166; Jin and Peterson (1995) Arch. Biochem. Biophys., 323:47-53; Wanener and Collett (1995) J. Virol. 69:1720-
1726). Finally, the NS5B proteins of pestiviruses and hepaciviruses have the predicted
RNA-directed RNA polymerases activity (Behrens et al.(1996) EMBO J. 15:12-22;
Lchmannet al.(1997) J. Virol. 71:8416-8428; Yuan et al.(1997) Biochem. Biophys. Res.
Comm. 232:231-235; Hagedorn, PCT WO 97/12033; Zhong et al.(1998) J. Virol. 72.9365- 9369).
Hepatitis C Virus
The hepatitis C virus (HCV) is the leading cause of chronic liver disease worldwide. (Boyer, N. et al. J. Hepatol 32:98-112, 2000). HCV causes a slow growing viral infection and is the major cause of cinhosis and hepatocellular carcinoma (Di Besceglie, A. M. and
Bacon, B. R.5 Scientific American, Oct.: 80-85, (1999); Boyer, N. et al. J. Hepatol. 32:98- 112, 2000). An estimated 170 million persons are infected with HCV worldwide. (Boyer, N. et al. J. Hepatol. 32:98-112, 2000). Cinhosis caused by chronic hepatitis C infection accounts for 8,000-12,000 deaths per year in the United States, and HCV infection is the leading indication for liver transplantation.
HCV is known to cause at least 80% of posttransfusion hepatitis and a substantial proportion of sporadic acute hepatitis. Preliminary evidence also implicates HCV in many cases of "idiopathic" chronic hepatitis, "cryptogenic" cinhosis, and probably hepatocellular carcinoma unrelated to other hepatitis viruses, such as Hepatitis B Virus (HBV). A small proportion of healthy persons appear to be chronic HCV earners, varying with geography and other epidemiological factors. The numbers may substantially exceed those for HBV, though information is still preliminary; how many of these persons have subclinical chronic liver disease is unclear. (The Merck Manual, ch. 69, p. 901, 16th ed., (1992)). HCV is an enveloped virus containing a positive-sense single-stranded RNA genome of approximately 9.4kb. The viral genome consists of a 5' untranslated region (UTR), a long open reading frame encoding a polyprotein precursor of approximately 3011 amino acids, and a short 3' UTR. The 5' UTR is the most highly conserved part of the HCV genome and is important for the initiation and control of polyprotein translation. Translation of the HCV genome is initiated by a cap-independent mechanism known as internal ribosome entry. This mechanism involves the binding of ribosomes to an RNA sequence known as the internal ribosome entry site (IRES). An RNA pseudoknot structure has recently been determined to be an essential structural element of the HCV IRES. Viral structural proteins include a nucleocapsid core protein (C) and two envelope glycoproteins, El and E2. HCV also encodes two proteinases, a zinc-dependent metalloproteinase encoded by the NS2-NS3 region and a serine proteinase encoded in the NS3 region. These proteinases are required for cleavage of specific regions of the precursor polyprotein into mature peptides. The carboxyl half of nonstructural protein 5, NS5B, contains the RNA- dependent RNA polymerase. The function of the remaining nonstructural proteins, NS4A and NS4B, and that of NS5A (the amino-terminal half of nonstructural protein 5) remain unknown.
A significant focus of cunent antiviral research is directed to the development of improved methods of treatment of chronic HCV infections in humans (Di Besceglie, A. M. and Bacon, B. R., Scientific American, Oct.: 80-85, (1999)).
Treatment of HCV Infection with Interferon
Interferons (IFNs) have been commercially available for the treatment of chronic hepatitis for nearly a decade. IFNs are glycoproteins produced by immune cells in response to viral infection. IFNs inhibit replication of a number of viruses, including HCV, and when used as the sole treatment for hepatitis C infection, IFN can in certain cases suppress serum HCV-RNA to undetectable levels. Additionally, IFN can normalize serum amino transferase levels. Unfortunately, the effect of JJFN is temporary and a sustained response occurs in only 8%-9% of patients chronically infected with HCV (Gary L. Davis. Gastroenterology 118:S104-S114, 2000). Most patients, however, have difficulty tolerating interferon treatment, which causes severe flu-like symptoms, weight loss, and lack of energy and stamina.
A number of patents disclose Flaviviridae, including HCV, treatments, using interferon-based therapies. For example, U.S. Patent No. 5,980,884 to Blatt et al. discloses methods for retreatment of patients afflicted with HCV using consensus interferon. U.S.
Patent No. 5,942,223 to Bazer et al. discloses an anti-HCV therapy using ovine or bovine interferon-tau. U.S. Patent No. 5,928,636 to Alber et al. discloses the combination therapy of interleukin- 12 and interferon alpha for the treatment of infectious diseases including HCV. U.S. Patent No. 5,849,696 to Chretien et al. discloses the use of thymosins, alone or in combination with interferon, for treating HCV. U.S. Patent No. 5,830,455 to Valtuena et al. discloses a combination HCV therapy employing interferon and a free radical scavenger. U.S. Patent No. 5,738,845 to Imakawa discloses the use of human interferon tau proteins for treating HCV. Other interferon-based treatments for HCV are disclosed in U.S. Patent No. 5,676,942 to Testa et al, U.S. Patent No. 5,372,808 to Blatt et al, and U.S. Patent No. 5,849,696. A number of patents also disclose pegylated forms of interferon, such as, U.S.
Patent Nos. 5,747,646, 5,792,834 and 5,834,594 to Hoffmann-La Roche Inc; PCT Publication No. WO 99/32139 and WO 99/32140 to Enzon; WO 95/13090 and US Patent Nos. 5,738,846 and 5,711,944 to Schering; and U.S. Patent No. 5,908,621 to Glue etal.
Interferon alpha-2a and interferon alpha-2b are cunently approved as monotherapy for the treatment of HCV. ROFERON®-A (Roche) is the recombinant form of interferon alpha-2a. PEGASYS® (Roche) is the pegylated (i.e. polyethylene glycol modified) form of interferon alpha-2a. INTRON®A (Schering Corporation) is the recombinant form of Interferon alpha-2b, and PEG-INTRON® (Schering Corporation) is the pegylated form of interferon alpha-2b. Other forms of interferon alpha, as well as interferon beta, gamma, tau and omega are currently in clinical development for the treatment of HCV. For example, INFERGEN (interferon alphacon-1) by InterMune, OMNIFERON (natural interferon) by Viragen, ALBUFERON by Human Genome Sciences, REBIF (interferon beta- la) by Ares-Serono, Omega Interferon by BioMedicine, Oral Interferon Alpha by Amarillo Biosciences, and interferon gamma, interferon tau, and interferon gamma- lb by InterMune are in development. Ribivarin
Ribavirin (l-β-D-ribofuranosyl-l-l,2,4-triazole-3-carboxamide) is a synthetic, non- interferon-inducing, broad spectrum antiviral nucleoside analog sold under the trade name, Virazole (The Merck Index, 11th edition, Editor: Budavari, S., Merck & Co., Inc., Rahway, NJ, pl304, 1989). United States Patent No. 3,798,209 and RE29,835 disclose and claim ribavirin. Ribavirin is structurally similar to guanosine, and has in vitro activity against several DNA and RNA viruses including Flaviviridae (Gary L. Davis. Gastroenterology 118:S104-S114, 2000).
Ribavirin reduces serum amino transferase levels to normal in 40% of patients, but it does not lower serum levels of HCV-RNA (Gary L. Davis. Gastroenterology 118:S104-
S114, 2000). Thus, ribavirin alone is not effective in reducing viral RNA levels. Additionally, ribavirin has significant toxicity and is known to induce anemia.
Ribavirin is not approved fro monotherapy against HCV. It has been approved in combination with interferon alpha-2a or interferon alpha-2b for the treatment of HCV.
Combination of Interferon and Ribavirin
The current standard of care for chronic hepatitis C is combination therapy with an alpha interferon and ribavirin. The combination of interferon and ribavirin for the treatment of HCV infection has been reported to be effective in the treatment of interferon naive patients (Battaglia, A.M. et al, Ann. Pharmacother. 34:487-494, 2000), as well as for treatment of patients when histological disease is present (Berenguer, M. et al. Antivir. Ther. 3(Suppl. 3): 125-136, 1998). Studies have show that more patients with hepatitis C respond to pegylated interferon-alpha/ribavirin combination therapy than to combination therapy with unpegylated interferon alpha. However, as with monotherapy, significant side effects develop during combination therapy, including hemolysis, flu-like symptoms, anemia, and fatigue. (Gary L. Davis. Gastroenterology 118:S104-S114, 2000).
Combination therapy with PEG-INTRON® (peginterferon alpha-2b) and REBETOL® (Ribavirin, USP) Capsules is available from Schering Corporation. REBETOL® (Schering Corporation) has also been approved in combination with INTRON® A (Interferon alpha-2b, recombinant, Schering Corporation). Roche's
PEGASYS® (pegylated interferon alpha-2a) and COPEGUS® (ribavirin) are also approved for the treatment of HCV.
PCT Publication Nos. WO 99/59621, WO 00/37110, WO 01/81359, WO 02/32414 and WO 03/024461 by Schering Corporation disclose the use of pegylated interferon alpha and ribavirin combination therapy for the treatment of HCV. PCT Publication Nos. WO 99/15194, WO 99/64016, and WO 00/24355 by Hoffmann-La Roche Inc also disclose the use of pegylated interferon alpha and ribavirin combination therapy for the treatment of HCV.
Additional Methods to Treat Flaviviridae Infections
The development of new antiviral agents for flaviviridae infections, especially hepatitis C, is cunently underway. Specific inhibitors of HCV-derived enzymes such as protease, helicase, and polymerase inhibitors are being developed. Drugs that inhibit other steps in HCV replication are also in development, for example, drugs that block production of HCV antigens from the RNA (IRES inhibitors), drugs that prevent the normal processing of HCV proteins (inhibitors of glycosylation), drugs that block entry of HCV into cells (by blocking its receptor) and nonspecific cytoprotective agents that block cell injury caused by the virus infection. Further, molecular approaches are also being developed to treat hepatitis C, for example, ribozymes, which are enzymes that break down specific viral RNA molecules, and antisense oligonucleotides, which are small complementary segments of DNA that bind to viral RNA and inhibit viral replication, are under investigation. A number of HCV treatments are reviewed by Bymock et al. in Antiviral Chemistry & Chemotherapy, 11:2; 79-95 (2000) and De Francesco et al. in Antiviral Research, 58: 1-16 (2003). Examples of classes of drugs that are being developed to treat Flaviviridae infections include:
(1) Protease inhibitors
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) are being investigated.
Non-substrate-based NS3 protease 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 /rørα-phenoxyphenyl group are also being investigated.
Sch 68631, a phenanthrenequinone, is an HCV protease inhibitor (Chu M. et al, Tetrahedron Letters 37:7229-7232, 1996). In another example by the same authors, Sch 351633, isolated from the fungus Penicillium griseofulvum, was identified as a protease inhibitor (Chu M. et al, Bioorganic and Medicinal Chemistry Letters 9:1949-1952). Nanomolar potency against the HCV NS3 protease enzyme has been achieved by the design of selective inhibitors based on the macromolecule eglin c. Eglin c, isolated from leech, is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, α- chymotrypsin, chymase and subtilisin. Qasim M.A. et al, Biochemistry 36:1598-1607,
1997.
Several U.S. patents disclose protease inhibitors for the treatment of HCV. For example, U.S. Patent No. 6,004,933 to Spruce et al. discloses a class of cysteine protease inhibitors for inhibiting HCV endopeptidase 2. U.S. Patent No. 5,990,276 to Zhang et al. discloses synthetic inhibitors of hepatitis C virus NS3 protease. The inhibitor is a subsequence of a substrate of the NS3 protease or a substrate of the NS4A cofactor. The use of restriction enzymes to treat HCV is disclosed in U.S. Patent No. 5,538,865 to Reyes et al. Peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/008251 to Corvas International, Inc, and WO 02/08187 and WO 02/008256 to Schering Corporation. HCV inhibitor tripeptides are disclosed in US Patent Nos. 6,534,523, 6,410,531, and
6,420,380 to Boehringer Ingelheim and WO 02/060926 to Bristol Myers Squibb. Diaryl peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/48172 to Schering Corporation. Imidazoleidinones as NS3 serine protease inhibitors of HCV are disclosed in WO 02/08198 to Schering Corporation and WO 02/48157 to Bristol Myers Squibb. WO 98/17679 to Vertex Pharmaceuticals and WO 02/48116 to Bristol Myers
Squibb also disclose HCV protease inhibitors.
(2) 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;
(3) Thiazolidines and benzanilides identified in Kakiuchi N. et al. J. EBS Letters 421, 217-220; TakeshitaN. et al. Analytical Biochemistry, 1997, 247, 242-246; (4) 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 griseofulvum, which demonstrates activity in a scintillation proximity assay (Chu M. et al, Bioorganic and Medicinal Chemistry Letters 9, 1949-1952);
(5) Helicase inhibitors (for example 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, Piper idine derivatives, pharmaceutical compositions thereof and their use in the treatment of hepatitis C, PCT WO 97/36554);
(6) Nucleotide polymerase inhibitors and 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);
(7) Antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5' non-coding region (NCR) of the virus (Alt M. et al,
Hepatology, 1995, 22, 707-717), or nucleotides 326-348 comprising the 3' end of the NCR and nucleotides 371-388 located in the core coding region of the HCV RNA (Alt M. et al, Archives of Virology, 1997, 142, 589-599; Galderisi U. et al, Journal of Cellular Physiology, 1999, 181, 251-257); (8) 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);
(9) Ribozymes, such as nuclease-resistant ribozymes (Maccjak, D. J. et al, Hepatology 1999, 30, abstract 995) and those disclosed in U.S. Patent No. 6,043,077 to Barber et al, and U.S. Patent Nos. 5,869,253 and 5,610,054 to
Draper et al; and
(10) Nucleoside analogs have also been developed for the treatment of Flaviviridae infections.
Idenix Pharmaceuticals discloses the use of branched nucleosides in the treatment of flaviviruses (including HCV) and pestiviruses in International Publication Nos. WO
01/90121 and WO 01/92282. Specifically, a method for the treatment of hepatitis C infection (and flaviviruses and pestiviruses) in humans and other host animals is disclosed in the Idenix publications that includes administering an effective amount of a biologically active 1', 2', 3' or 4'-branched β-D or β-L nucleosides or a pharmaceutically acceptable salt or derivative thereof, administered either alone or in combination with another antiviral agent, optionally in a pharmaceutically acceptable canier.
Other patent applications disclosing the use of certain nucleoside analogs to treat hepatitis C virus include: PCT/CAOO/01316 (WO 01/32153; filed November 3, 2000) and PCT/CA01/00197 (WO 01/60315; filed February 19, 2001) filed by BioChem Pharma, Inc.
(now Shire Biochem, Inc.); PCT/US02/01531 (WO 02/057425; filed January 18, 2002) and PCT/US02/03086 (WO 02/057287; filed January 18, 2002) filed by Merck & Co., Inc., PCT/EPOl/09633 (WO 02/18404; published August 21, 2001) filed by Roche, and PCT Publication Nos. WO 01/79246 (filed April 13, 2001), WO 02/32920 (filed October 18, 2001) and WO 02/48165 by Pharmasset, Ltd.
PCT Publication No. WO 99/43691 to Emory University, entitled "2'- Fluoronucleosides" discloses the use of certain 2'-fluoronucleosides to treat HCV.
Eldrup et al. (Oral Session V, Hepatitis C Virus, Flaviviridae; 16th International Conference on Antiviral Research (April 27, 2003, Savannah, Ga.)) described the structure activity relationship of 2 ' -modified nucleosides for inhibition of HCV.
Bhat et al. (Oral Session V, Hepatitis C Virus, Flaviviridae, 2003 (Oral Session V, Hepatitis C Virus, Flaviviridae; 16th International Conference on Antiviral Research (April 27, 2003, Savannah, Ga.); p A75) describe the synthesis and pharmacokinetic properties of nucleoside analogues as possible inhibitors of HCV RNA replication. The authors report that 2 '-modified nucleosides demonstrate potent inhibitory activity in cell-based replicon assays.
Olsen et al. (Oral Session V, Hepatitis C Virus, Flaviviridae; 16th International Conference on Antiviral Research (April 27, 2003, Savannah, Ga.) p A76) also described the effects of the 2 '-modified nucleosides on HCV RNA replication. (11) Other miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S.
Patent 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), benzimidazoles (U.S. Pat. No. 5,891,874 to Colacino et al), plant extracts (U.S. Patent No. 5,837,257 to Tsai et al, U.S. Patent No. 5,725,859 to Omer et al, and U.S. Patent No. 6,056,961), and piperidenes (U.S. Patent No. 5,830,905 to Diana et al). (12) Other compounds cunently in preclinical or clinical development for treatment of hepatitis C virus include: Interleukin- 10 by Schering-Plough, IP-501 by Intemeuron, Merimebodib (VX-497) by Vertex, AMANTADINE® (Symmetrel) by Endo Labs Solvay, HEPTAZYME® by RPI, IDN-6556 by Idun Pharma., XTL-002 by XTL., HCV/MF59 by Chiron, CIVACIR® (Hepatitis C Immune Globulin) by NABI,
LEVOVIRIN® by ICN/Ribapharm, VIRAMIDINE® by ICN/Ribapharm, ZADAXIN® (thymosin alpha- 1) by Sci Clone, thymosin plus pegylated interferon by Sci Clone, CEPLENE® (histamine dihydrochloride) by Maxim, VX 950 / LY 570310 by Vertex/Eli Lilly, ISIS 14803 by Isis Pharmaceutical/Elan, IDN-6556 by Idun Pharmaceuticals, Inc., JTK 003 by AKROS Pharma, BILN-2061 by Boehringer Ingelheim, CellCept
(mycophenolate mofetil) by Roche, T67, a β-tubulin inhibitor, by Tularik, a therapeutic vaccine directed to E2 by Innogenetics, FK788 by Fujisawa Healthcare, Inc., IdB 1016 (Siliphos, oral silybin-phosphatdylcholine phytosome), RNA replication inhibitors (VP50406) by ViroPharma/Wyeth, therapeutic vaccine by Intercell, therapeutic vaccine by Epimmune/Genencor, IRES inhibitor by Anadys, ANA 245 and ANA 246 by Anadys, immunotherapy (Therapore) by Avant, protease inhibitor by Corvas/SChering, helicase inhibitor by Vertex, fusion inhibitor by Trimeris, T cell therapy by CellExSys, polymerase inhibitor by Biocryst, targeted RNA chemistry by PTC Therapeutics, Dication by Immtech, Int., protease inhibitor by Agouron, protease inhibitor by Chiron/Medivir, antisense therapy by AVI BioPharma, antisense therapy by Hybridon, hemopurifier by Aethlon Medical, therapeutic vaccine by Merix, protease inhibitor by Bristol-Myers Squibb/Axys, Chron- VacC, a therapeutic vaccine, by Tripep, UT 23 IB by United Therapeutics, protease, helicase and polymerase inhibitors by Genelabs Technologies, IRES inhibitors by Immusol, R803 by Rigel Pharmaceuticals, INFERGEN® (interferon alphacon-1) by InterMune, OMNIFERON® (natural interferon) by Viragen, ALBUFERON® by Human Genome
Sciences, REBIF® (interferon beta-la) by Ares-Serono, Omega Interferon by BioMedicine, Oral Interferon Alpha by Amarillo Biosciences, interferon gamma, interferon tau, and Interferon gamma- lb by InterMune.
Nucleoside prodrugs have been previously described for the treatment of other forms of hepatitis. WO 00/09531 (filed August 10, 1999) and WO 01/96353 (filed June 15, 2001) to Idenix Pharmaceuticals discloses 2'-deoxy-β-L-nucleosides and their 3 '-prodrugs for the treatment of HBV. U.S. Patent No. 4,957,924 to Beauchamp discloses various therapeutic esters of acyclovir. In light of the fact that HCV infection has reached epidemic levels worldwide, and has tragic effects on the infected patient, there remains a strong need to provide new effective pharmaceutical agents to treat hepatitis C that have low toxicity to the host.
Further, given the rising threat of other flaviviridae infections, there remains a strong need to provide new effective pharmaceutical agents that have low toxicity to the host.
Therefore, it is an object of the present invention to provide a compound, method and composition for the treatment of a host infected with hepatitis C virus.
It is another object of the present invention to provide a method and composition generally for the treatment of patients infected with pestiviruses, flaviviruses, or hepaciviruses.
SUMMARY OF THE INVENTION
2' and 3'-prodrugs of 1', 2', 3' or 4'-branched β-D or β-L nucleosides, or their pharmaceutically acceptable salts or pharmaceutically acceptable formulations containing these compounds are useful in the prevention and treatment of Flaviviridae infections and other related conditions such as anti- Flaviviridae antibody positive and Flaviviridae - positive conditions, chronic liver inflammation caused by HCV, cinhosis, acute hepatitis, fulminant hepatitis, chronic persistent hepatitis, and fatigue. These compounds or formulations can also be used prophylactically to prevent or retard the progression of clinical illness in individuals who are anti-Flaviviridae antibody or Flaviviridae-antigen positive or who have been exposed to a Flaviviridae.
A method for the treatment of a Flaviviridae viral infection in a host, including a human, is also disclosed that includes administering an effective amount of a 2' or 3'- prodrug of a biologically active 1', , 3' or 4 '-branched β-D or β-L nucleoside or a pharmaceutically acceptable salt thereof, administered either alone or in combination or alternation with another anti-Flaviviridae agent, optionally in a pharmaceutically acceptable canier. The term 2 '-prodrug, as used herein, refers to a 1', 2', 3' or 4 '-branched β-D or β-L nucleoside that has a biologically cleavable moiety at the 2 '-position, including, but not limited to acyl, and in one embodiment, a natural or synthetic D or L amino acid, preferably an L-amino acid. The term 3 '-prodrug, as used herein, refers to a 1', 2', 3' or 4 '-branched β-D or β-L nucleoside that has a biologically cleavable moiety at the 3 '-position, including, but not limited to acyl, and in one embodiment, a natural or synthetic D or L amino acid, preferably an L-amino acid. Pharmaceutically acceptable salts include tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, α-ketoglutarate, and α- glycerophosphate, formate, fumarate, propionate, glycolate, lactate, pyruvate, oxalate, maleate, salicyate, sulfate, sulfonate, nitrate, bicarbonate, hydrobromate, hydrobromide, hydroiodide, carbonate, and phosphoric acid salts. A particularly preferred embodiment is the mono or dihydrochloride salt.
In one embodiment, the 1', 2', 3' or 4'-branched β-D or β-L nucleoside includes biologically cleavable moieties at the 2' and/or 5' positions. Prefened moieties are natural or synthetic D or L amino acid esters, including D or L-valyl, though preferably L-amino acid esters, such as L-valyl, and alkyl esters including acetyl. Therefore, this invention specifically includes 2'-D or L-amino acid ester and 2',5'-D or L-diamino acid ester, preferably L-amino acid ester, of V, 2', 3' or 4'-branched β-D or β-L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC50 of less than 15 micromolar, and even more preferably less than 10 micromolar; 2 '-(alkyl or aryl) ester or 2',5'-di(alkyl or aryl) ester of 1', 2', 3' or 4'-branched β-D or β-L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC50 of less than 10 or 15 micromolar; and prodrugs of 2',5'-diesters of 1', 2', 3' or 4'-branched β- D or β-L nucleosides wherein (i) the 2' ester is a natural or synthetic D or L-amino acid ester, though preferably an L-amino acid ester, and the 5 '-ester is an alkyl or aryl ester; (ii) both esters are independently natural or synthetic D or L-amino acid ester, though preferably both are L-amino acid esters; (iii) both esters are independently alkyl or aryl esters; and (iv) the 2' ester is independently an alkyl or aryl ester and the 5 '-ester is a natural or synthetic D or L-amino acid ester, though preferably an L-amino acid ester, wherein the parent drug optionally has an EC50 of less than 10 or 15 micromolar. Examples of prodrugs falling within the invention are 2'-D or L-valine ester of β-D-
2',6-dimethyl-cytidine; 2'-L-valine ester of β-D-2',6-dimethyl-thymidine; 2'-L-valine ester of β-D-2',8-dimethyl-adenosine; 2'-L-valine ester of β-D-2',8-dimethyl-guanosine; 2'-L- valine ester of β-D-2',6-dimethyl-5-fluorocytidine; 2'-L-valine ester of β-D-2',6-dimethyl- uridine; 2'-acetyl ester of β-D-2',6-dimethyl-cytidine; 2'-acetyl ester of β-D-2',6-dimethyl- thymidine; 2'-acetyl ester of β-D-2',8-dimethyl-adenosine; 2'-acetyl ester of β-D-2',8- dimethyl-guanosine; 2'-acetyl ester of β-D-2',6-dimethyl-5-fluoro-cytidine; and 2'-esters of β-D-2',6-dimethyl-(cytidine, 5-fluorocytidine, uridine or thymidine) or 2'-esters of β-D- 2',8-dimethyl-(guanosine, adenosine or inosine) wherein (i) the 2' ester is an amino acid ester; or (ii) the 2' ester is an alkyl or aryl ester.
Additional examples of prodrugs falling within the invention are 2',5'-L-divaline ester of β-D-2',6-dimethyl-cytidine (dival-2',6-diMe-L-dC); 2',5'-L-divaline ester of β-D- 2',6-dimethyl-thymidine; 2',5'-L-divaline ester of β-D-2',8-dirnethyl-adenosine; 2',5'-L- divaline ester of β-D-2',8-dimethyl-guanosine; 2',5'-L-divaline ester of β-D-2',6-dimethyl- 5-fluoro-cytidine; 2',5'-L-divaline ester of β-D-2',6-dimethyl-uridine; 2',5'-diacetyl ester of β-D-2',6-dimethyl-cytidine; 2',5'-diacetyl ester of β-D-2',6-dimethyl-thymidine; 2',5'- diacetyl ester of β-D-2',8-dimethyl-adenosine; 2',5'-diacetyl ester of β-D-2',8-dimethyl- guanosine; 2',5'-diacetyl ester of β-D-2',6-dimethyl-5-fluoro-cytidine; and 2',5 '-diesters of β-D-2',6-dimethyl-(cytidine, 5-fluorocytidine, uridine or thymidine) or 2',5'-diesters of β- D-2',8-dimethyl-(guanosine, adenosine or inosine) wherein (i) the 2' ester is an amino acid ester and the 5 '-ester is an alkyl or aryl ester; (ii) both esters are amino acid esters; (iii) both esters are independently alkyl or aryl esters; or (iv) the 2' ester is an alkyl or aryl ester and the 5 '-ester is an amino acid ester.
In another embodiment, the 1', 2', 3' or 4'-branched β-D or β-L nucleoside 3'- prodrug includes biologically cleavable moieties at the 3' and/or 5' positions. Preferred moieties are natural or synthetic D or L amino acid esters, such as valyl, though preferably L-amino acids, such as L-valyl, and alkyl esters including acetyl. Therefore, this invention specifically includes 3 '-L-amino acid ester and 3',5'-L-diamino acid ester of 1', 2', 3' or 4'- branched β-D or β-L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC50 of less than 15 micromolar, and even more preferably less than 10 micromolar; 3'-(alkyl or aryl) ester or 3',5'-L-di(alkyl or aryl) ester of 1', 2', 3' or 4 '-branched β-D or β-L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC5o of less than 10 or 15 micromolar; and prodrugs of
3 ',5 '-diesters of V, 2', 3' or 4'-branched β-D or β-L nucleosides wherein (i) the 3' ester is a natural or synthetic D or L amino acid ester and the 5 '-ester is an alkyl or aryl ester; (ii) both esters are natural or synthetic D or L-amino acid esters; (iii) both esters are independently alkyl or aryl esters; and (iv) the 3' ester is independently an alkyl or aryl ester and the 5 '-ester is a natural or synthetic D or L-amino acid ester, wherein the parent drug optionally has an EC50 of less than 10 or 15 micromolar.
Examples of prodrugs falling within the invention are 3'-L-valine ester of β-D-2',6- dimethyl-cytidine; 3'-L-valine ester of β-D-2',6-dimethyl-thymidine; 3 '-L- valine ester of β- D-2',8-dimethyl-adenosine; 3'-L-valine ester of β-D-2',8-dimethyl-guanosine; 3'-L-valine ester of β-D-2',6-dimethyl-5-fluorocytidine; 3'-L-valine ester of β-D-2',6-dimethyl-uridine; 3'-acetyl ester of β-D-2',6-dimethyl-cytidine; 3'-acetyl ester of β-D-2',6-dimethyl- thymidine; 3'-acetyl ester of β-D-2',8-dimethyl-adenosine; 3'-acetyl ester of β-D-2',8- dimethyl-guanosine; 3 '-acetyl ester of β-D-2',6-dimethyl-5-fluoro-cytidine; and 3 '-esters of β-D-2',6-dimethyl-(cytidine, 5-fluorocytidine, uridine or thymidine) or 3 '-esters of β-D- 2',8-dimethyl-(guanosine, adenosine or inosine) wherein (i) the 3' ester is an amino acid ester; or (ii) the 3' ester is an alkyl or aryl ester.
Additional examples of prodrugs falling within the invention are 3',5'-L-divaline ester of β-D-2',6-dimethyl-cytidine (dival-2',6-diMe-L-dC); 3',5'-L-divaline ester of β-D-
2',6-dimethyl-thymidine; 3',5'-L-divaline ester of β-D-2',8-dimethyl-adenosine; 3',5'-L- divaline ester of β-D-2',8-dimethyl-guanosine; 3',5'-L-divaline ester of β-D-2',6-dimethyl- 5-fluoro-cytidine; 3',5'-L-divaline ester of β-D-2',6-dimethyl-uridine; 3 ',5 '-diacetyl ester of β-D-2',6-dimethyl-cytidine; 3 ',5 '-diacetyl ester of β-D-2',6-dimethyl-thymidine; 3',5'- diacetyl ester of β-D-2',8-dimethyl-adenosine; 3 ',5 '-diacetyl ester of β-D-2',8-dimethyl- guanosine; 3 ',5 '-diacetyl ester of β-D-2',6-dimethyl-5-fluoro-cytidine; and 3 ',5 '-diesters of β-D-2',6-dimethyl-(cytidine, 5-fluorocytidine, uridine or thymidine) or 3',5'-diesters of β- D-2',8-dimethyl-(guanosine, adenosine or inosine) wherein (i) the 3' ester is an amino acid ester and the 5 '-ester is an alkyl or aryl ester; (ii) both esters are amino acid esters; (iii) both esters are independently alkyl or aryl esters; or (iv) the 3' ester is an alkyl or aryl ester and the 5 '-ester is an amino acid ester.
In another embodiment, the prodrug of V, 2', 3' or 4 '-branched β-D or β-L nucleoside includes biologically cleavable moieties at the 2', 3' and/or 5' positions. Prefened moieties are natural or synthetic D or L amino acid esters, including D or L-valyl, though preferably L-amino acid esters, such as L-valyl, and alkyl esters including acetyl.
Therefore, this invention specifically includes 2',3'-L or D-diamino acid ester and 2',3',5'- L or D-triamino acid ester of 1', 2', 3' or 4 '-branched β-D or β-L nucleosides, prefenably L-amino acid, with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC50 of less than 15 micromolar, and even more preferably less than 10 micromolar; 2',3'-di(alkyl or aryl) ester or 2',3',5'-L-tri(alkyl or aryl) ester of 1', 2', 3' or
4'-branched β-D or β-L nucleosides with any desired purine or pyrimidine base, wherein the parent drug optionally has an EC50 of less than 10 or 15 micromolar; and prodrugs of 2',3'- diesters of 1', 2', 3' or 4'-branched β-D or β-L nucleosides wherein (i) the 2' ester is an amino acid ester and the 3 '-ester is an alkyl or aryl ester; (ii) both esters are amino acid esters; (iii) both esters are independently alkyl or aryl esters; and (iv) the 2' ester is independently an alkyl or aryl ester and the 3 '-ester is an amino acid ester, wherein the parent drug optionally has an EC50 of less than 10 or 15 micromolar. Further, 2',3',5'- triesters of 1', 2', 3' or 4'-branched β-D or β-L nucleosides wherein (i) all three esters are amino acid esters; (ii) all three esters are independently alkyl or aryl esters; (iii) the 2' ester is an amino acid ester, the 3' ester is an amino acid ester and the 5 '-ester is an alkyl or aryl ester; (iv) the 2' ester is an amino acid ester, the 3' ester is an alkyl or aryl ester and the 5'- ester is an alkyl or aryl ester; (v) the 2' ester is an alkyl or aryl ester, the 3' ester is an alkyl or aryl ester and the 5 '-ester is an amino acid ester; (vi) the 2' ester is an alkyl or aryl ester, the 3' ester is an amino acid ester and the 5 '-ester is an amino acid ester; (vii) the 2' ester is an alkyl or aryl ester, the 3' ester is an amino acid ester and the 5 '-ester is an alkyl or aryl ester; and (viii) the 2' ester is an amino acid ester, the 3' ester is an alkyl or aryl ester and the 5 '-ester is an amino acid ester; wherein the parent drug optionally has an EC50 of less than 10 or 15 micromolar.
Examples of prodrugs falling within the invention include 2',3'-L-divaline ester of β-D-2',6-dimethyl-cytidine (dival-2',6-diMe-L-dC); 2',3'-L-divaline ester of β-D-2',6- dimethyl-thymidine; 2',3'-L-divaline ester of β-D-2',8-dimethyl-adenosine; 2',3'-L-divaline ester of β-D-2',8-dimethyl-guanosine; 2',3'-L-divaline ester of β-D-2',6-dimethyl-5-fluoro- cytidine; 2',3'-L-divaline ester of β-D-2',6-dimethyl-uridine; 2',3 '-diacetyl ester of β-D-
2',6-dimethyl-cytidine; 2',3 '-diacetyl ester of β-D-2',6-dimethyl-thymidine; 2 ',3 '-diacetyl ester of β-D-2',8-dimethyl-adenosine; 2',3 '-diacetyl ester of β-D-2',8-dimethyl-guanosine; 2',3 '-diacetyl ester of β-D-2',6-dimethyl-5-fluoro-cytidine; and 2',3 '-diesters of β-D-2',6- dimethyl-(cytidine, 5-fluorocytidine, uridine or thymidine) or 2',3'-diesters of β-D-2',8- dimethyl-(guanosine, adenosine or inosine) wherein (i) the 2' ester is an amino acid ester and the 3 '-ester is an alkyl or aryl ester; (ii) both esters are amino acid esters; (iii) both esters are independently alkyl or aryl esters; or (iv) the 2' ester is an alkyl or aryl ester and the 3 '-ester is an amino acid ester.
Additional examples of prodrugs falling within the invention include 2',3',5'-L- trivaline ester of β-D-2',6-dimethyl-cytidine (trival-2',6-diMe-L-dC); 2',3',5'-L-trivaline ester of β-D-2',6-dimethyl-thymidine; 2',3',5'-L-trivaline ester of β-D-2',8-dimethyl- adenosine; 2',3',5'-L-trivaline ester of β-D-2',8-dimethyl-guanosine; 2',3',5'-L-trivaline ester of β-D-2',6-dimethyl-5-fluoro-cytidine; 2',3',5'-L-trivaline ester of β-D-2',6- dimethyl-uridine; 2',3',5'-triacetyl ester of β-D-2',6-dimethyl-cytidine; 2',3',5'-triacetyl ester of β-D-2',6-dimethyl-thymidine; 2',3',5'-triacetyl ester of β-D-2',8-dimethyl- adenosine; 2',3',5'-triacetyl ester of β-D-2',8-dimethyl-guanosine; 2',3',5'-triacetyl ester of β-D-2',6-dimethyl-5-fluoro-cytidine; and 2',3',5'-triesters of β-D-2',6-dimethyl-(cytidine, 5-fluorocytidine, uridine or thymidine) and 2',3 ',5 '-triesters of β-D-2',8-dimethyl- (guanosine, adenosine or inosine) wherein (i) all three esters are amino acid esters; (ii) all three esters are independently alkyl or aryl esters; (iii) the 2' ester is an amino acid ester, the 3' ester is an amino acid ester and the 5'-ester is an alkyl or aryl ester; (iv) the 2' ester is an amino acid ester, the 3' ester is an alkyl or aryl ester and the 5 '-ester is an alkyl or aryl ester; (v) the 2' ester is an alkyl or aryl ester, the 3' ester is an alkyl or aryl ester and the 5 '-ester is an amino acid ester; (vi) the 2' ester is an alkyl or aryl ester, the 3' ester is an amino acid ester and the 5 '-ester is an amino acid ester; (vii) the 2' ester is an alkyl or aryl ester, the 3' ester is an amino acid ester and the 5 '-ester is an alkyl or aryl ester; and (viii) the 2' ester is an amino acid ester, the 3' ester is an alkyl or aryl ester and the 5 '-ester is an amino acid ester.
In a first principal embodiment, a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (I):
Figure imgf000018_0001
or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein:
R1, R2 and R3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate); straight chained, branched or cyclic alkyl (including lower alkyl); acyl (including lower acyl); CO-alkyl, CO-aryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO- substituted aryl, 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; alkylsulfonyl, arylsulfonyl, aralkylsulfonyl, a lipid, including a phospholipid; an amino acid; and amino acid residue, a carbohydrate; a peptide; cholesterol; or other pharmaceutically acceptable leaving group which is capable of providing a compound wherein R1, R2 and/or R3 is independently H or phosphate (including mono-, di- or triphosphate), for example when administered in vivo; wherein in one embodiment R2 and/or R3 is not phosphate (including mono-, di- or triphosphate or a stabilized phosphate prodrug); wherein at least one of R2 and R3 is not hydrogen; and wherein: Y1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR4, NH2, NHR4, NR4R5, SH or SR4; X1 is a straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3,
2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO- Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; and X2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3,
2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO- Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; and wherein each Y3 is independently H, F, Cl, Br or I; each R4 and R5 is independently hydrogen, acyl (including lower acyl), alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl), lower alkyl, alkenyl, alkynyl or cycloalkyl.
In the embodiments described herein, R1, R2 and/or R3 may be a pharmaceutically acceptable leaving group which is capable of providing a compound wherein R1, R2 and/or
R3 is independently H or phosphate (including mono-, di- or triphosphate), for example when administered in vivo. In a second principal embodiment, a compound of Formula (II) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (II):
Figure imgf000020_0001
(II) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: R1, R2, R3, R4, R5, Y1, Y3, X1 and X2 are as defined above.
In a third principal embodiment, a compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric, or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV) or (V):
Figure imgf000020_0002
(III) (IV) (V) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: Base is selected from the group consisting of
Figure imgf000021_0001
(A) (B)
Figure imgf000021_0002
(C) (D) (E) (F)
Figure imgf000021_0003
(G) (H)
Figure imgf000021_0004
(I) (J) (K) (L)
Figure imgf000022_0001
(M) (N)
Figure imgf000022_0002
(O) (P) (Q) (R)
Figure imgf000022_0003
(S) (T)
Figure imgf000022_0004
(U) (V) (W) (X)
Figure imgf000023_0001
00 (Z)
Figure imgf000023_0002
(AA) (AB) (AC) (AD)
Figure imgf000023_0003
(AE) (AF)
Figure imgf000023_0004
(AG) (AH) (Al) (AJ)
Figure imgf000024_0001
(BA) (BB)
Figure imgf000024_0002
(BC) (BD) (BE) (BF)
Figure imgf000024_0003
(BG) (BH)
Figure imgf000024_0004
(Bl) (BJ) (BK) (BL)
Figure imgf000025_0001
(BM) (BN)
Figure imgf000025_0002
(BO) (BP) (BQ) (BR)
Figure imgf000025_0003
(BS) (BT)
Figure imgf000026_0001
(BU) (BV) (B ) (BX)
Figure imgf000026_0002
(BAE) (BAF)
R1, R2 R3' R4, and R5, are as defined above; each W1, W2, W3 and W4 is independently N, CH, CF, CI, CBr, CCl, CCN, CCH3, CCF3,
CCH2CH3, CC(O)NH2, CC(O)NHR4, CC(O)N(R4)2, CC(O)OH, CC(O)OR4 or CX3; each W* is independently O, S, NH or NR4; X is O, S, SO2, CH2, CH2OH, CHF, CF2, C(Y3)2, CHCN, C(CN)2, CHR4 or C(R4)2; X* is CH, CF, CY3 or CR4;
X2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO- Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH,
OR4, NH2, NHR4, NR4R5, SH or SR5; each X3 is independently a straight chained, branched or cyclic optionally substituted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl,
Br-vinyl, optionally substituted alkynyl, haloalkynyl, N3, CN, -C(O)OH, -C(O)OR4, -C(O)0(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, OH, OR4, -O(acyl), -O(lower acyl), -O(alkyl), -O(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), chloro, bromo, fluoro, iodo, NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2; each Y is independently selected from the group consisting of H, optionally substituted .. .. lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH2, CH NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R, (CH2)mCOOH,
(CH2)mCOOR, (CH2)mCONH2, (CH2)mCONR2, and (CH2)mCONHR; wherein R is H, alkyl or acyl;
Y1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR4, NH2, NHR4, NR4R5, SH or SR4; each Y2 is independently O, S, NH or NR4; each Y3 is independently H, F, Cl, Br or I; wherein for Base (B), W4 cannot be CH if W1, W2 and W3 are N; wherein for Base (E), (F), (K), (L), (W) and (X), W4 cannot be CH if W1 is N; each R6 is independently an optionally substituted alkyl (including lower alkyl), CH3,
CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3,
CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(0)NH2, -CH2C(0)NHR4, -CH2C(0)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(0)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(0)O(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR 44, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2 or cyano; each R7 is independently OH, OR2, optionally substituted alkyl (including lower alkyl),
CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3) , CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring), optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N), optionally substituted heteroaryl (preferably a 3-7 membered heteroaromatic ring having one or more O, S and/or N), -CH2C(O)OH, -CH2C(O)OR4, -CH2C(0)0(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(0)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4,
-(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(O)S(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)SH, -C(O)SR4, -C(O)S(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(0)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, -O(acyl), -O(lower acyl), -O(R4), -O(alkyl),
-0(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), NO2, NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo (fluoro, chloro, bromo, iodo); alternatively, R6 and R7 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); and each m is independently 0, 1 or 2. In a fourth principal embodiment, a compound of Formula (VI) or (VII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or (VII):
Figure imgf000029_0001
(VI) (VII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: Base, R, R1, R2, R3, R4, R5, R6, R7, Y, Y1, Y2, Y3, W1, W2, W3, W4, W*, X, X*, X1, X2, and
X3 are as defined above; wherein, in one embodiment, R8 in Formula (VI) is -OH or -NH2 only when X is carbon; and wherein; each R8 and R11 is independently hydrogen, an optionally substituted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH2C(O)OH, -CH2C(O)OR4, -CH2C(0)0(lower alkyl), -CH2C(0)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(0)NH(lower alkyl), -C(O)N(R4)2, -C(0)N(lower alkyl)* cyano, NH-acyl or N(acyl)2; each R9 and R10 are independently hydrogen, OH, OR2, optionally substituted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring), optionally substituted heterocycle (preferably 2004/002999
a 3-7 membered heterocyclic ring having one or more O, S and/or N), optionally substituted heteroaryl (preferably a 3-7 membered heteroaromatic ring having one or more O, S and/or N), -CH2C(O)OH, -CH2C(0)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(0)OR4,
-(CH2)mC(0)0(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(O)S(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)0(lower alkyl), -C(0)SH, -C(0)SR4, -C(0)S(lower alkyl), -C(0)NH2, -C(0)NHR4, -C(0)NH(lower alkyl), -C(0)N(R4)2, -C(0)N(lower alkyl)2, -O(acyl), -O(lower acyl), -O(R4), -O(alkyl),
-O(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), NO2, NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo (fluoro, chloro, bromo, iodo); each m is independently 0, 1 or 2; and alternatively, R6 and R10, R7 and R9, R8 and R7 or R9 and Rn can come together to form a bridged compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); or alternatively, R6 and R7 or R9 and R10 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N).
2004/002999
In a fifth principal embodiment, a compound of Formula (VIII), (IX) or (X), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VIII), (DQ or (X):
Figure imgf000031_0001
(VIII) (IX) (X) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: R1, R2, R3, R4, R5, X, Y3, and X* are as defined above; Base* is a purine or pyrimidine base as defined herein; each R12 is independently a substituted alkyl (including lower alkyl), CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, substituted alkenyl, haloalkenyl (but not Br-vinyl), substituted alkynyl, haloalkynyl, -CH2C(0)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(0)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)0(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)* -C(O)OH, -C(O)OR4, -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2; each R13 is independently substituted alkyl (including lower alkyl), CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, substituted alkenyl, haloalkenyl (but not Br-vinyl), substituted alkynyl, haloalkynyl, optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring), optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N), optionally substituted heteroaryl (preferably a 3-7 membered heteroaromatic ring having one or more O, S and/or N), -CH C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(O)NH2, 2004/002999
-CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(0)N(R4)2, -CH2C(0)N(lower alkyl)* -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(O)S(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(0)OR4, -C(0)SH, -C(0)SR4, -C(O)S(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)* -O(R4), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), -NHR4, -NR4R5, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), SCN, OCN, NCO or fluoro; alternatively, R12 and R13 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); and each m is independently 0, 1 or 2.
In a sixth principal embodiment, a compound of Formula (XI) or (XII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XI) or (XII):
Figure imgf000032_0001
(XI) (XII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: alternatively, Base*, is replaced with Base in Formulas (XI) and (XII); and
Base, Base*, R, R1, R2, R3, R4, R5, R12, R13, Y, Y1, Y2, Y3, W*, W1, W2, W3, W4, X, X*, X1,
X2, and X3 are as defined above; wherein, in one embodiment, R8 in Formula (XI) is -OH or -NH2 only when X is carbon; and wherein; 2004/002999
each R8 and R11 is independently hydrogen, an optionally substituted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH2C(0)OH, -CH2C(O)OR4, -CH2C(0)0(lower alkyl),
-CH2C(0)NH* -CH2C(0)NHR4, -CH2C(0)NH(lower alkyl), -CH2C(0)N(R4)* -CH2C(0)N(lower alkyl)* -(CH2)mC(0)OH, -(CH2)mC(0)OR4, -(CH2)mC(0)O(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)* -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)* cyano, NH-acyl or N(acyl)2; each R9 and R10 are independently hydrogen, OH, OR2, optionally substituted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)* CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl,
Br-vinyl, optionally substituted alkynyl, haloalkynyl, optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring), optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N), optionally substituted heteroaryl (preferably a 3-7 membered heteroaromatic ring having one or more O, S and/or N), -CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4,
-CH2C(O)S(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)* -CH2C(O)N(lower alkyl)* -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(0)0(lower alkyl), -(CH2)mC(0)SH, -(CH2)mC(0)SR4, -(CH )mC(0)S(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(0)N(R4)* -(CH2)mC(0)N(lower alkyl)* -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(0)SH, -C(0)SR4, -C(0)S(lower alkyl), -C(O)NH* -C(O)NHR4, -C(O)NH(lower alkyl), -C(0)N(R4)2, -C(0)N(lower alkyl)2, -O(acyl), -O(lower acyl), -O(R4), -O(alkyl), -O(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), N02, NH* -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)* -NH(alkenyl),
-NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo (fluoro, chloro, bromo, iodo); each m is independently 0, 1 or 2; and alternatively, R8 and R13, R9 and R13, R9 and R11 or R10 and R12 can come together to form a bridged compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); or alternatively, R12 and R13 or R9 and R10 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N).
In a particular aspect of the invention, a compound of Formula (XI) or (XII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XI) or (XII):
Figure imgf000034_0001
(XIII) (XIV) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein:
R3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; acyl; a sulfonate ester; optionally substituted alkyl sulfonyl; optionally substituted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R3 is independently H, or mono-, di- or triphosphate; X" is selected from the group consisting of one or more O, S, SO, S02, N, NH, NR and CH2 wherein any of the aforementioned may be optionally substituted and may be variably positioned so as to form a 3-7 membered ring;
R is H, alkyl or acyl; and
B indicates a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or
N);
Base is selected from the group consisting of:
Figure imgf000035_0001
(9) (h) (j) and
Figure imgf000035_0002
wherein: each R', R", R'" and R"" are independently selected from the group consisting of
H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH2, NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* C02-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH, (CH2)mCN, (CH2)mNO2 and (CH2)mCONH2; m is 0 or 1; W is C-R" orN;
T and V independently are CH or N; Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N; Q and Q2 independently are N or C-R;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof. In a second particular aspect of the invention, a compound of Formula (XV), (XVI) or (XVII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XV), (XVI) or (XVII):
Figure imgf000036_0001
(XV) (XVI) (XVII) or its pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: G and E independently are selected from the group consisting of CH3, CH2OH, CH2F,
CH2N3, CH2CN, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH* (CH2)mCONR2,
(CH2)mCONHR and N-acyl; m is O or 1;
R is H, alkyl or acyl; and R', R", R'", R"", and R3 and Base are as defined for Formula (XIII).
Alternatively, for compound of Formula (XVII), at most one of G and E can further be hydrogen.
In a third particular aspect of the invention, a compound of Formula (XVIII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a
Flaviviridae comprising administering an effective treatment amount of compound of
Formula (XVIII):
Figure imgf000036_0002
(XVIII) or its pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein M is selected from the group consisting of S, SO, and SO2; and R', R", R"\ R"", and R3 and Base are as defined for Formula (XIII).
In a fourth particular aspect of the invention, a compound of Formula (XIX), (XX), (XXI) (XXII) or (XXIII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XIX), (XX), (XXI) (XXII) or (XXIII):
Figure imgf000037_0001
(XXIII) or its pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof wherein:
A is selected from the group consisting of optionally substituted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCO- NH* (CH2)mCONR* and (CH2)mCONHR;
Y is selected from the group consisting of H, optionally substituted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH* (CH2)mCONR2, and (CH2)mCONHR; X is selected from the group consisting of -OH, optionally substituted alkyl, cycloalkyl, alkenyl, alkynyl, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aryl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N- dialkyl, NH-alkenyl, NH-alkynyl, NH-aryl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S- alkenyl, S-alkynyl, S-aryl, S-aralkyl, S-acyl, S-cycloalkyl, C02-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH2, (CH2)mCONR2, (CH2)mCONHR, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; m is O or l;
R is H, alkyl or acyl;
R3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; substituted or unsubstituted alkyl; acyl; a sulfonate ester; optionally substituted alkyl sulfonyl; optionally substituted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R3 is independently H, or mono-, di- or triphosphate; and Base is a non-natural base selected from the group of:
Figure imgf000038_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH2, NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO2-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH, (CH2)mCN, (CH2)mN02 and (CH2)mCONH2; m is O or l;
W is C-R" orN; T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
Qi and Q2 independently are N or C-R""; and
Q3, Q4, Q5 and Q6 independently are N or CH; with the proviso that in bases (g) and (i), R', R"" are not H, OH, or NH2; and Q,
T, V, Q* Q5 and Q6 are not N.
In one embodiment, the amino acid residue is of the formula C(0)C(Rπ)(R12)(NR13R14), wherein:
R11 is the side chain of an amino acid and wherein, as in proline, R11 can optionally be attached to R13 to form a ring structure; or alternatively, Rn is an alkyl, aryl, heteroaryl or heterocyclic moiety;
R12 is hydrogen, alkyl (including lower alkyl) or aryl; and
R13 and R14 are independently hydrogen, acyl (including an acyl derivative attached to R11) or alkyl (including but not limited to methyl, ethyl, propyl, and cyclopropyl). In another prefened embodiment, at least one of R2 and R3 is an amino acid residue, and is preferably L-valinyl.
The β-D- and β-L-nucleosides of this invention may inhibit Flaviviridae polymerase activity. Nucleosides can be screened for their ability to inhibit Flaviviridae 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-Flaviviridae 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 ECso). In preferred embodiments the parent of the prodrug compound exhibits an EC50 of less than 25, 15, 10, 5, or 1 micromolar. In prefened embodiments the compound exhibits an EC50 of less than 15 or 10 micromolar, when measured according to the polymerase assay described in Fenari et al, Jnl of Vir., 73:1649-1654, 1999; Ishii et al, Hepatology, 29:1227-1235,1999; Lohmann et al, Jnl. of Bio. Chem., 274:10807-10815, 1999; or Yamashita et al, Jnl. of Bio. Chem., 273:15479-15486, 1998.
In another embodiment, combination and/or alternation therapy are provided. In combination therapy, an effective dosage 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.
The invention provides combinations of at least two of the herein described prodrugs. The invention further provides at least one of the described 2' and 3'-prodrugs in combination or alternation with a second nucleoside that exhibits activity against a Flaviviridae, including but not limited to a parent drug of any of the prodrugs defined herein, i.e. β-D-2',6-dimethyl-cytidine, β-D-2',6-dimethyl-thymidine, β-D-2',8-dimethyl- adenosine, β-D-2',8-dimethyl-guanosine, β-D-2',6-dimethyl-5-fluorocytidine and/or β-D- 2',6-dimethyl-uridine. Alternatively, the 2' or 3'-prodrugs can be administered in combination or alternation with other anti-Flaviviridae agent exhibits an EC50 of less than 10 or 15 micromolar, or their prodrugs or pharmaceutically acceptable salts. Nonlimiting examples of antiviral agents that can be used in combination with the compounds disclosed herein include: 1) an interferon and/or ribavirin; (2) Substrate-based NS3 protease inhibitors;. (3) Non-substrate-based inhibitors; (4) Thiazolidine derivatives; (5) Thiazolidines and benzanilides; (6) A phenan-threnequinone; (7) NS3 inhibitors; (8) HCV helicase inhibitors; (9) polymerase inhibitors, including RNA-dependent RNA- polymerase inhibitors; (10) Antisense oligodeoxynucleotides; (11) Inhibitors of IRES- dependent translation; (12) Nuclease-resistant ribozymes; and (13) other compounds that exhibit activity against a flaviviridae. The invention further includes administering the prodrug in combination or alternation with an immune modulator or other pharmaceutically active modifer of viral replication, including a biological material such as a protein, peptide, oligonucleotide, or gamma globulin, including but not limited to interfereon, interleukin, or an antisense oligonucleotides to genes which express or regulate Flaviviridae replication.
The invention further includes administering the prodrug in combination or alternation with an immune modulator or other pharmaceutically active modifer of viral replication, including a biological material such as a protein, peptide, oligonucleotide, or gamma globulin, including but not limited to interfereon, interleukin, or an antisense oligonucleotides to genes which express or regulate Flaviviridae replication. In particular, the present invention provides the following: (a) a compound of Formula (XIII) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof;
(b) pharmaceutical compositions comprising a compound of Formula (XIII) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable canier or dileuent;
(c) pharmaceutical compositions comprising a compound of Formula (XIII) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, with one or more other effective antiviral agent, optionally with a pharmaceutically acceptable canier or dileuent; (d) pharmaceutical compositions for the treatment of a Flaviviridae infection in a host comprising a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable canier or dileuent;
(e) pharmaceutical compositions for the treatment of a Flaviviridae infection in a host comprising a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, with one or more other effective antiviral agent, optionally with a pharmaceutically acceptable carrier or dileuent;
(f) methods for the treatment of a Flaviviridae infection in a host comprising a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, optionally with a pharmaceutically acceptable canier or dileuent; (g) methods for the treatment of a Flaviviridae infection in a host comprising a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, with one or more other effective antiviral agent, optionally with a pharmaceutically acceptable canier or dileuent; (h) uses for a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, optionally with a pharmaceutically acceptable canier or dileuent, for the treatment of a Flaviviridae infection in a host; (i) uses for a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, with one or more other effective antiviral agent, optionally with a pharmaceutically acceptable carrier or dileuent, for the treatment of a Flaviviridae infection in a host;
(j) uses for a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, optionally with a pharmaceutically acceptable canier or dileuent, in the manufacture of a medicament for the treatment of a Flaviviridae infection in a host; and (k) uses for a compound of Formula (I) - (XXIII), or its pharmaceutically acceptable salt or prodrug thereof, with one or more other effective antiviral agent, optionally with a pharmaceutically acceptable canier or dileuent, in the manufacture of a medicament for the treatment of a Flaviviridae infection in a host. In an alternative embodiment, the parent nucleoside compound of any of the 2'- or 3'- prodrugs (i.e., the nucleosides without the 2'- or 3'- cleavable moieties) are provided for the treatment of a Flaviviridae infection and in particular a hepatitis C infection.
BRIEF DESCRIPTION OF THE FIGURES Figure 1 provides the structure of various non-limiting examples of nucleosides of the present invention, as well as other known nucleosides, in particular FIAU and ribavirin.
Figure 2 provides a non-limiting example of the steps involved in esterification of the V, 2', 3' or 4'-branched β-D or β-L nucleoside to obtain a 2'-prodrug. The same general procedure can be used to obtain the 3 '-prodrug by selectively protecting the 2' and 5 '-hydroxyl groups or protecting the 2', 3' and 5 '-hydroxyl groups and selectively deprotecting the 3 ' -hydroxyl.
Figure 3 provides a non-limiting example of the steps involved in esterification of the 1', 2', 3' or 4'-branched β-D or β-L nucleoside to obtain a 3 '-prodrug.
Figure 4 provides a non-limiting example of the steps involved in esterification of the 1', 2', 3' or 4'-branched β-D or β-L nucleoside to obtain a 2',3 '-prodrug.
DETAILED DESCRIPTION OF THE INVENTION
The invention as disclosed herein is a compound, a method and composition for the treatment of a Flaviviridae infection in humans and other host animals. The method includes the administration of an effective anti-Flaviviridae treatment amount of a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4'-branched β-D or β-L nucleoside as described herein or a pharmaceutically acceptable salt, derivative or prodrug thereof, optionally in a pharmaceutically acceptable canier. The compounds of this invention either possesses antiviral (i.e., anti-HCV) activity, or are metabolized to a compound that exhibits such activity. HCV is a member of the Flaviviridae family. HCV has been placed in a new monotypic genus, hepacivirus. Therefore, in one embodiment, the Flaviviridae is HCV. In an alternate embodiment, the Flaviviridae is a flavivirus or pestivirus. The 2' and/or 3 '-prodrugs of a 1', 2', 3' or 4'-branched β-D or β-L nucleoside are acyl derivates of a secondary or tertiary alcohol alpha to a secondary or tertiary carbon. Due to the steric hindrance of these prodrugs over the 5 '-prodrugs, an acyl derivative of a primary alcohol, these prodrugs differentially modulate the biological properties of the molecule in vivo. It has been discovered that the 2' and/or 3'-prodrugs of a V, , 3' or 4'- branched β-D or β-L nucleoside can provide a drug with increased half-life and improved pharmacokinetic profile.
The 2' or 3 '-prodrug in a preferred embodiment is a cleavable acyl group, and most particularly, an amino acid moiety, prepared from any naturally occurring and synthetic α, β γ or δ amino acid, including but is not limited to, glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine. In a prefened embodiment, the amino acid is in the L-configuration. Alternatively, the amino acid can be a derivative of alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, β-alanyl, β-valinyl5 β- leucinyl, β-isoleuccinyl, β-prolinyl, β-phenylalaninyl, β-tryptophanyl, β-methioninyl, β- glycinyl, β-serinyl, β-threoninyl, β-cysteinyl, β-tyrosinyl, β-asparaginyl, β-glutaminyl, β- aspartoyl, β-glutaroyl, β-lysinyl, β-argininyl or β-histidinyl. In one particular, embodiment, the moiety is a valine ester. On particularly prefened compound is the 3 '-valine ester of
2',6-dimethyl-ribo-cytidine.
The oral bio-availability of 1', 2', 3' or 4'-branched β-D or β-L nucleoside as the neutral base and the HCl salt is low in rodents and non-human primates. It has been discovered that there is significant competition of V, , 3' or 4'-branched β-D or β-L nucleoside with other nucleosides or nucleoside analogs for absorption, or transport, from the gastrointestinal tract and competition of other nucleosides or nucleoside analogs for the absorption with 1', 2', 3' or 4'-branched β-D or β-L nucleoside. In order to improve oral bioavailability and reduce the potential for drug-drug interaction, 2' and 3 '-prodrugs of 1', 2 3' or 4'-branched β-D or β-L nucleoside were obtained with higher oral bioavailability than the parent molecule and a reduced effect on the bioavailability of other nucleosides or nucleoside analogs used in combination.
The 2', 3', and/or 5'-mono, di or frivaline ester of a 1', 2', 3' or 4'-branched β-D or β-L nucleoside have higher oral bioavailability than the parent 1', 2', 3' or 4 '-branched β-D or β-L nucleoside and reduced interaction with other nucleosides or nucleoside analogs when used in combination as compared to V, 2', 3' or 4'-branched β-D or β-L nucleoside.
The 2', 3', and/or 5'-mono, di or trivaline ester of a 1', 2', 3' or 4'-branched β-D or β-L nucleoside can be converted to the parent V, 2', 3' or 4'-branched β-D or β-L nucleoside through de-esterification in the gastrointestinal mucosa, blood or liver. The 2',
3', and/or 5 '-mono, di or trivaline ester of a 1', 2', 3' or 4 '-branched β-D or β-L nucleoside can be actively transported from the gastrointestinal lumen after oral delivery into the bloodstream by an amino acid transporter function in the mucosa of the gastrointestinal tract. This accounts for the increase in oral bioavailability compared to the parent 1', 2', 3' or 4'-branched β-D or β-L nucleoside that is transported primarily by a nucleoside transporter function. There is also reduced competition for uptake of the 2', 3', and/or 5'- mono, di or trivaline ester of V, 2', 3' or 4'-branched β-D or β-L nucleoside with other nucleosides or nucleoside analogs that are transported by the nucleoside transporter function and not the amino acid transporter function. As partial de-esterification of the di or trivaline ester of 1', 2', 3' or 4 '-branched β-D or β-L nucleoside occurs prior to complete absorption, the mono or divaline ester continues to be absorbed using the amino acid transporter function. Therefore, the desired outcome of better absorption, or bioavailability, and reduced competition with other nucleosides or nucleoside analogs for uptake into the bloodstream can be maintained. In summary, the present invention includes the following features:
(a) a 2' and/or 3'-prodrug of a 1', 2', 3' or 4'-branched β-D or β-L nucleoside, as described herein, and pharmaceutically acceptable salts and compositions thereof;
(b) a 2' and/or 3 '-prodrug of a V, 2 3' or 4 '-branched β-D or β-L nucleoside as described herein, and pharmaceutically acceptable salts and compositions thereof for use in the treatment and/or prophylaxis of a Flaviviridae infection, especially in individuals diagnosed as having a Flaviviridae infection or being at risk of becoming infected by hepatitis C;
(c) a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4'-branched β-D or β-L nucleoside, or their pharmaceutically acceptable salts and compositions as described herein substantially in the absence of the opposite enantiomers of the described nucleoside, or substantially isolated from other chemical entities;
(d) processes for the preparation of a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4'-branched β-D or β-L nucleoside, as described in more detail below; (e) pharmaceutical formulations comprising a 2' and/or 3'-prodrug of a V, 2', 3' or 4'- branched β-D or β-L nucleoside or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable canier or diluent;
(f) pharmaceutical formulations comprising a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4'- branched β-D or β-L nucleoside or a pharmaceutically acceptable salt thereof together with one or more other effective anti-HCV agents, optionally in a pharmaceutically acceptable canier or diluent;
(g) pharmaceutical formulations comprising a 2' and/or 3'-prodrug of a 1', 2', 3' or 4'- branched β-D or β-L nucleoside or a pharmaceutically acceptable salt thereof together with the parent of a different a V, 2', 3' or 4'-branched β-D or β-L nucleoside, optionally in a pharmaceutically acceptable carrier or diluent;
(h) a method for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of a 2' and/or 3 '-prodrug of a
V, 2', 3' or 4 '-branched β-D or β-L nucleoside, its pharmaceutically acceptable salt or composition;
(i) a method for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4 '-branched β-D or β-L nucleoside, its pharmaceutically acceptable salt or composition in combination and/or alternation with one or more effective anti- HCV agent;
(j) a method for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4 '-branched β-D or β-L nucleoside, or its pharmaceutically acceptable salt or composition with the parent of a different a 1', 2', 3' or 4'-branched β-D or β- L nucleoside;
(k) a method for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of a 2' and/or 3 '-prodrug of a β-D-2'-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof; (1) a method for the treatment and/or prophylaxis of a host infected with Flaviviridae that includes the administration of an effective amount of the 2'-valyl or acetyl ester of β-D-2'-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof; (m) use of a 2' and/or 3'-prodrug of a 1', 2', 3' or 4'-branched β-D or β-L nucleoside, and pharmaceutically acceptable salts and compositions thereof for the treatment and/or prophylaxis of a Flaviviridae infection in a host; (n) use of a 2' and/or 3 '-prodrug of a 1', 2', 3' or 4 '-branched β-D or β-L nucleoside, its pharmaceutically acceptable salt or composition in combination and/or alternation with one or more effective anti-HCV agent for the treatment and/or prophylaxis of a
Flaviviridae infection in a host; (o) use of a 2' and/or 3'-prodrug of a V, 2', 3' or 4'-branched β-D or β-L nucleoside, or its pharmaceutically acceptable salt or composition with the parent of a different a 1 ', 2', 3' or 4'-branched β-D or β-L nucleoside for the treatment and/or prophylaxis of a Flaviviridae infection in a host; (p) use of a 2' and/or 3 '-prodrug of a β-D-2 '-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof for the treatment and/or prophylaxis of a
Flaviviridae infection in a host; (q) use of the 3'-valyl or acetyl ester of β-D-2 '-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof for the treatment and/or prophylaxis of a
Flaviviridae infection in a host; (r) use of a 2' and/or 3'-prodrug of a V, 2', 3' or 4'-branched β-D or β-L nucleoside, and pharmaceutically acceptable salts and compositions thereof in the manufacture of a medicament for treatment and/or prophylaxis of a Flaviviridae infection;
(s) use of a 2' and/or 3'-prodrug of a 1 ', 2', 3' or 4'-branched β-D or β-L nucleoside, its pharmaceutically acceptable salt or composition in combination and/or alternation with one or more effective anti-HCV agent in the manufacture of a medicament for the treatment and/or prophylaxis of a Flaviviridae infection in a host; (t) use of a 2' and/or 3'-prodrug of a V, 2', 3' or 4'-branched β-D or β-L nucleoside, or its pharmaceutically acceptable salt or composition with the parent of a different a
1', 2', 3' or 4'-branched β-D or β-L nucleoside in the manufacture of a medicament for the treatment and/or prophylaxis of a Flaviviridae infection in a host; (u) use of a 2' and/or 3 '-prodrug of a β-D-2 '-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof in the manufacture of a medicament for the treatment and/or prophylaxis of a Flaviviridae infection in a host; and (v) use of the 2'-valyl or acetyl ester of β-D-2 '-methyl-cytidine, or its pharmaceutically acceptable salt or composition thereof in the manufacture of a medicament for the treatment and/or prophylaxis of a Flaviviridae infection in a host. Flaviviridae 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. In a particular embodiment of the invention, the Flaviviridae is HCV. In an alternate embodiment of the invention, the Flaviviridae is a flavivirus or pestivirus. 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 hemonhagic 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 dianhea virus ("BVDV"), classical swine fever virus ("CSFV," also called hog cholera virus), and border disease virus ("BDV").
/. Active Compounds
In a first principal embodiment, a compound of Formula (I), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (I):
Figure imgf000047_0001
or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein:
R1, R2 and R3 are independently H, phosphate (including mono-, di- or triphosphate and a stabilized phosphate); straight chained, branched or cyclic alkyl (including lower alkyl); acyl (including lower acyl); CO-alkyl, CO-aryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO- substituted aryl, 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; alkylsulfonyl, arylsulfonyl, aralkylsulfonyl, a lipid, including a phospholipid; an amino acid; and amino acid residue, a carbohydrate; a peptide; cholesterol; or other pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 and/or R3 is independently H or phosphate (including mono-, di- or triphosphate); wherein in one embodiment R2 and/or R3 is not phosphate (including mono-, di- or triphosphate or a stabilized phosphate prodrug); wherein at least one of R2 and R3 is not hydrogen;
Y1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR4, NH* NHR4, NR4R5, SH or SR4; X1 is a straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO- Oalkoxyalkyl, CONH* CONHR4, CON(R4)* chloro, bromo, fluoro, iodo, CN, N3s OH,
OR4, NH* NHR4, NR4R5, SH or SR5;
X2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO- Oalkoxyalkyl, CONH* CONHR4, CON(R4)* chloro, bromo, fluoro, iodo, CN, N3, OH,
OR4, NH* NHR4, NR4R5, SH or SR5; and wherein each Y3 is independently H, F, Cl, Br or I; each R4 and R5 is independently hydrogen, acyl (including lower acyl), alkyl (including but not limited to methyl, ethyl, propyl and cyclopropyl), lower alkyl, alkenyl, alkynyl or cycloalkyl.
In a prefened subembodiment, a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (I) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein:
R1 is H or phosphate (preferably H);
R2 and R3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R2 and R3 is acyl or an amino acid residue;
X1 is CH3, CF3 or CH2CH3;
X2 is H orNH2; and
Y is hydrogen, bromo, chloro, fluoro, iodo, NH or OH.
In a second principal embodiment, a compound of Formula (II) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (II):
Figure imgf000049_0001
(II) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: R1, R2, R3, R4, R5, Y1, Y3, X1 and X2 are as defined above.
In a preferred subembodiment, a compound of Formula (II), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (II) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: R1 is H or phosphate (preferably H);
R2 and R3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R2 and R3 is acyl or an amino acid residue; X1 is CH3, CF3 or CH2CH3; 2004/002
X2 is H, F, Cl, Br, I or CH3; and
Y is hydrogen, bromo, chloro, fluoro, iodo, NH2 or OH.
In a third principal embodiment, a compound of Formula (III), (IN) or (V) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV), or (V):
Figure imgf000050_0001
(III) (IV) (V) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein : R1, R2, R3, R4, R5, Y, Y1 and X2 are as defined above; Base is selected from the group consisting of :
Figure imgf000050_0002
(A) (B)
Figure imgf000050_0003
(C) 0>) (E) (F)
Figure imgf000051_0001
(G) (H)
Figure imgf000051_0002
(I) (J) (K) (L)
Figure imgf000051_0003
(M) (N)
Figure imgf000051_0004
(O) (P) (Q) (R)
Figure imgf000052_0001
(S) (T)
Figure imgf000052_0002
(ϋ) (V) (W) (X)
Figure imgf000052_0003
00 (Z)
Figure imgf000052_0004
(AA) (AB) (AC) (AD)
Figure imgf000053_0001
(AE) (AF)
Figure imgf000053_0002
(AG) (AH) (Al) (AJ)
Figure imgf000053_0003
(BA) (BB)
Figure imgf000053_0004
(BC) (BD) (BE) (BF)
Figure imgf000054_0001
(BG) (BH)
Figure imgf000054_0002
(Bl) (BJ) (BK) (BL)
Figure imgf000054_0003
(BM) (BN)
Figure imgf000054_0004
(BO) (BP) (BQ) (BR)
Figure imgf000055_0001
(BS) (BT)
Figure imgf000055_0002
(BTJ) (BV) (BW) (BX)
Figure imgf000055_0003
(BY) (BZ)
Figure imgf000055_0004
(BAA) (BAB) (BAC) (BAD)
Figure imgf000056_0001
(BAE) (BAF) each W1, W2, W3 and W4 is independently N, CH, CF, CI, CBr, CCl, CCN, CCH3, CCF3, CCH2CH3, CC(O)NH2, CC(O)NHR4, CC(O)N(R4)* CC(O)OH, CC(0)OR4 or CX3; each W* is independently O, S, NH or NR4; wherein for Base (B), W4 cannot be CH if W1, W2 and W3 are N; wherein for Base (E), (F), (K), (L), (W) and (X), W4 cannot be CH if W1 is N; X is O, S, SO* CH* CH2OH, CHF, CF* C(Y3)* CHCN, C(CN)2, CHR4 or C(R4)2; X* is CH, CF, CY3 or CR4; each X3 is independently a straight chained, branched or cyclic optionally substituted alkyl
(including lower alkyl), CH3, CH2CN, CH2N3, CH2NH* CH2NHCH3, CH2N(CH3)* CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, N3, CN, -C(O)OH, -C(O)OR4, -C(0)O(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)*
-C(O)N(lower alkyl)2, OH, OR4, -O(acyl), -0(lower acyl), -O(alkyl), -0(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), chloro, bromo, fluoro, iodo, NH* -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2; each Y2 is independently O5 S, NH or NR4; each Y3 is independently H, F, Cl, Br or I; each R6 is independently an optionally substituted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3,
CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH C(O)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)* -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(0)0(lower alkyl), -(CH2)mC(O)NH* -(CH2)mC(O)NHR 44, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)NH* -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)* -C(O)N(lower alkyl)2 or cyano; each R7 is independently OH, OR2, optionally substituted alkyl (including lower alkyl),
CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring), optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N), optionally substituted heteroaryl (preferably a 3-7 membered heteroaromatic ring having one or more O, S and/or N), -CH2C(0)OH, -CH2C(0)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(0)S(lower alkyl), -CH2C(0)NH* -CH2C(0)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)* -CH2C(O)N(lower alkyl)* -(CH2)mC(0)OH, -(CH2)mC(0)OR4,
-(CH2)mC(0)O(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(0)S(lower alkyl), -(CH2)mC(O)NH* -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)SH, -C(O)SR4, -C(O)S(lower alkyl), -C(O)NH* -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)* -C(0)N(lower alkyl)2, -O(acyl), -0(lower acyl), -0(R4), -O(alkyl),
-0(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), N02, NH* -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo (fluoro, chloro, bromo, iodo); alternatively, R6 and R7 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); and each m is independently 0, 1 or 2.
In a first subembodiment, the compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, or the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV), or (V) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, wherein:
R1 is H or phosphate (preferably H);
R2 and R3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R2 and R3 is acyl or an amino acid residue;
W4 is CX3;
X3 is CH3, CF3 or CH2CH3;
R6 is alkyl; and
X is O, S, SO2 or CH* In a second subembodiment, the compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, or the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, wherein:
R1 is H or phosphate (preferably H);
R2 and R3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R2 and R3 is an amino acid residue;
W4 is CX3; X3 is CH3, CF3 or CH2CH3;
R6 is alkyl; and
X is O, S, SO2 or CH*
In a third subembodiment, the compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, or the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (III), (IV) or (V), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, wherein:
R1 is H or phosphate (preferably H); R2 and R3 are independently H, phosphate, acyl or an amino acid residue, wherein at least one of R2 and R3 is acyl or an amino acid residue;
W4 is CX3;
X3 is CH3, CF3 or CH2CH3;
R is alkyl; and X is O.
In even more prefened subembodiment, the compound of Formula (IV(a)), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, as well as the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (IV(a)):
Figure imgf000059_0001
(IV(a)) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein:
Base is as defined herein; optionally substituted with an amine or cyclopropyl (e.g., 2- amino, 2,6-diamino or cyclopropyl guanosine);
R7 is halo (F, Cl, Br or I), though preferably F;
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; 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 one embodiment R2 is not phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug); and R2 is 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; 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 one embodiment R2 is not phosphate (including monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug). In a fourth principal embodiment, a compound of Formula (VI) or (VII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, as well as a method for the freatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or (VII):
Figure imgf000060_0001
(VI) (VII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, wherein: Base, R, R1, R4, R5, R6, R7, Y, Y1, Y2, Y3, W*, W1, W2, W3, W4, X, X*, X1, X2, and X3are as defined above; wherein, in one embodiment, R8 in Formula (VI) is -OH or -NH2 only when X is carbon; and wherein; each R8 and R11 is independently hydrogen, an optionally substituted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH* CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(0)NH* -CH2C(0)NHR4, -CH2C(0)NH(lower alkyl), -CH2C(0)N(R4)*
-CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)* -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(0)OR4, -C(0)0(lower alkyl), -C(0)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)* cyano, NH-acyl or N(acyl)2; each R9 and R10 are independently hydrogen, OH, OR2, optionally substituted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)* CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C15 CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring), optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N), optionally substituted heteroaryl (preferably a 3-7 membered heteroaromatic ring having one or more O, S and/or N), -CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(O)NH* -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl),
-CH2C(O)N(R4)* -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(O)S(lower alkyl), -(CH2)mC(O)NH* -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)SH, -C(O)SR4, -C(O)S(lower alkyl), -C(O)NH* -C(O)NHR4, -C(0)NH(lower alkyl), -C(0)N(R4)* -C(0)N(lower alkyl)2, -O(acyl), -O(lower acyl), -O(R4), -O(alkyl), -0(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), NO2, NH* -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo (fluoro, chloro, bromo, iodo); each m is independently 0, 1 or 2; and alternatively, R6 and R10, R7 and R9, R8 and R7 or R9 and R11 can come together to form a bridged compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle
(preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); or alternatively, R6 and R7 or R9 and R10 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O5 S and/or N).
In a particularly prefened embodiment, a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, in which: • X is O, S, SO or SO2; and/or • each R6 is independently an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, or (CH2)mCONHR4; and/or
• each R7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, CO2-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3> CF2CF3, CH2CO2R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or
• each R9 is independently hydrogen, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-5 O-acyl, F, Cl, Br, I,
CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S-cycloalkyl, C02-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3> CF2CF3, CH2C02R4,
(CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)2, (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or • each R10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substimted cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2C02R4, (CH2)mCOOH, (CH2)mC00R4, (CH2)mCONH2,
(CH2)mCON(R4)2, or (CH2)mC0NHR4; and/or each R8 and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, (CH2)mCONHR4 and N-acyl; and/or each m is independently 0 or 1; and/or
Base is selected from one of the following:
Figure imgf000063_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of
H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* C02-alkyl, CONH-alkyl,
CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN5 (CH2)mN02 and (CH2)mCONH2; is C-R" orN;
T and V independently are CH or N; Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
Q and Q2 independently are N or C-R; wherein R is H, alkyl or acyl;
Q3j Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In a particularly prefened alternative embodiment, a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, in which:
• X is O, S5 SO or S02; and/or
• R6 and R7 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• each R9 is independently hydrogen, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, C Br, I,
CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl5 S-aralkyl, S-acyl, S-cycloalkyl, CO2-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3s CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4,
(CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or • each R10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2C02R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* or (CH2)mCONHR4; and/or • each R8 and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4 and N-acyl; and/or
• each m is independently 0 or 1 ; and/or Base is selected from one of the following:
Figure imgf000065_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of
H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* C02-alkyl, CONH-alkyl,
CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mNO2 and (CH2)mCONH2;
W is C-R" or N;
T and V independently are CH or N; Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
Qj and Q2 independently are N or C-R; wherein R is H, alkyl or acyl;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof. In another particularly prefened embodiment, a compound of Formula (VI), or its pharmaceutically acceptable salt or prodrug thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which:
• X is O, S, SO or SO2; and/or
• each R6 is independently an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH2OH,
CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2C02R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)2, or (CH2)mCONHR4; and/or
• each R7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, CO2-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)*
CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4, (CH2)mCOOH5 (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or
• R9 and R10 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or • each R8 and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4 andN-acyl; and/or
• each m is independently 0 or 1 ; and/or
• Base is selected from one of the following:
Figure imgf000067_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO2-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mN02 and (CH2)mCONH2;
W is C-R" orN;
T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N; Qi and Q2 independently are N or C-R; wherein R is H, alkyl or acyl;
Q3> Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In another particularly prefened embodiment, a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which:
• X is O, S, SO or SO* and/or
• R6 and R7 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• R9 and R10 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• each R8 and Rn is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4 andN-acyl; and/or
• each m is independently 0 or 1 ; and/or • Base is selected from one of the following:
Figure imgf000068_0001
(9) (h)
Figure imgf000068_0002
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, 0-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- 2004/002999
cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO2-alkyl, CONH-alkyl,
CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH* (CH2)mCOOH,
(CH2)mCN, (CH2)mNO2 and (CH2)mCONH2;
W is C-R" orN; T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN;
QΪ and Q2 independently are N or C-R; wherein R is H, alkyl or acyl;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In a particularly prefened embodiment, a compound of Formula (VI), or its pharmaceutically acceptable salt or prodrug thereof, or a stereoisomeric, tautomeric or polymorphic form thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, in which:
• X is CH* CH2OH, CHF, CF* C(Y3)* CHCN, C(CN)* CHR4 or C(R )2; and/or
• each R6 is independently an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3,
CF2CF3, CH2CO2R4, (CH2)mCOOH, (CH2)mC00R4, (CH2)mCONH* (CH2)mCON(R4)* or (CH2)mCONHR4; and/or
• each R7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN,
OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, C02-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)fflCONH2, (CH2)mCON(R4)2, (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered 2004/002999
heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or
• each R9 is independently hydrogen, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I,
CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S-cycloalkyl, C02-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4,
(CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)2, (CH2)mCONHR4, an optionally substimted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or • each R10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2C02R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, or (CH2)mCONHR4; and/or • each R8 and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, (CH2)mCONHR4 andN-acyl; and/or
• each m is independently 0 or 1 ; and/or
• Base is selected from one of the following:
2004/002999
Figure imgf000071_0001
(9) (h)
Figure imgf000071_0002
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* C02-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)fflN02 and (CH2)mCONH2;
W is C-R" orN;
T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N; Qi and Q2 independently are N or C-R;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In a particularly preferred alternative embodiment, a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, as well as a method for the freatment of a host infected with a
Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: 2004/002999
• X is CH* CH2OH, CHF, CF* C(Y3)* CHCN, C(CN)* CHR4 or C(R4)2; and/or
• R6 and R7 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• each R9 is independently hydrogen, optionally substituted lower alkyl, optionally substimted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl,
S-acyl, S-cycloalkyl, C02-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or
• each R10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3,
CF3j CF2CF3, CH2C02R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)* or (CH2)mCONHR4; and/or
• each R8 and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)2, (CH2)mCONHR4 andN-acyl; and/or • each m is independently 0 or 1; and/or
• Base is selected from one of the following:
Figure imgf000073_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* C02-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mNO2 and (CH2)mCONH2;
W is C-R" orN;
T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN; O and Q2 independently are N or C-R; wherein R is H, alkyl or acyl;
Q3; Q4s Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In another particularly prefened embodiment, a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which:
• X is CH* CH2OH, CHF, CF* C(Y3)* CHCN, C(CN)2, CHR4 or C(R4)2; and/or
• each R6 is independently an optionally substimted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substimted cycloalkyl, CH2OH,
CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, or (CH2)mCONHR4; and/or
• each R7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl5 S- cycloalkyl, CO2-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)2,
CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, (CH2)mCONHR4, an optionally substimted 3-7 membered carbocyclic, and an optionally substimted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or
• R9 and R10 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or • each Rs and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4 andN-acyl; and/or
• each m is independently 0 or 1; and/or
• Base is selected from one of the following:
Figure imgf000075_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* C02-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mNO2 and (CH2)mCONH2;
W is C-R" orN;
T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN; O and Q2 independently are N or C-R; wherein R is H, alkyl or acyl;
Q3j Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In another particularly prefened embodiment, a compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which:
• X is CH* CH2OH, CHF, CF* C(Y3)* CHCN, C(CN)* CHR4 or C(R4)2; and/or
• R6 and R7 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• R9 and R10 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• each R8 and Rn is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4 andN-acyl; and/or
• each m is independently 0 or 1 ; and/or • Base is selected from one of the following:
Figure imgf000076_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, CL Br, I, CN, COOH, CONH* C02-alkyl, CONH-alkyl,
CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mNO2 and (CH2)mCONH2;
W is C-R" orN; T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
QΪ and Q2 independently are N or C-R; wherein R is H, alkyl or acyl;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In a particularly prefened embodiment, a compound of Formula (VII), or its pharmaceutically acceptable salt or prodrug thereof, or a stereoisomeric, tautomeric or polymorphic form thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which:
• X* is CH, CF, CY3 or CR4; and/or
• each R6 is independently an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substimted alkynyl, optionally substimted cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3,
CF2CF3, CH2C02R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* or (CH2)mCONHR4; and/or
• each R7 is independently -OH, optionally substituted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN,
OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, CO2-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4, (CH2)mCOOH5
(CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or
• each R9 is independently hydrogen, optionally substimted lower alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I,
CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S-cycloalkyl, CO2-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4,
(CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substimted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or • each R10 is independently hydrogen, an optionally substituted lower alkyl, optionally substituted alkenyl, optionally substimted alkynyl, optionally substimted cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2C02R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)* or (CH2)mCONHR4; and/or • each R8 and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)* (CH2)mCONHR4 andN-acyl; and/or
• each m is independently 0 or 1 ; and/or
• Base is selected from one of the following:
Figure imgf000079_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO2-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mN02 and (CH2)mCONH2;
W is C-R" orN;
T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN; Qi and Q2 independently are N or C-R; wherein R is H, alkyl or acyl;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In a particularly prefened alternative embodiment, a compound of Formula (VII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, as well as a method for the freatment of a host infected with a
Flaviviridae comprising administering an effective freatment amount of compound of Formula (Nil) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which:
• X* is CH, CF, CY3 or CR4; and/or
• R6 and R7 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more Ν, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• each R9 is independently hydrogen, optionally substituted lower alkyl, optionally substimted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, - OH, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I,
CΝ, ΝC, SCΝ, OCΝ, ΝCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S-cycloalkyl, CO2-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH- alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3j CF2CF3, CH2CO2R4,
(CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)* (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substimted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or • each R10 is independently hydrogen, an optionally substituted lower alkyl, optionally substimted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2C02R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* or (CH2)mCONHR4; and/or • each R8 and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)2, (CH2)mCONHR4 and N-acyl; and/or
• each m is independently 0 or 1; and/or
• Base is selected from one of the following:
Figure imgf000081_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* C02-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mN02 and (CH2)mCONH2;
W is C-R" orN;
T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN; Qi and Q2 independently are N or C-R;
R is H, alkyl or acyl;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In another particularly prefened embodiment, a compound of Formula (VII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which:
• X* is CH, CF, CY3 or CR4; and/or
• each R6 is independently an optionally substituted lower alkyl, optionally substimted alkenyl, optionally substimted alkynyl, optionally substimted cycloalkyl, CH2OH,
CH2NH* CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R4, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)2, or (CH2)mCONHR4; and/or
• each R7 is independently -OH, optionally substituted lower alkyl, optionally substimted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, -O-alkyl, -O- alkenyl, -O-alkynyl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aralkyl, S-acyl, S- cycloalkyl, CO2-alkyl, CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)2,
CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3> CF2CF3, CH2C02R4, (CH2)mCOOH, (CH )mCOOR4, (CH2)mCONH2, (CH2)mCON(R4)* (CH2)mCONHR4, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; and/or
• R9 and R10 come together to form a spiro compound selected from the group consisting of optionally substituted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or • each R8 and R11 is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH,
(CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4 and N-acyl; and/or
• each m is independently 0 or 1; and/or
• Base is selected from one of the following:
Figure imgf000083_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO2-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mN02 and (CH2)mCONH2; W is C-R" or N;
T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* orN; Qi and Q2 independently are N or C-R;
R is H, alkyl or acyl;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In another particularly preferred embodiment, a compound of Formula (VII), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which:
• X* is CH, CF, CY3 or CR4; and/or
• R6 and R7 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• R9 and R10 come together to form a spiro compound selected from the group consisting of optionally substimted 3-7 membered spiro carbocyclic or heterocyclic compound having one or more N, O and/or S atoms, said heteroatoms independently taken alone or in combination with one another; and/or
• each R8 and Ru is independently H, CH3, CH2OH, CH2F, CH2N3, (CH2)mCOOH, (CH2)mCOOR4, (CH2)mCONH* (CH2)mCON(R4)* (CH2)mCONHR4 and N-acyl; and/or
• each m is independently 0 or 1; and/or • Base is selected from one of the following:
Figure imgf000084_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substimted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO2-alkyl, CONH-alkyl,
CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH* (CH2)mCOOH,
(CH2)mCN, (CH2)mN02 and (CH2)mCONH2;
W is C-R" or N; T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
Qi and Q2 independently are N or C-R;
R is H, alkyl or acyl;
Q3; Q4, Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In a first subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) 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 substimted 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; 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; (2) R6 is alkyl; (3) R7 and R9 are independently OR2, alkyl, alkenyl, alkynyl, Br-vinyl, O- alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino or di(loweralkyl)amino;
(4) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; (5) X is O, S, S02 or CH2; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3.
In a second subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the freatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) 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 substimted 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; 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; (2) R6 is alkyl, alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino, or di(loweralkyl)amino; (3) R7 and R9 are independently OR2; (4) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; (5) X is O, S, SO2 or CH2; (6) W4 is CX3; and (7) X3 is CH3,
CF3 or CH2CH3.
In a third subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) 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; 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; (2) R6 is alkyl, alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino or di(loweralkyl)amino; (3) R7 and R9 are independently OR2, alkyl, alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino or di(loweralkyl)amino; (4) R8 and R10 are H; (5) X is O, S, SO2 or CH2; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3. In a fourth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the freatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) 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; 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; (2) R6 is alkyl, alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino, or di(loweralkyl)amino; (3) R7 and R9 are independently OR2, alkyl, alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino, or di(loweralkyl)amino; (4) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; (5) X is O; (6) W is CX3; and (7) X3 is CH3, CF3 or CH2CH3. In a fifth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, and the method for the freatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) 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; 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; (2) R6 is alkyl; (3) R7 and R9 are independently OR1; (4) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine; (5) X is O, S, S02 or CH2; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3.
In a sixth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, and the method for the freatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) R1 is independently H or phosphate (including monophosphate, diphosphate, friphosphate, 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; 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; (2) R6 is alkyl; (3) R7 and R9 are independently OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino, or di(loweralkyl)-amino; (4) R8 and R10 are H; (5) X is O, S, SO* or CH2; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3.
In a seventh subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, and the method for the freatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, in which: (1) 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 substimted 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; 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; (2) R6 is alkyl; (3) R7 and R9 are independently OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O-alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino or di(loweralkyl)-amino; (4) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine; (5) X is O; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3.
In a eighth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) 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; 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; (2) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino or di(loweralkyl)amino; (3) R7 and R9 are independently OR2; (4) R8 and R10 are hydrogen; (6) X is O, S, S02 or CH2; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3.
In a ninth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) 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 substimted 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; 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; (2) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino or di(loweralkyl)amino; (3) R7 and R9 are independently OR2; (4) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine or iodine; (5) X is O; (6) W4 is CX3; and (7) X3 is CH3, CF3 or
CH2CH3.
In a tenth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the freatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) 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 substimted 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; 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; (2)
R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino or di(loweralkyl)amino; (3) R7 and R9 are independently OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, O- alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino, or di(loweralkyl)amino; (4) R8 and R10 are hydrogen; (5) X is O; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3.
In an eleventh subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) R1 is independently H or phosphate; (2) R6 is alkyl (including lower alkyl), alkenyl, alkynyl, Br-vinyl, hydroxy, O-alkyl, O-alkenyl, chloro, bromo, fluoro, iodo, NO* amino, loweralkylamino or di(loweralkyl)amino; (3) R7 and R9 are independently OR2; (4) R8 and R10 are hydrogen; (5) X is O, S, SO2 or CH2; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3.
In a twelfth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) R1 is independently H or phosphate; (2) R6 is alkyl; (3) R7 and R9 are independently OR2; (4) R8 and R10 are hydrogen; (5) X is O, S, SO* or CH2; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3. In a thirteenth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) R1 is independently H or phosphate; (2) R6 is alkyl; (3) R7 and R9 are independently OR2; (4) R8 and R10 are independently H, alkyl (including lower alkyl), chlorine, bromine, or iodine; (5) X is O; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3. In a fourteenth subembodiment, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, in which: (1) R1 is independently H or phosphate; (2) R6 is alkyl;
(3) R7 and R9 are independently OR2, alkyl (including lower alkyl), alkenyl, alkynyl, Br- vinyl, O-alkenyl, chlorine, bromine, iodine, NO* amino, loweralkylamino or di(loweralkyl)amino; (4) R8 and R10 are hydrogen; (5) X is O; (6) W4 is CX3; and (7) X3 is CH3, CF3 or CH2CH3. In even more prefened subembodiments, the compound of Formula (VI), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, and the method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (VI) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, in which:
(1) Base is 8-methyladenine; (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 8-methylguanine; (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 6-methylcytosine; (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 6-methylthymidine; (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; 2004/002999
(1) Base is 6-methyluracil; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R and R ,10 are hydrogen; and (6) X is O;
(1) Base is 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (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 8-methyladenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is SO*
(1) Base is 8-methyladenine; (2) R1 is hydrogen; (3) R6 is methyl; (4) R7 and R9 are hydroxyl; (5) R8 and R10 are hydrogen; and (6) X is CH*
In a fifth principal embodiment, a compound of Formula (VIII), (IX) or (X) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric, or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (VIII), (IX), or (X):
Figure imgf000092_0001
(VIII) (IX) (X) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: R1, R2, R3, R4, R5, Y3, X, and X* are as defined above; Base* is a purine or pyrimidine base as defined herein; each R12 is independently a substimted alkyl (including lower alkyl), CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, substimted alkenyl, haloalkenyl (but not Br-vinyl), substituted alkynyl, haloalkynyl, -CH2C(0)OH, -CH2C(O)OR4, -CH2C(O)0(lower alkyl), -CH2C(0)NH* -CH2C(0)NHR4,
-CH2C(0)NH(lower alkyl), -CH2C(0)N(R4)* -CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(0)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)* -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)NH2, -C(0)NHR4, -C(0)NH(lower alkyl), -C(0)N(R4)* -C(0)N(lower alkyl)2; each R13 is independently substimted alkyl (including lower alkyl), CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, substimted alkenyl, haloalkenyl (but not Br-vinyl), substimted alkynyl, haloalkynyl, optionally substimted carbocycle (preferably a 3-7 membered carbocyclic ring), optionally substimted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N), optionally substimted heteroaryl (preferably a 3-7 membered heteroaromatic ring having one or more O, S and/or N), -CH2C(0)OH, -CH2C(0)OR4, -CH2C(0)0(lower alkyl), -CH2C(0)SH, -CH2C(0)SR4, -CH2C(0)S(lower alkyl), -CH2C(0)NH* -CH2C(0)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(0)N(R4)* -CH2C(0)N(lower alkyl)*
-(CH2)mC(0)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)0(lower alkyl), -(CH2)mC(0)SH, -(CH2)mC(0)SR4, -(CH2)mC(O)S(lower alkyl), -(CH2)mC(O)NH* -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)* -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)SH, -C(O)SR4, -C(O)S(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, -O(R4), -O(alkynyl),
-O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), -NHR4, -NR4R5, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), SCN, OCN, NCO or fluoro; alternatively, R12 and R13 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); and each m is independently 0, 1 or 2. In a sixth principal embodiment, a compound of Formula (XI) or (XII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric, or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (XI) or (XII):
Figure imgf000094_0001
(XI) (XII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: Base*, R, R1, R2, R3, R4, R5, R12, R13, Y, Y1, Y2, Y3, W*, W1, W2, W3, W4, X, X*, X2 and
X3 are as defined above; wherein, in one embodiment, R8 in Formula (XI) is -OH or -NH2 only when X is carbon; and wherein; each R8 and Rπ is independently hydrogen, an optionally substimted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH* CH2NHCH3, CH2N(CH3)* CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH2C(0)OH, -CH2C(0)OR4, -CH2C(0)0(lower alkyl), -CH2C(O)NH* -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(0)N(R4)* -(CH2)mC(0)N(lower alkyl)2, -C(0)OH, -C(0)OR4, -C(0)0(lower alkyl), -C(0)NH2, -C(O)NHR4, -C(0)NH(lower alkyl), -C(0)N(R4)2, -C(0)N(lower alkyl)2, cyano, NH-acyl or N(acyl)2; each R9 and R10 are independently hydrogen, OH, OR2, optionally substimted alkyl
(including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)* CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, optionally substimted carbocycle (preferably a 3-7 membered carbocyclic ring), optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N), optionally substimted heteroaryl (preferably a 3-7 membered heteroaromatic ring having one or more O, S and/or N), -CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(O)NH* -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)* -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(0)OR4,
-(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(O)S(lower alkyl), -(CH2)mC(O)NH* -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(O)N(R4)* -(CH2)mC(0)N(lower alkyl)2, -C(0)OH, -C(O)OR4, -C(O)0(lower alkyl), -C(O)SH, -C(O)SR4, -C(0)S(lower alkyl), -C(0)NH* -C(0)NHR4, -C(0)NH(lower alkyl), -C(O)N(R4)* -C(O)N(lower alkyl)2, -O(acyl), -Oøower acyl), -O(R4), -O(alkyl),
-O(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), NO* NH* -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo (fluoro, chloro, bromo, iodo); each m is independently 0, 1 or 2; and alternatively, R8 and R13, R9 and R13, R9 and Rn or R10 and R12 can come together to form a bridged compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substimted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N); or alternatively, R12 and R13 or R9 and R10 can come together to form a spiro compound selected from the group consisting of optionally substimted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substituted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or N).
In a particular apect of the invention, compounds of the Formula (XIII) or (XIV) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective freatment amount of compound of Formula (XIII) or (XIV):
Figure imgf000096_0001
(XIII) (XIV) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: R3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; acyl; a sulfonate ester; optionally substimted alkyl sulfonyl; optionally substituted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R3 is independently H, or mono-, di- or triphosphate; X" is selected from the group consisting of one or more O, S, SO, SO* N, NH, NR and CH2 wherein any of the aforementioned may be optionally substimted and may be variably positioned so as to form a 3-7 membered ring;
R is H, alkyl or acyl;
B indicates a spiro compound selected from the group consisting of optionally substituted carbocycle (preferably a 3-7 membered carbocyclic ring) or optionally substimted heterocycle (preferably a 3-7 membered heterocyclic ring having one or more O, S and/or
N); and Base is selected from the group consisting of:
Figure imgf000097_0001
wherein: each R', R", R'" and R"" are independently selected from the group consisting of
H, OH, substimted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO2-alkyl, CONH-alkyl,
CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH* (CH2)mCOOH,
(CH2)mCN, (CH2)mNO2 and (CH2)mCONH2; m is 0 or 1;
W is C-R" orN; T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
Qi and Q2 independently are N or C-R;
R is H, alkyl or acyl;
Q Q , Q5 and Q6 independently are N or CH; and tautomeric forms thereof.
In a second particular apect of the invention, a compound of Formula (XV), (XVI) or (XVII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XV), (XVI), or (XVII):
Figure imgf000098_0001
or its pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein:
G and E independently are selected from the group consisting of H, CH3, CH2OH5 CH F, CH2N3, CH2CN, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH* (CH2)mCONR* (CH2)mCONHR and N-acyl; R is H, alkyl or acyl; m is O or 1; and R3 and Base are as defined for Formula (XIII).
Alternatively, for compound of Formula (XVII), at most one of G and E can further be hydrogen.
In a third particular apect of the invention, a compound of Formula (XVIII) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, as well as a method for the freatment of a host infected with a
Flaviviridae comprising administering an effective treatment amount of compound of Formula (XVIII):
Figure imgf000098_0002
(XVIII) or its pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, wherein: M is selected from the group consisting of S, SO, and SO2; and
R3 and Base are as defined for Formula (XIII). In a fourth particular aspect of the invention, a compound of Formula (XIX), (XX), (XXI) (XXII) or (XXπi) or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymoφhic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (XIX), (XXI), (XXII), or (XXIII):
Figure imgf000099_0001
(XXIII) or its pharmaceutically acceptable salt or prodrug or a stereoisomeric, tautomeric or polymorphic form thereof, wherein:
A is selected from the group consisting of optionally substituted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C15 CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCO- NH* (CH2)mCONR* and (CH2)mCONHR;
Y is selected from the group consisting of H, optionally substimted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)* CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH* (CH2)mCONR2, and (CH2)mCONHR; R is H, alkyl or acyl;
X is selected from the group consisting of -OH, optionally substituted alkyl, cycloalkyl, alkenyl, alkynyl, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aryl, -O-aralkyl, -O-cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO* NH* N3, NH-acyl, NH-alkyl, N- dialkyl, NH-alkenyl, NH-alkynyl, NH-aryl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S- alkenyl, S-alkynyl, S-aryl, S-aralkyl, S-acyl, S-cycloalkyl, C02-alkyl, CONH-alkyl,
CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH-cycloalkyl, CH2OH, CH2NH* CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3, CF2CF3, CH2C02R5 (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH* (CH2)mCONR2, (CH2)mCONHR, an optionally substituted 3-7 membered carbocyclic, and an optionally substituted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; m is O or 1; R3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; substituted or unsubstituted alkyl; acyl; a sulfonate ester; optionally substituted alkyl sulfonyl; optionally substituted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein Ri is independently H, or mono-, di- or triphosphate; and
Base is a non-natural base selected from the group of:
Figure imgf000100_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of H, OH, substimted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH* NH-alkyl, N-dialkyl, NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH* CO2-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH, (CH2)mCN, (CH2)mNO2 and (CH2)mCONH2; m is 0 or 1; W is C-R" or N;
T and V independently are CH or N; Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH* or N;
Qi and Q2 independently are N or C-R""; and
Q3, Q4, Q5 and Q6 independently are N or CH; with the proviso that in bases (g) and (i), R', R"" are not H, OH, or NH* and Q,
T, V, Q* Q5 and Q6 are not N.
In another prefened embodiment, a compound of Formula (IX), or a pharmaceutically acceptable salt or prodrug, or a stereoisomeric, tautomeric or polymorphic form thereof, is provided, as well as a method for the treatment of a host infected with a Flaviviridae comprising administering an effective treatment amount of compound of Formula (IX):
Figure imgf000101_0001
(IX) or a stereoisomeric, tautomeric or polymoφhic form thereof, or a pharmaceutically acceptable salt thereof, wherein:
R1, R2 and R3 are independently H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl; CO-aryl; CO-alkoxyalkyl; CO-aryloxyalkyl; CO-substituted aryl; sulfonate ester; benzyl, wherein the phenyl group is optionally substituted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; or a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 and/or R3 is independently H or phosphate;
X is O, S, SO2 or CH2;
Base* is a purine or pyrimidine base;
R12 is C(Y3)3;
Y3 is independently H, F, Cl, Br or I; and
R13 is fluoro.
In one subembodiment X is O, and Y3 is H. In another subembodiment, when X is O and Y3 is H, R1, R2 and R3 are also H. II. Stereochemistry
It is appreciated that nucleosides of the present invention have several chiral centers and may exist in and be isolated in optically active and racemic forms. Some compounds may exhibit polymoφhism. It is to be understood that the present invention encompasses any racemic, optically-active, diastereomeric, polymorphic, or stereoisomeric form, or mixtures thereof, of a compound of the invention, which possess the useful properties described herein. It being well known in the art how to prepare optically active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).
In particular, since the 1' and 4' carbons of the nucleoside are chiral, their nonhydrogen substituents (the base and the CHOR groups, respectively) can be either cis (on the same side) or trans (on opposite sides) with respect to the sugar ring system. The four optical isomers therefore are represented by the following configurations (when orienting the sugar moiety in a horizontal plane such that the oxygen atom is in the back): cis (with both groups "up", which corresponds to the configuration of naturally occuning β- D nucleosides), cis (with both groups "down", which is a nonnaturally occuning β-L configuration), trans (with the C2' substituent "up" and the C4' substituent "down"), and trans (with the C2' substituent "down" and the C4' substituent "up"). The "D-nucleosides" are cis nucleosides in a natural configuration and the "L-nucleosides" are cis nucleosides in the nonnaturally occuning configuration.
Likewise, most amino acids are chiral (designated as L or D, wherein the L enantiomer is the naturally occuning configuration) and can exist as separate enantiomers. Examples of methods to obtain optically active materials are known in the art, and include at least the following. i) physical separation of crystals - a technique whereby macroscopic crystals of the individual enantiomers are manually separated. This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct; ii) simultaneous crystallization - a technique whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state; iii) enzymatic resolutions - a technique whereby partial or complete separation of a racemate by virtue of differing rates of reaction for the enantiomers with an enzyme; iv) enzymatic asymmetric synthesis - a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer; v) chemical asymmetric synthesis - a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries; vi) diastereomer separations - a technique whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers. The resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer; vii) first- and second-order asymmetric transformations - a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer. The desired enantiomer is then released from the diastereomer; viii) kinetic resolutions - this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions; ix) enantiospecific synthesis from non-racemic precursors - a synthetic technique whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis; x) chiral liquid chromatography - a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase. The stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions; xi) chiral gas chromatography - a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase; xii) extraction with chiral solvents - a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent; xiii) transport across chiral membranes - a technique whereby a racemate is placed in contact with a thin membrane barrier. The barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane banier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
III. 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 C\ to
Cio, and specifically includes methyl, CF3, CC13, CFC1* CF2C1, ethyl, CH2CF3, CF2CF3, propyl, isopropyl, cyclopropyl, butyl, isobutyl, secbutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 3-methylpentyl, 2,2- dimethylbutyl, and 2,3-dimethylbutyl. The term includes both substimted and unsubstituted alkyl groups, and particularly includes halogenated alkyl groups, and even more particularly fluorinated alkyl groups. Non-limiting examples of moieties with which the alkyl group can be substimted are selected from the group consisting of halogen (fluoro, chloro, bromo or iodo), hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, nifro, 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 incoφorated by reference.
The term "lower alkyl", as used herein, and unless otherwise specified, refers to a Ci to C4 saturated straight, branched, or if appropriate, a cyclic (for example, cyclopropyl) alkyl group, including both substituted and unsubstituted moieties.
The term "alkylamino" or "arylamino" refers to an amino group that has one or two alkyl or aryl substituents, respectively. Unless otherwise specifically stated in this application, when alkyl is a suitable moiety, lower alkyl is prefened. Similarly, when alkyl or lower alkyl is a suitable moiety, unsubstituted alkyl or lower alkyl is prefened.
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 puφoses. 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 unsubstimted moieties. The aryl group can be substimted with any described moiety, including, but not limited to,one or more moieties selected from the group consisting of halogen (fluoro, chloro, bromo or iodo), 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-alkylaminopurine, 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-iodopyrimidine, C6- iodo-pyrimidine, C5-Br-vinyl pyrimidine, C6-Br-vinyl pyrimidine, C5-nitropyrimidine, C5-amino-pyrimidine, N2-alkylpurines, N2-alkyl-6-thiopurines, 5-azacytidinyl,
5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pynolopyrimidinyl, and pyrazolopyrimidinyl. 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 frimethylsilyl, dimethylhexylsilyl, t- butyldimethylsilyl, and t-butyldiphenylsilyl, frityl, alkyl groups, and acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl.
The term "acyl" or "O-linked ester" refers to a group of the formula C(O)R', wherein R' is an straight, branched, or cyclic alkyl (including lower alkyl), amino acid, aryl including phenyl, alkaryl, aralkyl including benzyl, alkoxyalkyl including methoxymethyl, aryloxyalkyl such as phenoxymethyl; or substimted alkyl (including lower alkyl), aryl including phenyl optionally substituted with chloro, bromo, fluoro, iodo, to C4 alkyl or d to C4 alkoxy, sulfonate esters such as alkyl or aralkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxy-trityl, substituted benzyl, alkaryl, aralkyl including benzyl, alkoxyalkyl including methoxymethyl, aryloxyalkyl such as phenoxymethyl. Aryl groups in the esters optimally comprise a phenyl group. In particular, acyl groups include acetyl, trifluoroacetyl, methylacetyl, cyclopropylacetyl, cyclopropyl carboxy, propionyl, butyryl, hexanoyl, heptanoyl, octanoyl, neo-heptanoyl, phenylacetyl, 2-acetoxy-2-phenylacetyl, diphenylacetyl, α-methoxy-α- trifluoromethyl-phenylacetyl, bromoacetyl, 2-nitro-benzeneacetyl, 4-chloro-benzeneacetyl, 2-chloro-2,2-diphenylacetyl, 2-chloro-2-phenylacetyl, frimethylacetyl, chlorodifluoroacetyl, perfluoroacetyl, fluoroacetyl, bromodifluoroacetyl, methoxyacetyl, 2-thiopheneacetyl, chlorosulfonylacetyl, 3-methoxyphenylacetyl, phenoxyacetyl, tert-butylacetyl, trichloroacetyl, monochloro-acetyl, dichloroacetyl, 7H-dodecafluoro-heptanoyl, perfluoro- heptanoyl, 7H-dodeca-fluoroheptanoyl, 7-chlorododecafluoro-heptanoyl, 7-chloro- dodecafluoro-heptanoyl, 7H-dodecafluoroheptanoyl, 7H-dodeca-fluoroheptanoyl, nona- fluoro-3,6-dioxa-heptanoyl, nonafluoro-3,6-dioxaheptanoyl, perfluoroheptanoyl, methoxybenzoyl, methyl 3-amino-5-phenylthiophene-2-carboxyl, 3,6-dichloro-2-methoxy- benzoyl, 4-(l,l,2,2-tetrafluoro-ethoxy)-benzoyl, 2-bromo-propionyl, omega-aminocapryl, decanoyl, n-pentadecanoyl, stearyl, 3-cyclopentyl-propionyl, 1-benzene-carboxyl, O- acetylmandelyl, pivaloyl acetyl, 1-adamantane-carboxyl, cyclohexane-carboxyl, 2,6- pyridinedicarboxyl, cyclopropane-carboxyl, cyclobutane-carboxyl, perfluorocyclohexyl carboxyl, 4-methylbenzoyl, chloromethyl isoxazolyl carbonyl, perfluorocyclohexyl carboxyl, crotonyl, 1 -methyl- lH-indazole-3 -carbonyl, 2-propenyl, isovaleryl, 1- pyrrolidinecarbonyl, 4-phenylbenzoyl. When the term acyl is used, it is meant to be a specific and independent disclosure of acetyl, trifluoroacetyl, methylacetyl, cyclopropylacetyl, propionyl, butyryl, hexanoyl, heptanoyl, octanoyl, neo-heptanoyl, phenylacetyl, diphenylacetyl, α-frifluoromethyl-phenylacetyl, bromoacetyl, 4-chloro- benzeneacetyl, 2-chloro-2,2-diphenylacetyl, 2-chloro-2-phenylacetyl, frimethylacetyl, chlorodifluoroacetyl, perfluoroacetyl, fluoroacetyl, bromodifluoroacetyl, 2-thiopheneacetyl, tert-butylacetyl, trichloroacetyl, monochloro-acetyl, dichloroacetyl, methoxybenzoyl, 2- bromo-propionyl, decanoyl, n-pentadecanoyl, stearyl, 3-cyclopentyl-propionyl, 1-benzene- carboxyl, pivaloyl acetyl, 1-adamantane-carboxyl, cyclohexane-carboxyl, 2,6- pyridinedicarboxyl, cyclopropane-carboxyl, cyclobutane-carboxyl, 4-methylbenzoyl, crotonyl, l-methyl-lH-indazole-3-carbonyl, 2-propenyl, isovaleryl, 4-phenylbenzoyl.
The term "amino acid" includes naturally occuning and synthetic , β γ or δ amino acids, and includes but is not limited to, amino acids found in proteins, i.e. glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine. In a prefened embodiment, the amino acid is in the L-configuration. Alternatively, the amino acid can be a derivative of alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, β- alanyl, β-valinyl, β-leucinyl, β-isoleuccinyl, β-prolinyl, β-phenylalaninyl, β-tryptophanyl, β-methioninyl, β-glycinyl, β-serinyl, β-threoninyl, β-cysteinyl, β-tyrosinyl, β-asparaginyl, β-glutaminyl, β-aspartoyl, β-glutaroyl, β-lysinyl, β-argininyl or β-histidinyl. Tables 1-24 set out examples of species within the present invention. When the term amino acid is used, it is considered to be a specific and independent disclosure of each of the esters of α, β γ or δ glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine in the D and L-configurations.
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%, 98 %, 99% or 100% by weight, of the designated enantiomer of that nucleoside. In a prefened 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%, 90%, 95%, 98%, 99%, or 100% by weight, of the nucleoside, the remainder comprising other chemical species or enantiomers.
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 Flaviviridae viral 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 Flaviviridae genome and animals, in particular, primates (including chimpanzees) and humans. In most animal applications of the present invention, the host is a human patient. Veterinary applications, in certain indications, however, are clearly anticipated by the present invention (such as chimpanzees).
The 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 a Flaviviridae, or are metabolized to a compound that exhibits such activity.
IV. Prodrugs and Derivatives
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 refened to as "physiologically acceptable salts"), and a compound, which has been alkylated, acylated, or otherwise modified 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.
A. Pharmaceutically Acceptable Salts
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 by addition of acids, which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorate, α-ketoglutarate, α-glycerophosphate, formate, fumarate, propionate, glycolate, lactate, pyruvate, oxalate, maleate, and salicylate. Suitable inorganic salts may also be formed, including, sulfate, nitrate, bicarbonate, carbonate salts, hydrobromate and phosphoric acid. In a prefened embodiment, the salt is a mono- or di- hydrochloride salt.
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. In one embodiment, the salt is a hydrochloride salt of the compound. In another embodiment, the pharmaceutically acceptable salt is a dihydrochloride salt. B. Nucleotide Prodrug Formulations
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 reduces polarity and allows passage into cells. 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. Bischoferger, Antiviral Research, 1995, 27:1-17. Any of these can be used in combination with the disclosed nucleosides to achieve a desired effect. In an alternative embodiment, the nucleoside is delivered as a phosphonate or a
SATE derivative.
The active nucleoside can also be provided as a 2', 3' and/or 5'-phosphoether lipid or a 2', 3' and/or 5'-ether lipid. Non-limiting examples are described include 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. 1990. "Novel membrane-interactive ether lipid analogs that inhibit infectious HJN-1 production and induce defective virus formation." AIDS Res. Hum. Retro Viruses. 6:491-501; Piantadosi, C, J. Marasco C.J., S.L. Morris-Νatschke, K.L. Meyer, F. Gumus, J.R. Surles, K.S. Ishaq, L.S. Kucera, Ν. Iyer, CA. Wallen, S. Piantadosi, and E.J. Modest. 1991. "Synthesis and evaluation of novel ether lipid nucleoside conjugates for anti-HIV activity." J. Med. Chem. 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. 1992. "Greatly enhanced inhibition of human immunodeficiency virus type 1 replication in CEM and HT4-6C cells by 3'-deoxythymine diphosphate dimyristoylglycerol, a lipid prodrug of 3,-deoxythymine." Antimicrob. Agents Chemother.
36:2025.2029; Hosetler, K.Y., L.M. Stuhmiller, H.B. Lenting, H. van den Bosch, and D.D. Richman, 1990. "Synthesis and antiretroviral activity of phospholipid analogs of azidothymine and other antiviral nucleosides." J. Biol Chem. 265:61127.
Nonlimiting examples of U.S. patents that disclose suitable lipophilic substituents that can be covalently incorporated into the nucleoside, preferably at the 2', 3' and/or 5'-OH position of the nucleoside or lipophilic preparations, include U.S. Patent Nos. 5,149,794 (Sep. 22, 1992, Yatvin et al); 5,194,654 (Mar. 16, 1993, Hostetler et al, 5,223,263 (June 29, 1993, Hostetler et al); 5,256,641 (Oct. 26, 1993, Yatvin et al); 5,411,947 (May 2, 1995, Hostetler et al); 5,463,092 (Oct. 31, 1995, Hostetler et al); 5,543,389 (Aug. 6, 1996, Yatvin et al); 5,543,390 (Aug. 6, 1996, Yatvin et al); 5,543,391 (Aug. 6, 1996, Yatvin et al); and 5,554,728 (Sep. 10, 1996; Basava et al), all of which are incoφorated 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, W0 90/00555, W0 91/16920, W0 91/18914, W0 93/00910, W0 94/26273, W0 96/15132, EP 0350287, EP 93917054.4, and W0 91/19721.
Aryl esters, especially phenyl esters, are also provided. Nonlimiting examples are disclosed in DeLambert et al., J. Med. Chem. 37: 498 (1994). Phenyl esters containing a carboxylic ester ortho to the phosphate are also provided. Khamnei and Tonence, J. Med. Chem.; 39:4109-4115 (1996). In particular, benzyl esters, which generate the parent compound, in some cases using substituents at the ortho- or para-position to accelerate hydrolysis, are provided. Examples of this class of prodrugs are described by Mitchell et al., J. Chem. Soc. Perkin Trans. I 2345 (1992); Brook, et al. WO 91/19721; and Glazier et al. WO 91/19721.
Cyclic and noncyclic phosphonate esters are also provided. Nonlimiting examples are disclosed in Hunston et al., J. Med. Chem. 27: 440-444 (1984) and Starrett et al. J. Med.
Chem. 37: 1857-1864 (1994). Additionally, cyclic 3',5'-phosphate esters are provided. Nonlimiting examples are disclosed in Meier et al. J. Med. Chem. 22: 811-815 (1979). Cyclic l',3'-propanyl phosphonate and phosphate esters, such as ones containing a fused aryl ring, i.e. the cyclosaligenyl ester, are also provided (Meier et al., Bioorg. Med. Chem. Lett. 7: 99-104 (1997)). Unsubstituted cyclic l',3'-ρropanyl esters of the monophosphates are also provided (Farquhar et al., J. Med. Chem. 26: 1153 (1983); Farquhar et al., J. Med. Chem. 28: 1358 (1985)) were prepared. In addition, cyclic l',3'-propanyl esters substimted with a pivaloyloxy methyloxy group at C-l' are provided (Freed et al., Biochem. Pharmac. 38: 3193 (1989); Biller et al., U.S. Pat. No. 5,157,027).
Cyclic phosphoramidates are known to cleave in vivo by an oxidative mechanism. Therefore, in one embodiment of the present invention, a variety of substituted l',3' propanyl cyclic phosphoramidates are provided. Non-limiting examples are disclosed by Zon, Progress in Med. Chem. 19, 1205 (1982). Additionally, a number of 2'- and 3'- substimted proesters are provided. 2'-Substituents include methyl, dimethyl, bromo, trifluoromethyl, chloro, hydroxy, and methoxy; 3 '-substituents including phenyl, methyl, trifluoromethyl, ethyl, propyl, i-propyl, and cyclohexyl. A variety of l'-substituted analogs are also provided.
Cyclic esters of phosphorus-containing compounds are also provided. Non-limiting examples are described in the following:
• [1] di and tri esters of phosphoric acids as reported in Nifantyev et al., Phosphorus, Sulfur Silicon and Related Eelements, 113: 1 (1996); Wijnberg et al., EP-180276 Al;
• [2] phosphorus (III) acid esters. Kryuchkov et al., Izv. Akad. Nauk SSSR, Ser. Khim. 6: 1244 (1987). Some of the compounds were claimed to be useful for the asymmetric synthesis of L-Dopa precursors. Sylvain et al., DE3512781 Al;
• [3] phosphoramidates. Shih et al., Bull. Inst. Chem. Acad. Sin, 41: 9 (1994); Edmundson et al., J. Chem. Res. Synop. 5: 122 (1989); and
• [4] phosphonates. Neidlein et al., Heterocycles 35: 1185 (1993).
Further, nonlimiting examples of U.S. and International Patent Applications that disclose suitable cyclic phosphoramidate prodrugs include U.S. Patent No. 6,312,662; WO 99/45016; WO 00/52015; WO 01/47935; and WO 01/18013 to Erion, et al. from Metabasis Therapeutics, Inc. Specifically, prodrugs of the formula below are provided:
Figure imgf000112_0001
wherein:
• together V and Z are connected via an additional 3-5 atoms to form a cyclic group containing 5-7 atoms, optionally 1 heteroatom, substimted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from both O groups attached to the phosphorus; or
• together V and Z are connected via an additional 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryl group at the beta, and gamma position to the O attached to the phosphorus;
• together V and W are connected via an additional 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to one of said carbon atoms that is three atoms from an O attached to the phosphorus;
• together Z and W are connected via an additional 3-5 atoms to form a cyclic group, optionally containing one heteroatom, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
• together W and W are connected via an additional 2-5 atoms to form a cyclic group, optionally containing 0-2 heteroatoms, and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; • Z is selected from the group consisting of -CHR2 OH, -CHR2 OC(O)R3, -CHR2
OC(S)R3, -CHR2 OC(S)OR3, -CHR2 OC(0)SR3, -CHR2 OC02 R3, -OR2, -SR2, - CHR2 N3, -CH2 aryl, -CH(aryl)OH, -CH(CH=CR2 2)OH, -CH(C.ident.CR2)OH, -R2, -NR2* -OCOR3, -OCO2 R3, -SCOR3, -SCO2 R3, -NHCOR2, -NHCO2 R3, -CH2
NHaryl, -(CH2)P -OR12, and -(CH2)P -SR12 ;
• p is an integer 2 or 3;
• with the provisos that: • a) V, Z, W, W are not all -H; and
• b) when Z is -R2, then at least one of V, W, and W is not -H, alkyl, aralkyl, or alicyclic;
• R2 is selected from the group consisting of R3 and -H;
• R3 is selected from the group consisting of alkyl, aryl, alicyclic, and aralkyl; • R12 is selected from the group consisting of -H, and lower acyl;
• M is the biologically active agent, and that is attached to the phosphorus in formula I via the 2', 3' and/or 5'-hydroxyl.
V. Combination or Alternation Therapy The active compounds of the present invention can be administered in combination or alternation with another anti-flavivirus or pestivirus agent, or in particular an anti-HCV agent to treat any of the conditions described herein. In combination therapy, effective dosages of two or more agents are administered together, whereas in alternation or sequential-step therapy, an effective dosage of each agent is administered serially or sequentially. The dosages given will depend on absoφtion, 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. In prefened embodiments, an anti-HCV (or anti-pestivirus or anti-flavivirus) compound that exhibits an EC50 of 10-15 μM, or preferably less than 1-5 μM, is desirable.
It has been recognized that drug-resistant variants of flaviviruses, pestiviruses or HCV 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 the viral 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 prefened over alternation therapy because it induces multiple simultaneous stresses on the virus. Any of the viral treatments described in the Background of the Invention can be used in combination or alternation with the compounds described in this specification. Nonlimiting examples include:
1) Protease inhibitors Examples include substrate-based NS3 protease inhibitors (Attwood et al., Antiviral peptide derivatives, PCT WO 98/22496, 1998; Attwood et al., Antiviral Chemistry and Chemotherapy 1999, 10, 259-273; Attwood et al., Preparation and use of amino acid derivatives as anti-viral agents, German Patent Pub. 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); Non-substrate-based NS3 protease 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 substimted on the amide with a 14 carbon chain and the latter processing a para-phenoxyphenyl group; and Sch 68631, a phenanthrenequinone, an HCV protease inhibitor (Chu M. et al., Tetrahedron Letters 37:7229-7232, 1996).
Sch 351633, isolated from the fungus Penicillium griseofulvum, was identified as a protease inhibitor (Chu M. et al., Bioorganic and Medicinal Chemistry Letters 9:1949-
1952). Eglin c, isolated from leech, is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, α-chymotrypsin, chymase and subtilisin. Qasim M.A. et al., Biochemistry 36:1598-1607, 1997.
U.S. patents disclosing protease inhibitors for the treatment of HCV include, for example, U.S. Patent No. 6,004,933 to Spruce et al. which discloses a class of cysteine protease inhibitors for inhibiting HCV endopeptidase 2; U.S. Patent No. 5,990,276 to Zhang et al. which discloses synthetic inhibitors of hepatitis C virus NS3 protease; U.S. Patent No. 5,538,865 to Reyes et a; WO 02/008251 to Corvas International, Inc, and WO 02/08187 and WO 02/008256 to Schering Corporation. HCV inhibitor tripeptides are disclosed in US Patent Nos. 6,534,523, 6,410,531, and 6,420,380 to Boehringer Ingelheim and WO 02/060926 to Bristol Myers Squibb. Diaryl peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/48172 to Schering Coφoration. Imidazoleidinones as NS3 serine protease inhibitors of HCV are disclosed in WO 02/08198 to Schering Coφoration and WO 02/48157 to Bristol Myers Squibb. WO 98/17679 to
Vertex Pharmaceuticals and WO 02/48116 to Bristol Myers Squibb also disclose HCV protease inhibitors.
2) 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;
3) 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;
4) 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 griseofulvum, which demonstrates activity in a scintillation proximity assay
(Chu M. et al., Bioorganic and Medicinal Chemistry Letters 9, 1949-1952);
5) Helicase inhibitors (Diana G.D. et al., Compounds, compositions and methods for freatment 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);
6) Nucleotide polymerase inhibitors and gliotoxin (Fenari R. et al. Journal of Virology, 1999, 73, 1649-1654), and the natural product cerulenin (Lohmann V. et al., Virology, 1998, 249, 108-118);
7) Antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5' non-coding region (NCR) of the virus (Alt M. et al., Hepatology, 1995, 22, 707-717), or nucleotides 326-348 comprising the 3' end of the NCR and nucleotides 371-388 located in the core coding region of the HCV RNA (Alt M. et al., Archives of Virology, 1997, 142, 589-599; Galderisi U. et al, Journal of Cellular Physiology, 1999, 181, 251-257); 8) 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 freatment of viral diseases, Japanese Patent Pub. JP-10101591); 9) Ribozymes, such as nuclease-resistant ribozymes (Maccjak, D. J. et al.,
Hepatology 1999, 30, abstract 995) and those disclosed in U.S. Patent No. 6,043,077 to Barber et al., and U.S. Patent Nos. 5,869,253 and 5,610,054 to Draper et al.; and
10) Nucleoside analogs have also been developed for the treatment of Flaviviridae infections.
11) any of the compounds described by Idenix Pharmaceuticals in International Publication Nos. WO 01/90121 and WO 01/92282; .
12) Compound in other patent applications disclosing the use of certain nucleoside analogs to treat hepatitis C virus include: PCT/CAOO/01316 (WO 01/32153; filed November 3, 2000) and PCT/CA01/00197 (WO 01/60315; filed February
19, 2001) filed by BioChem Pharma, Inc. (now Shire Biochem, Inc.); PCT/US02/01531 (WO 02/057425; filed January 18, 2002) and PCT/US02/03086 (WO 02/057287; filed January 18, 2002) filed by Merck & Co., Inc., PCT/EPOl/09633 (WO 02/18404; published August 21, 2001) filed by Roche, and PCT Publication Nos. WO 01/79246 (filed April 13, 2001), WO
02/32920 (filed October 18, 2001) and WO 02/48165 by Pharmasset, Ltd.
13) PCT Publication No. WO 99/43691 to Emory University, entitled "2'- Fluoronucleosides" discloses the use of certain 2'-fluoronucleosides to treat HCV. 14) Other miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S.
Patent 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.), benzimidazoles (U.S. Pat. No. 5,891,874 to Colacino et al.), plant extracts (U.S. Patent No. 5,837,257 to Tsai et al., U.S. Patent No. 5,725,859 to Omer et al., and U.S. Patent No. 6,056,961), and piperidenes (U.S. Patent No. 5,830,905 to Diana et al.). 15) Other compounds cunently in preclinical or clinical development for treatment of hepatitis c virus include: Interleukin- 10 by Schering-Plough, IP-501 by Intemeuron, Merimebodib (VX-497) by Vertex, AMANTADINE®
(Symmefrel) by Endo Labs Solvay, HEPTAZYME® by RPI, IDN-6556 by Idun Pharma., XTL-002 by XTL., HCV/MF59 by Chiron, CIVACIR® (Hepatitis C Immune Globulin) by NABI, LEVOVIRIN® by ICN/Ribapharm, VIRAMIDINE® by ICN/Ribapharm, ZADAXIN® (thymosin alpha-1) by Sci Clone, thymosin plus pegylated interferon by Sci Clone, CEPLENE®
(histamine dihydrochloride) by Maxim, VX 950 / LY 570310 by Vertex/Eli Lilly, ISIS 14803 by Isis Pharmaceutical/Elan, IDN-6556 by Idun Pharmaceuticals, Inc., JTK 003 by AKROS Pharma, BILN-2061 by Boehringer Ingelheim, CellCept (mycophenolate mofetil) by Roche, T67, a β- tubulin inhibitor, by Tularik, a therapeutic vaccine directed to E2 by
Innogenetics, FK788 by Fujisawa Healthcare, Inc., IdB 1016 (Siliphos, oral silybin-phosphatdylcholine phytosome), RNA replication inhibitors (VP50406) by ViroPharma/Wyeth, therapeutic vaccine by Intercell, therapeutic vaccine by Epimmune/Genencor, IRES inhibitor by Anadys, ANA 245 and ANA 246 by Anadys, immunotherapy (Therapore) by Avant, protease inhibitor by Corvas/SChering, helicase inhibitor by Vertex, fusion inhibitor by Trimeris, T cell therapy by CellExSys, polymerase inhibitor by Biocryst, targeted RNA chemistry by PTC Therapeutics, Dication by Immtech, Int., protease inhibitor by Agouron, protease inhibitor by Chiron/Medivir, antisense therapy by AVI BioPharma, antisense therapy by Hybridon, hemopurifier by
Aethlon Medical, therapeutic vaccine by Merix, protease inhibitor by Bristol- Myers Squibb/Axys, Chron-VacC, a therapeutic vaccine, by Tripep, UT 23 IB by United Therapeutics, protease, helicase and polymerase inhibitors by Genelabs Technologies, IRES inhibitors by Immusol, R803 by Rigel Pharmaceuticals, INFERGEN® (interferon alphacon-1) by InterMune,
OMNIFERON® (natural interferon) by Viragen, ALBUFERON® by Human Genome Sciences, REBIF® (interferon beta- la) by Ares-Serono, Omega Interferon by BioMedicine, Oral Interferon Alpha by Amarillo Biosciences, interferon gamma, interferon tau, and Interferon gamma- lb by InterMune. VI. Pharmaceutical Compositions
Hosts, including humans, infected with pestivirus, flavivirus, HCV infection, or any other condition described herein, or another organism replicating through a RNA-dependent RNA viral polymerase, or for treating any other disorder described herein, 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 canier or dilutent. 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 pestivirus, flavivirus or HCV 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. Lower doses may be preferable, for example doses of 0.5-100 mg, 0.5-50 mg, 0.5-10 mg, or 0.5-5 mg per kilogram body weight per day. Even lower doses may be useful, and thus ranges can include from 0.1-0.5 mg per kilogram body weight 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. An oral dosage of 50-1000 mg is usually convenient, including in one or multiple dosage forms of 50, 100, 200, 250, 300, 400, 500, 600, 700, 800, 900 or 1000 mgs. Lower doses may be preferable, for example from 10-100 or 1-50 mg. Also contemplated are doses of 0.1-50 mg, or 0.1-20 mg or 0.1-10.0 mg. Furthermore, lower doses may be utilized in the case of administration by a non-oral route, as, for example, by injection or inhalation.
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 absoφtion, 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 prefened mode of administration of the active compound is oral. Oral compositions will generally include an inert diluent or an edible canier. They may be enclosed in gelatin capsules or compressed into tablets. For the p pose of oral therapeutic administration, the active compound can be incoφorated with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can e 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, sucutaneous, 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 ethylenediaminetefraacetic 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, prefened caniers are physiological saline or phosphate buffered saline (PBS). In a prefened embodiment, the active compounds are prepared with caniers 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 Coφoration.
Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) are also prefened 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. Patent No. 4,522,811 (which is incoφorated 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.
VII 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 ofl '-C-Branched Nucleosides l'-C-Branched ribonucleosides of the following structure:
Figure imgf000121_0001
wherein Base, R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, Y, W1, W2, W3, X, X1, X2 and X3 are as defined herein 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 substimted 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 l'-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.
Scheme 1
Figure imgf000122_0001
Activation
1) Coupling
2) Optional Deprotection
1) Optional Protection
2) Optional
Figure imgf000122_0002
Reduction
Figure imgf000122_0003
Optional Deprotection
Figure imgf000122_0004
2. Alternative method for the preparation ofl '-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), substimtion 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 l'-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.
Scheme 2
Figure imgf000123_0001
In addition, the L-enantiomers conesponding to the compounds of the invention can be prepared following the same general methods (1 or 2), beginning with the conesponding 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 imgf000123_0002
wherein Base, R1, R2, R3, R4, R5, R6, R7, R9, R10, Y, W1, W2, W3, X, X1, X2 and X3 are as defined herein 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 substimted 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* ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H2O2- ammonium molybdate, NaBr02-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.
Scheme 3
Figure imgf000125_0001
Protect on
1) Coupling
2) Optional Deprotection
1) Optional Protection
2) Optional
Figure imgf000125_0002
Figure imgf000125_0003
Reduction
Optional Deprotection
Figure imgf000125_0004
2. Modification of a pre- formed nucleoside
The key starting material for this process is an appropriately substimted 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, MhO* ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H202-ammonium molybdate, NaBr02-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.
Scheme 4
Figure imgf000126_0001
Optional Deprotection
ase
Figure imgf000126_0002
Optional Deprotection
Figure imgf000126_0003
In another embodiment of the invention, the L-enantiomers are desired. Therefore, the L-enantiomers can be conesponding to the compounds of the mvention can be prepared following the same foregoing general methods, beginning with the conesponding 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 imgf000127_0001
wherein Base, R1, R2, R3, R4, R5, R6, R7, R8, R9, Y, W1, W2, W3, X, X1, X2 and X3 are as defined herein 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, substimtion, 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* ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H202- ammonium molybdate, NaBr02-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 trimethylsilylfriflate 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.
Scheme 5
Figure imgf000128_0001
Protect on
1) Coupling
2) Optional Deprotection
1) Optional Protection
2) Optional
Figure imgf000128_0003
Reduction
tion
Figure imgf000128_0002
2. Modification of a pre-formed nucleoside
The key starting material for this process is an appropriately substimted 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 temperamre 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* rathenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H202-ammonium molybdate, NaBr02-CAN, NaOCl in HO Ac, 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.
Scheme 6
Figure imgf000130_0001
Optional Deprotection
1) Optional Protection
2) Optional
Figure imgf000130_0002
Reduction
on
Figure imgf000130_0003
In another embodiment of the invention, the L-enantiomers are desired. Therefore, the L-enantiomers can be conesponding to the compounds of the invention can be prepared following the same foregoing general methods, beginning with the conesponding L-sugar or nucleoside L-enantiomer as starting material. D. General Synthesis of 4 '-C-Branched Nucleosides
4'-C-Branched ribonucleosides of the following structure:
Figure imgf000130_0004
wherein Base, R1, R2, R3, R4, R5, R6, R7, Rs, R9, R10, Y, W1, W2, W3, X, X1, X2 and X3 are as defined herein can be prepared by one of the following general methods. 1. Modification from the pentodialdo-furanose
The key starting material for this process is an appropriately substituted pentodialdo- furanose. The pentodialdo-furanose can be purchased or can be prepared by any known means including standard epimerization, substimtion and cyclization techniques.
In a preferred embodiment, the pentodialdo-furanose is prepared from the appropriately substimted hexose. The hexose can be purchased or can be prepared by any known means including standard epimerization (e.g. via alkaline treatment), substimtion and coupling techniques. The hexose can be either in the furanose form, or cyclized via any means known in the art, such as methodology taught by Townsend Chemistry of Nucleosides and Nucleotides, Plenum Press, 1994, preferably by selectively protecting the hexose, to give the appropriate hexafuranose.
The 4'-hydroxymethylene of the hexafuranose then can be oxidized with the appropriate oxidizing agent in a compatible solvent at a suitable temperamre to yield the 4'- aldo-modified sugar. Possible oxidizing agents are Swern reagents, 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* ruthenium tetroxide, phase transfer catalysts such as chromic acid or permanganate supported on a polymer, Cl2-pyridine, H202-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, though preferably using H3PO , DMSO and DCC in a mixture of benzene/pyridine at room temperature.
Then, the pentodialdo-furanose 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. In the presence of a base, such as sodium hydroxide, the protected pentodialdo-furanose can then be coupled with a suitable electrophilic alkyl, halogeno-alkyl (i.e. CF3), alkenyl or alkynyl (i.e. allyl), to obtain the 4'-alkylated sugar. Alternatively, the protected pentodialdo-furanose can be coupled with the conesponding carbonyl, such as formaldehyde, in the presence of a base, such as sodium hydroxide, with the appropriate polar solvent, such as dioxane, at a suitable temperamre, which can then be reduced with an appropriate reducing agent to give the 4 '-alkylated sugar. In one embodiment, the reduction is canied out using PhOC(S)Cl, DMAP, preferably in acetonitrile at room temperamre, followed by treatment of ACCΝ and TMSS refluxed in toluene. 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 temperamre. 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 4 '-C-branched ribonucleoside is desired. Alternatively, deoxyribonucleoside is desired. To obtain these deoxyribo-nucleosides, a formed ribo-nucleoside 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.
In another embodiment of the invention, the L-enantiomers are desired. Therefore, the L-enantiomers can be conesponding to the compounds of the invention can be prepared following the same foregoing general methods, beginning with the conesponding L- pentodialdo-furanose as starting material. E. General Synthesis of 2 ' and/or 3 '-Prodrugs
The key starting material for this process is an appropriately substituted 1', 2', 3' or 4'-branched β-D or β-L nucleosides. The branched nucleoside can be purchased or can be prepared by any known means including the techniques disclosed herein. The branched nucleoside can be optionally protected with a suitable protecting group, preferably with a 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 branched nucleoside can then be coupled with a suitable acyl doner, such as an acyl chloride and/or an acyl anhydride with the appropriate protic or aprotic solvent at a suitable temperamre, to give the 2' and/or 3' prodrug of 1', 2', 3' or 4 '-branched β-D or β-L nucleoside. Alternatively, the protected branched nucleoside can then be coupled with a suitable acyl, such as a carboxylic acid, such as alkanoic acid and/or amino acid residue, optionally with a suitable coupling agent, with the appropriate aprotic solvent at a suitable temperature, to give the 2' and/or 3' prodrug of 1', 2', 3' or 4 '-branched β-D or β-L nucleoside. Possible coupling reagents are any reagents that promote coupling, including but are not limiting to, Mitsunobu reagents (e.g. diisopropyl azodicarboxylate and diethyl azodicarboxylate) with friphenylphosphine or various carbodiimides.
For example, simple amino-alcohols can be esterified using acid chlorides in refluxing acetonitrile-benzene mixture (See Scheme 7 below: Synthetic Communications, 1978, 8(5), 327-333; hereby incorporated by reference). Alternatively, esterification can be achieved using an anhydride, as described in J Am. Chem. Soc, 1999, 727(24), 5661-5664, which is hereby incoφortated by reference. See Figures 2, 3 and 4.
Scheme 7
Figure imgf000133_0001
ACN: acetonitrile The present invention is described by way of illustration, in the following examples. It will be understood by one of ordinary skill in the art that these examples are in no way limiting and that variations of detail can be made without departing from the spirit and scope of the present invention.
EXAMPLE 1 : PREPARATION OF 1 '-C-METHYLRIBOADENINE VIA 6-AMINO-9-(l-DEOXY-β-
D-PSICOFURANOSYL)PURINE
Melting points were determined on a Mel-temp II apparatus and are unconected. NMR spectra were recorded on a Bruker 400 AMX spectrometer at 400 MHz for 1H NMR and 100 MHz for 13C NMR with TMS as internal standard. Chemical shifts (δ) are reported in parts per million (ppm), and signals are reported as s (singlet), d (doublet), t (triplet), q
(quartet), m (multiplet), or bs (broad singlet). IR spectra were measured on a Nicolet 51 OP FT-IR spectrometer. Mass spectra were recorded on a Micromass Autospec high-resolution mass spectrometer. TLC were performed on Uniplates (silica gel) purchased from Analtech Co. Column chromatography was performed using either silica gel-60 (220-440 mesh) for flash chromatography or silica gel G (TLC grade, > 440 mesh) for vacuum flash column chromatography. UV spectra were obtained on a Beckman DU 650 spectrophotometer. Elemental analysis was performed by Atlantic Microlab, Inc., Norcross, GA, or Galbraith Laboratories, Inc., Knoxville, TN. HPLC was performed with a Waters HPLC system (Millipore Corporation, Milford, MA) equipped with a Model 600 controller, a Model 996 photodiode anay detector and a Model 717 plus autosampler. Millennium 2010 software was used for system control, data acquisition and processing. A chiralyser polarimetric detector, Perkin-Elmer Model 24 IMC polarimeter (Wilton, CT), was used for the determination of optical rotations.
Synthesis ofl '-C-methylribo-8-methyladenine
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-(l-deoxy-β-D-psicofuranosyl)purine" Collect. Czech. Chem. Commun. 1967, 32, 2663-2667; J. Farkas", Collect. Czech. Chem. Commun. 1966, 31, 1535) (Scheme 8).
Scheme 8
C
6-Benzamidopurine chloromercuri salt
Figure imgf000134_0002
Figure imgf000134_0001
Figure imgf000134_0003
In a similar manner, but using the appropriate sugar and purine bases, the following nucleosides of Formula XXIV are prepared.
Figure imgf000134_0004
(XXIV)
Figure imgf000134_0005
X vl1, - XV2, and Y are defined herein. Alternatively, the following nucleosides of Formula XXV are prepared, using the appropriate sugar and pyrimidine bases.
Figure imgf000135_0001
(XXV) wherein R »ι , R , R X A, - Xv-2, and Y are defined herein.
Alternatively, the following nucleosides of Formula XXVI are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000135_0002
(XXVI) wherein R , 1 , R , R , R , X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXVII are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000135_0003
(XXVII) wherein R .1 , R , R , X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXVIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000136_0001
(XXVIII) wherein R ,ι , R , R , X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXIX are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000136_0002
(XXIX) wherein
Figure imgf000136_0003
R°, R', R , X, R ,9y, τ R»10, and Base are defined herein.
EXAMPLE 2: PREPARATION OF 2'-C-METHYLRIBO-8-METHYLADENINE
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.Og. Chem. 1997, 62, 1754-1759) (Scheme 9).
Scheme 9
BzO
Figure imgf000136_0004
(a) Dess-Martin periodinane; (b) MeMgBr / TiCl ; (c) BzCl, DMAP, Et3N; (d) bis(trimethylsilyl)acetamide, N6-benzoyl adenine, TMSOTf; (e) NH3 /MeOH The 3 '-prodrug of the 2'-branched nucleoside was prepared according to published procedure (Synthetic Communications, 1978, 8(5), 327-333; J. Am. Chem. Soc, 1999, 121(24), 5661-5664). Alternatively, the 2'-branched nucleoside can be esterified without protection (Scheme 9b). Carbonyldiimidazole (377 mg, 2.33 mmol) was added to a solution of N-(tert-butoxycarbonyl)-L-valine (507 mg, 2.33 mmol) in 15 mL of anhydrous tetrahydrofuran. The mixmre was stirred at 20 °C for one hour and at 50 °C for 10 minutes and then added to a solution of 4-Amino-l-(3,4-dihydroxy-5-hydroxymethyl-3-methyl- tetrahydro-furan-2-yl)-7H-pyrimidine-2-one (500 mg, 1.95 mmol), 4- (dimethylamino)pyridine (25 mg, 0.195 mmol), friethylamine (5 mL) in anhydrous N,N- dimethylformamide (10 mL), which is also stirring at 50 °C. The reaction mixture was stined at 50 °C for one hour and then examined by ΗPLC. ΗPLC analysis indicated the formation of 52% of the desired ester, 17% of starting material in addition to undesired byproducts. The 3'-OΗ of 4-amino-l-(3,4-dihydroxy-5-hydroxymethyl-3-methyl-tefrahydro- furan-2-yl)-iH-pyrimidine-2-one tends to react selectively when coupled with BOC-Val.
In a similar manner, but using the appropriate sugar and purine bases, the following nucleosides of Formula XXX are prepared.
Figure imgf000137_0001
wherein R , 11, τ R>2 ,
Figure imgf000137_0002
X vl, v X2, and Y are defined herein.
Alternatively, the following nucleosides of Formula XXXI are prepared, using the appropriate sugar and pyrimidine bases.
Figure imgf000138_0001
(XXXI) wherein R ,1 , R , R , X Λ , X v2 , and Y are defined herein.
Alternatively, the following nucleosides of Formula XXXII are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000138_0002
(XXXII) wherein R1, R2, R3, R6, X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXXIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000138_0003
(XXXIII) wherein R1, R2, R6, X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXXIV are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000139_0001
(XXXIV) wherein R1, R2, R6, X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXXV are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000139_0002
(XXXV) wherein R »ι , R , R , R , R , 10 , X, and Base are defined herein.
EXAMPLE 3: PREPARATION OF 3'-C-METHYLRIBO-8-METHYLADENINE
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 10).
Scheme 10
Figure imgf000139_0003
(a) Ru02 / NaI04; (b) MeMgl / TiCl4; (c) HCl / MeOH / H20; (d) BzCl / pyridine; (e) AcBr, HBr / AcOH; (f) chloromercuri-6-benzamidopurine; (g) NH3 / MeOH. In a similar manner, but using the appropriate sugar and purine bases, the following nucleosides of Formula XXXVI are prepared.
Figure imgf000140_0001
(XXXVI) wherein R , R , R , X r\ , V X2 , and Y are defined herein.
Alternatively, the following nucleosides of Formula XXXVII are prepared, using the appropriate sugar and pyrimidine bases.
Figure imgf000140_0002
(XXXVII) wherein R1, R2, R3, X1, X2, and Y are defined herein.
Alternatively, the following nucleosides of Formula XXXVIII are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000140_0003
(XXXVIII) wherein R »ι , R , R , R , X, and Base are defined herein. Alternatively, the following nucleosides of Formula XXXLX are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000141_0001
(XXXIX) wherein R1, R2, R6, X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXXX are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000141_0002
(XXXX) wherein R , R , R , X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXXXI are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000141_0003
(XXXXI) wherein R1, R6, R7, R8, R9, X, and Baseare defined herein.
EXAMPLE 4: PREPARATION OF l-O-MUETHYL-2,3-O-ISOPROPYLIDENE-β-D-
RlBOFURANOSE - (1) The title compound can be prepared according to a published procedure (Leonard,
N. J.; Canaway, K. L. "5-Amino-5-deoxyribose derivatives. Synthesis and use in the preparation of "reversed" nucleosides" J. Heterocycl. Chem. 1966, 3, 485-489). A solution of 50.0 g (0.34 mole) of dry D-ribose in 1.0 L of acetone, 100 mL of 2,2- dimethoxypropane, 200 mL of methanol containing 20 mL of methanol saturated with hydrogen chloride at 0°C was stined overnight at room temperamre. The resulting solution was neutralized with pyridine and evaporated under reduced pressure. The resulting oil was 5 partitioned between 400 mL of water and 400 mL of methylene chloride. The water layer was extracted twice with methylene chloride (400 mL). The combined organic extracts were dried over sodium sulfate and evaporated under reduced pressure. The residue was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (1-2%) in methylene chloride] to give pure 1 (52.1 g, 75%) as a yellow syrup. 1H-NMR (CDC13): δ 10 5.00 (s, IH, H-l), 4.86 (d, IH, H-2, J2-3 = 5.9 Hz), 4.61 (d, IH, H-3, J3-2 = 5.9 Hz), 4.46 (t,
IH, H-4, J4-5 = 2.7 Hz), 3.77-3.61 (m, 2H, H-5 and H-5'), 3.46 (s, IH, OCH3), 3.0-2.4 (br s, IH, OH-5), 1.51 (s, 3H CH3), 1.34 (s, 3H CH3); MS (matrix GT): FAB>0 m/z 173 (M- OCH3)+.
EXAMPLE 5: PREPARATION OF l-O-METHYL-2,3-O-ISOPROPYLIDENE-β-D-
15 PENTODIALDO-RIBOFURANOSE - (2)
The title compound can be prepared according to a published procedure (Jones, G. H.; Moffatt, J. G. Oxidation of carbohydrates by the sulfoxide-carbodiimide and related methods. Oxidation with dicyclohexylcarbodiimide-DMSO, diisopropylcarbodiimide- DMSO, acetic anhydride-DMSO, and phosphorus pentoxide-DMSO: in Methods in
20 Carbohydrate Chemistry; Whisler, R. L. and Moffatt, J. L. Eds; Academic Press: New
York, 1972; 315-322).
Compound 1 was co-evaporated twice with anhydrous pyridine. Dicyclohexylcarbodi-imide (DCC, 137.8 g, 0.67 mol) was added to a solution of 1 (68.2 g, 0.33 mole) in anhydrous benzene (670 mL), DMSO (500 mL) and pyridine (13.4 mL). To
25. the resulting solution, cooled to 0°C, was added a solution of anhydrous crystalline orthophosphoric acid (16.4 g, 0.167 mmol) in anhydrous DMSO (30 mL). The mixture was stined for 1.5 hours at 0°C and 18 hours at room temperature under argon atmosphere, diluted with ethyl acetate (1000 mL). A solution of oxalic acid dihydrate (63.1 g, 038 mol) in DMSO (30 mL) was added and the reaction mixture was stined at room temperature
30 during 1 hour and then filtered to eliminate precipitated dicyclohexylurea (DCU). The filtrate was concentrated to a volume of about 600 mL under reduced pressure and neutralized with a saturated aqueous sodium hydrogen carbonate solution (400 mL). Brine (200 mL) was added and the organic layer was extracted with ethyl acetate (4x 1000 mL). The combined organic layers were concentrated to a volume of about 2000 mL, washed with a samrated aqueous sodium hydrogen carbonate solution (2x 700 mL), and with brine (2x 700 mL) before being dried over sodium sulfate and evaporated under reduced pressure. A small fraction of the crude residue was purified on silica gel chromatography [eluent: chloroform/ethyl ether, 8:2] in order to confirm the strucmre of 2 which was obtained as a pale yellow solid. 1H-NMR (CDC13): δ 9.61 (s, IH, H-5), 5.12 (s, IH, H-l), 5.08 (d, IH, H- 2, J2-3 = 5.9 Hz), 4.53 (d, IH, H-3, J3-2 = 6.0 Hz), 4.51 (s, IH, H-4), 3.48 (s, IH, OCH3), 1.56 (s, 3H CH3), 1.36 (s, 3H CH3); MS (matrix GT): FAB>0 m/z 203 (M+H)+, 171 (M- OCH3)+.
EXAMPLE 6: PREPARATION OF 4-C-HYDROXYMETHYL-1-O-METHYL-2,3-O-
ISOPROPYLIDENE-β-D-RIBOFURANOSE - (3)
The title compound can be prepared according to a published procedure (Leland, D. L.; Kotick, M. P. "Studies on 4-C-(hydroxymethyl)pentofuranoses. Synthesis of 9-[4-C- (hydroxymethyl)-a-L-threo-pentofuranosyl]adenine" Carbohydr. Res. 1974, 38, C9-C11;
Jones, G. H.; Taniguchi, M.; Tegg, D.; Moffatt, J. G. "4'-substituted nucleosides. 5. Hydroxylation of nucleoside 5'-aldehydes" J. Org. Chem. 1979, 44, 1309-1317; Gunic, E.; Girardet, J.-L.; Pietrzkowski, Z.; Esler, C; Wang, G. "Synthesis and cytotoxicity of 4'-C- and 5'-C-substituted Tovocamvcins" Bioorg. Med. Chem.2001. 9, 163-170). To a solution of the crude material (2) obtained above and 37% aqueous formaldehyde (167 mL) in dioxane (830 mL) was added aqueous sodium hydroxyde (2N, 300 mL). The mixture was stined at room temperamre for 4 hours and neutralized by addition of Dowex 50 W X 2 (H* form). The resin was filtered, washed with methanol, and the combined filtrates were concentrated to dryness and coevaporated several times with absolute ethanol. Sodium formate which was precipitated from absolute ethanol was removed by filtration, the filtrate was concentrated to dryness and the residue was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (0-4%) in chloroform] to give pure 3 (42.2 g, 54% from 1), which was recrystallized from cyclohexane. Mp = 94-95 (dec.) (lit.94-96.5; 97-98 : Refs :3,4), 1H-NMR (DMSO-d6): δ 4.65 (s, IH, H-l), 4.44-4.37 (m, 3H, H-2, H-3 and OH-6), 4.27 (t, IH, OH-5, J = 5.6 Hz, J =
6.0 Hz), 3.42-3.34 (m, 2H, H-5 and H-6) 3.29 (dd, IH, H-5*, J5'-OH = 5.4 Hz, J5-5' = 11.4 Hz), 3.11 (dd, IH, H-6', J6'-0H = 5.7 Hz, J6-6' = 10.9 Hz), 3.03 (s, 3H, OCH3), 1.48 (s, 3H CH3), 1.05 (s, 3H CH3); MS (matrix GT): FAB>0 m/∑ 469 (2M+H)+, 235 (M+H)+, 203 (M- OCH3)+ FABO m/z 233 (M-H)-.
EXAMPLE 7: PREPARATION OF 6-O-MONOMETHOXYTRITYL-4-C-HYDROXYMETHYL-1-
O-METHYL-2,3-O-ISOPROPYLIDENE-β-D-RIBOFURANOSE - (4) The title compound can be prepared according to a published procedure (Gunic, E.;
Girardet, J.-L.; Pietrzkowski, Z.; Esler, C; Wang, G. "Synthesis and cytotoxicity of 4'-C- and 5'-C-substituted Toyocamycins" Bioorg. Med. Chem.2001, 9, 163-170).
To a solution of 3 (41.0 g, 175 mmol) in pyridine (700 ml) was added by portions dimethoxytrityl chloride (60.5 g, 178 mmol) at +4°C. The reaction mixmre was stined for 3 hours at room temperature. After addition of methanol, the reaction mixture was concentrated (200 ml) and then dissolved with ethyl acetate (2 L). The organic layer was washed with a 5% aqueous sodium hydrogen carbonate solution, with water and dried over sodium sulfate and then evaporated to dryness. Purification by silica gel column chromatography [eluent: ethyl acetate / hexane 15/85] afforded pure 4 (63.0 g, 68%) as a syrup. 1H-NMR (CDC13): δ 7.5-6.9 (m, 13H, MMTr), 4.89 (s, IH, H-l), 4.72-4.62 (m, 3H,
H-2, H-3 and OH-5), 3.82 (dd, IH, H-5, J5-OH = 5.5 Hz, J5-5' = 10.5 Hz), 3.79 (s, 6H, OCH3), 3.54 (dd, IH, H-5', J5'-OH = 4.9 Hz, J5>-5 = 10.5 Hz), 3.31 (s, 3H, OCH3), 3.24 (d, IH, H-6, J6-6> = 9.2 Hz), 3.13 (d, IH, H-6', J6>-6 = 9.2 Hz), 1.24 (s, 3H CH3), 1.15 (s, 3H CH3); MS (matrix GT): FAB>0 m/z 303 (DMTr)+.
EXAMPLE 8: PREPARATION OF 5-O-BENZOYL-4-C-HYDROXYMETHYL-1-O-METHYL-
2,3-O-ISOPROPYLIDENE-β-D-RIBO-FURANOSE - (5)
The title compound can be prepared according to a published procedure (Gunic, E.; Girardet, J.-L.; Pietrzkowski, Z.; Esler, C; Wang, G. "Synthesis and cytotoxicity of 4'-C- and 5"-C-substituted Toyocamycins" Bioorg. Med. Chem. 2001, 9, 163-170). To a solution of 4 (2.51 g, 4.68 mmol) in anhydrous pyridine (37 mL) was added under argon benzoyl chloride (1.09 mL, 9.36 mmol) and the reaction mixmre was stined for 13 hours at to room temperature. Then the reaction was cooled to 0°C and stopped with ice- cold water (100 mL). The water layer was extracted with methylene chloride (3D 200 mL). The combined organic layers were washed with a saturated aqueous sodium hydrogen carbonate solution (2x 150 mL), with water (lx 150 mL) and then dried over sodium sulfate and evaporated under reduced pressure. The residue was dissolved in 80% acetic acid (70.2 mL) and the mixture was stined at room temperamre for 3hr and concentrated to dryness. Purification by silica gel column chromatography [eluent: chloroform] afforded pure 5 (1.40 g, 88%) as a syrup. 1H-NMR (CDC13): δ 8.1-7.4 (m, 5H, C6H5CO), 5.08 (s, IH, H-l), 4.77 (dd, 2H, H-2 and H-3, J = 6.1 Hz, J = 8.2 Hz), 4.51 (q, 2H, H-5 and H-5', J = 11.5 Hz, J5-5' = 23.8 Hz), 3.91 (t, 2H, H-6 and H-6', J = 12.3 Hz), 4.38 (s, IH, OCH3), 2.2-1.8 (brs, IH,
OH-6), 1.57 (s, 3H CH3), 1.38 (s, 3H CH3); MS (matrix GT): FAB>0 m/z 677 (2M+H)+, 339 (M+H)+, 307 (M-OCH3)+, 105 (C6HsCO)+ FAB<0 m/z 121 (C6H5CO2)-.
EXAMPLE 9: PREPARATION OF 5-O-BENZOYL-4-C-METHYL-1-O-METHYL-2,3-O-
ISOPROPYLIDENE-β-D-RIBOFURANOSE - (6) The title compound can be prepared according to a published procedure (Gunic, E.;
Girardet, J.-L.; Pietrzkowski, Z.; Esler, C; Wang, G. "Synthesis and cytotoxicity of 4'-C- and 5'-C-substituted Toyocamycins" Bioorg. Med. Chem. 2001, 9, 163-170).
A solution of 5 (37.6 g, 0.111 mol), 4-dimethylaminopyridine (DMAP, 40.7 g, 0.333 mol) and phenoxythiocarbonyle chloride in anhydrous acetonitrile (1000 mL) was stined at room temperamre for 1 hour and concentrated to dryness. The residue was dissolved in methylene chloride (500 mL) and successively washed with 0.2 M hydrochloric acid (2x 500 mL) and water (500 mL) before being dried over sodium sulfate, evaporated under reduced pressure and coevaporated several times with anhydrous toluene. The crude material was dissolved in anhydrous toluene (880 mL) and tris(trimethylsilyl)silane (TMSS, 42.9 mL, 0.139 mol), and l,l'-azobis(cyclohexanecarbonitrile) (ACCN, 6.8 g, 27.8 mmol) were added. The reaction mixture was stined under reflux for 45 minutes, cooled to room temperature and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography [eluent: stepwise gradient of diethyl ether (5-20%) in petroleum ether] to give pure 6 (26.4 g, 74%) as a pale yellow syrup. 'H-NMR (DMSO-d6): δ 8.0-7.5 (m, 5H, C6H5CO), 4.85 (s, IH, H-l), 4.63 (dd, 2H, H-2 and H-3, J = 6.1 Hz, J =
11.6 Hz), 4.24 (d, IH, H-5, J5.5' = 11.1 Hz), 4.10 (d, IH, H-5', J5.-5 = 11.1 Hz), 3.17 (s, IH, OCH3), 1.38 (s, 3H CH3), 1.30 (s, 3H CH3), 1.25 (s, 3H CH3); MS (matrix GT): FAB>0 m/z 291 (M-OCH3)+, 105 (C6H5CO)+ FABO m/z 121 (C6H5C02)". EXAMPLE 10: PREPARATION OF 5-O-BENZOYL-4-C-METHYL-l,2,3-O-ACETYL-0t,β-D-
RIBOFURANOSE - (7)
Compound 6 (22.5 g, 70 mmol) was suspended in a 80% aqueous acetic acid solution (250 mL). The solution was heated at 100°C for 3 hours. The volume was then reduced by half and coevaporated with absolute ethanol and pyridine. The oily residue was dissolved in pyridine (280 mL) and then cooled at 0°C. Acetic anhydride (80 mL) and 4- dimethylamino-pyridine (500 mg) were added. The reaction mixmre was stined at room temperature for 3 hours and then concentrated under reduced pressure. The residue was dissolved with ethyl acetate (1 L) and successively washed with a saturated aqueous sodium hydrogen carbonate solution, a 1 M hydrochloric acid and water. The organic layer was dried over sodium sulfate and evaporated under reduced pressure. The resulting residue was purified by silica gel column chromatography [eluent: stepwise gradient of diethyl ether (30-40%) in petroleum ether] to give pure 7 (16.2 g, 60%) as a pale yellow syrup. A small fraction of the material was re-purified on silica gel chromatography [same eluent: system] in order separate the α and the β anomers. α anomer: 1H-NMR (DMSO-d6): δ 8.1-7.5 (m, 5H, C6H5CO), 6.34 (pt, IH, H-l, J = 2.4 Hz, J = 2,1 Hz), 5.49 (m, 2H, H-2 and H-3), 4.33 (q, 2H, H-5 and H-5', J = 11.6 Hz, J = 18.7 Hz), 2.15 (s, 3H, CH3CO2), 2.11 (s, 3H, CH3CO2), 2.07 (s, 3H, CH3CO2), 1.37 (s, 3H, CH3); MS (matrix GT): FAB>0 m/z 335 (M-CH3CO2 +, 275 (M-CH3CO2AH)+,105 (C6H5CO)+, 43 (CH3CO)+ FAB<0 m/z 121 (C6H5CO2)\ 59 (CH3CO2)\ β anomer: 1H-NMR (DMSO-d6): δ 8.1-7.5 (m, 5H, C6H5CO), 5.99 (s, IH, H-l), 5.46 (d, IH, H-2, J2-3 = 5.3 HZ), 5.30 (d, IH, H-2, J2-3 = 5.3 Hz), 4.39 (d, IH, H-5, J5-5> = 11.7 Hz), 4.19 (d, IH, H-5', J5 -5 = 11.7 Hz), 2.10 (s, 3H, CH3CO2), 2.06 (s, 3H, CH3CO2), 2.02 (s, 3H, CH3CO2), 1.30 (s, 3H, CH3); MS (matrix GT): FAB>0 m/z 335 (M-CH3CO2-)+, 275 (M- CH3CO2-+H)+, 105 (C6H5CO)+, 43 (CH3CO)+ FAB<0 m/z 121 (C6H5CO2)-3 59 (CH3CO2)-.
EXAMPLE 11: PREPARATION OF O-6-DlPHENYLCARBAMOYL-N2-ISOBUTYRYL-9-(2,3- DI-O-ACETYL-5-O-BENZOYL-4-C-METHYL-β-D-RIBOFURANOSYL)-8-METHYLGUANINE -
(18)
To a suspension of O-6-diphenylcarbamoyl-8-methyl-N2-isobutyrylguanine in anhydrous toluene (20 mL) was added N,O-bis(trimethylsilyl)acetamide (1.92 mL, 7.9 mmol). The reaction mixmre was allowed to warm under reflux for 1 hour. Compound 7
(1.55 g, 3.93 mmol) was dissolved in toluene (10 mL) and trimethylsilyltrifluoro- methanesulfonate (TMSTf) (915 mL, 4.72 mmol) was added. The mixmre was heated under reflux for 30 minutes. The solution was then cooled to room temperamre and neutralized with a 5% aqueous sodium hydrogen carbonate solution. The reaction mix re was diluted with ethyl acetate (200 mL). The organic phase was washed with a 5% aqueous sodium hydrogen carbonate solution (150 mL) and with water (2x 150 mL). The organic layer was dried over Na2SO and evaporated to dryness. The residue was purified by silica gel column chromatography [eluent: stepwise gradient of diethyl ether (70-90%) in petroleum ether] to afford 18.
EXAMPLE 12: PREPARATION OF 9-(4-C-METHYL-β-D-RIBOFURANOSYL)-8- METHYLGUANINE - (19)
The title compound can be prepared according to a published procedure from 18 (Waga, T.; Nishizaki, T.; Miyakawa, I.; Orhui, H.; Meguro, H. "Synthesis of 4'-C- methvlnucleosides" Biosci. Biotechnol. Biochem. 1993. 57, 1433-1438).
A solution of 18 in methanolic ammonia (previously saturated at -10°C) (20 mL) was stined at room temperature overnight. The solvent was evaporated under reduced pressure and the residue was partitioned between methylene chloride (60 mL) and water (60 mL). The aqueous layer was washed with methylene chloride (2x 60 mL), concentrated under reduced pressure. The residue was purified by an RP18 column chromatography [eluent water/acetonitrile 95/5] to afford 19.
EXAMPLE 13: 9-(2,3-DI-O-ACETYL-5-O-BENZOYL-4-C-METHYL-β-D-RIBOFURANOSYL)-
8-METHYLADENINE - (20)
A solution of 7 (1.10 g, 2.79 mmol) in anhydrous acetonitrile (50 ml) was treated with 8-methyladenine and stannic chloride (SnCl4, 660 μL, 5.58 mmol) and stined at room temperature overnight. The solution was concentrated under reduced pressure, diluted with chloroform (100 mL) and treated with a cold saturated aqueous solution of NaHCO3 (100 ml). The mixture was filtered on celite, and the precipitate was washed with hot chloroform. The filtrates were combined, washed with water (100 ml) and brine (100 ml), dried (Na2SO4), and evaporated under reduced pressure. The residue was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (3-5%) in dichloromethane] to afford 20. EXAMPLE 14: PREPARATION OF 9-(4-C-METHYL-β-D-RIBOFURANOSYL)-8-
METHY ADENINE - (21)
The title compound can be prepared according to a published procedure from 20 (Waga, T.; Nishizaki, T.; Miyakawa, I.; Orhui, H.; Meguro, H. "Synthesis of 4'-C- methvlnucleosides" Biosci. Biotechnol. Biochem. 1993. 57, 1433-1438).
A solution of 20 in methanolic ammonia (previously saturated at -10°C) (50 mL) was stirred at room temperature overnight. The solvent was evaporated under reduced pressure and the residue was partitioned between methylene chloride (100 ml) and water (100 ml). The aqueous layer was washed with methylene chloride (2x 100 mL), and concentrated under reduced pressure. The residue was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (10-30%) in ethyl acetate] to afford 21.
In a similar manner, but using the appropriate sugar and purine bases, the following nucleosides of Formula XXXXII are prepared.
Figure imgf000148_0001
(XXXXII) wherein R .11, Rz, -
Figure imgf000148_0002
Xv-1,
Figure imgf000148_0003
and Y are defined herein.
EXAMPLE 15: PREPARATION OF l-(5-O-BENZOYL-4-C-METHYL-2,3-O-ACETYL-β-D- RIBOFURANOSYL)-6-METHYLURACIL - (8)
A suspension of 6-methyluracil was treated with hexamethyldisilazane (HMDS, 21 mL) and a catalytic amount of ammonium sulfate during 17 hours under reflux. After cooling to room temperature, the mixture was evaporated under reduced pressure, and the residue, obtained as a colorless oil, was diluted with anhydrous 1,2-dichloroethane (7.5 mL). To the resulting solution was added 7 (0.99 g, 2.51 mmol) in anhydrous 1,2- dichloroethane (14 mL), followed by addition of trimethylsilyl frifluoromethanesulfonate (TMSTf, 0.97 mL, 5.02 mmol). The solution was stined for 2.5 hours at room temperature under argon atmosphere, then diluted with chloroform (150 mL), washed with the same volume of a samrated aqueous sodium hydrogen carbonate solution and finally with water (2x 100 mL). The organic phase was dried over sodium sulfate, then evaporated under reduced pressure. The resulting crude material was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (0-2%) in chloroform] to afford pure 8.
EXAMPLE 16: PREPARATION OF l-(4-C-METHYL-β-D-RIBOFURANOSYL)-6-
METHYLURACIL - (9)
The title compound can be prepared according to a published procedure from 8 (Waga, T.; Nishizaki, T.; Miyakawa, I.; Orhui, H.; Meguro, H. "Synthesis of 4'-C- methylnucleosides" Biosci. Biotechnol. Biochem. 1993, 57, 1433-1438).
A solution of 8 in methanolic ammonia (previously samrated at -10°C) (27 mL) was stined at room temperamre overnight. The solvent was evaporated under reduced pressure and the residue was partitioned between methylene chloride (40 mL) and water (40 mL). The aqueous layer was washed with methylene chloride (2x 40 mL), concentrated under reduced pressure and coevaporated several times with absolute ethanol. Recrystallization from a mixmre absolute ethanol/methanol gave 9.
EXAMPLE 17: PREPARATION OF l-(5-O-BENZOYL-4-C-METHYL-2,3-O-ACETYL-β-D- RIBOFTJRANOSYL)-4-THIO-6-METHYL-URACIL - (10) Lawesson's reagent (926 mg, 2.29 mmol) was added under argon to a solution of 8 in anhydrous 1,2-dichloroethane (65 mL) and the reaction mixmre was stirred overnight under reflux. The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (1-2%) in chloroform] to give pure 10.
EXAMPLE 18: PREPARATION OF l-(4-C-METHYL-β-D-RIBOFURANOSYL)-4-THIO-6-
METHYLURACIL - (11)
A solution of 10 in methanolic ammonia (previously saturated at -10°C) (27 mL) was stined at room temperamre overnight. The solvent was evaporated under reduced pressure and the residue was partitioned between methylene chloride (40 ml) and water (40 mL). The aqueous layer was washed with methylene chloride (2x 40 mL), concentrated under reduced pressure. The crude material was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (5-7%) in methylene chloride] to give 11, which was lyophilized.
EXAMPLE 19: PREPARATION OF l-(4-C-METHYL-β-D-RIBOFURANOSYL)-6- METHYLCYTOSINE, HYDROCHLORIC FORM - (12)
Compound 11 was treated with methanolic ammonia (previously saturated at - 10°C), (12 mL) at 100°C in a stainless-steel bomb for 3 hours, then cooled to room temperature. The solvent was evaporated under reduced pressure and the residue was partitioned between methylene chloride (40 mL) and water (40 mL). The aqueous layer was washed with methylene chloride (2x 40 mL), concentrated under reduced pressure. The crude material was purified by silica gel column chromatography [eluent: methylene chloride/ methanol/ammonium hydroxide 65:30:5]. The collected fractions were evaporated under reduced pressure and in absolute ethanol (6.3 mL). To the solution was added a 2N hydrochloric acid solution (1.5 mL) and the mixmre was stined before being concentrated under reduced pressure. The procedure was repeated twice and 12 was precipitated from absolute ethanol.
EXAMPLE 20: PREPARATION OF l-(5-O-BENZOYL-4-C-METHYL-2,3-O-ACETYL-β-D- RIBOFURANOSYL)-6-METHYLTHYMINE - (13)
A suspension of 6-methylthymine was treated with hexamethyldisilazane (HMDS, 17 mL) and a catalytic amount of ammonium sulfate overnight under reflux. After cooling to room temperature, the mixture was evaporated under reduced pressure, and the residue, obtained as a colorless oil, was diluted with anhydrous 1,2-dichloroethane (6 mL). To the resulting solution was added 7 (1.0 g, 2.53 mmol) in anhydrous 1,2-dichloroethane (14 mL), followed by addition of trimethylsilyl trifluoromethanesulfonate (TMSTf, 0.98 mL, 5.06 mmol). The solution was stined for 5 hours at room temperamre under argon atmosphere, then diluted with chloroform (150 mL), washed with the same volume of a saturated aqueous sodium hydrogen carbonate solution and finally with water (2x 100 mL). The organic phase was dried over sodium sulfate, then evaporated under reduced pressure. The resulting crude material was purified by silica gel column chromatography [eluent: 2% of methanol in chloroform] to afford pure 13. EXAMPLE 21: PREPARATION OF l-(4-C-METHYL-β-D-RIBOFURANOSYL)-6-
METHYLTHYMINE - (14)
The title compound can be prepared according to a published procedure from 13 (Waga, T.; Nishizaki, T.; Miyakawa, I.; Orhui, H.; Meguro, H. "Synthesis of 4'-C- methylnucleosides" Biosci. Biotechnol. Biochem. 1993, 57, 1433-1438).
A solution of 13 in methanolic ammonia (previously saturated at -10°C) (60 mL) was stined at room temperature overnight. The solvent was evaporated under reduced pressure and the residue was partitioned between methylene chloride (60 mL) and water (60 mL). The aqueous layer was washed with methylene chloride (2x 60 mL), concentrated under reduced pressure and coevaporated several times with absolute ethanol.
Recrystallization from methanol gave 14.
EXAMPLE 22: PREPARATION OF l-(5,2,3-TM-O-ACETYL-4-C-METHYL-β-D- RIBOFURANOSYL)-6-METHYLTHYMINE - (15)
A solution of 14 in anhydrous pyridine (7.4 mL) was treated with acetic anhydride (1.2 mL) and stined at room temperamre for 3 hours. The solvent was evaporated under reduced pressure, and the residue was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (0-5%) in methylene chloride] to afford 15.
EXAMPLE 23: PREPARATION OF l-(5,2,3-TRI-O-ACETYL-4-C-METHYL-β-D- RIBOFURANOSYL)-4-THIO-6-METHYLTHYMINE - (16) Lawesson's reagent (119 mg, 0.29 mmol) was added under argon to a solution of 15 in anhydrous 1,2-dichloroethane (11 mL) and the reaction mixmre was stirred overnight under reflux. The solvent was evaporated under reduced pressure and the residue was purified by silica gel column chromatography [eluent: stepwise gradient of methanol (1-2%) in chloroform] to give 16.
EXAMPLE 24: PREPARATION OF l-(4-C-METHYL-β-D-RIBOFΪJRANOSYL)-5-METHYL-6-
METHYLCYTOSINE - (17), HYDROCHLORIDE FORM
Compound 16 was treated with methanolic ammonia (previously saturated at -
10°C), (10 mL) at 100°C in a stainless-steel bomb for 3 hours, then cooled to room temperature. The solvent was evaporated under reduced pressure and the residue was partitioned between methylene chloride (30 mL) and water (30 mL). The aqueous layer was washed with methylene chloride (2x 30 mL), concentrated under reduced pressure. The crude material was purified by silica gel column chromatography [eluent: 20% methanol in methylene chloride] to afford 17. This compound was dissolved in EtOH 100 (1.5 mL), treated with a 2N hydrochloric acid solution (0.3 mL), and the mixture was stined before being concenfrated under reduced pressure. The procedure was repeated twice and 17 was precipitated from absolute ethanol.
Alternatively, the following nucleosides of Formula XXXXIII are prepared, using the appropriate sugar and pyrimidine bases.
Figure imgf000152_0001
(XXXXIII) wherein R1, R2, R3, X1, X2, and Y are defined herein.
Alternatively, the following nucleosides of Formula XXXXIV are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000152_0002
(XXXXIV) wherein R ► 1 , r R>2 , T R)3 , T R>6 , X, and Base are defined herein. Alternatively, the following nucleosides of Formula XXXXV are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000153_0001
(XXXXV) wherein R »ι , R , R , X, and Base are defined herein.
Alternatively, the following nucleosides of Formula XXXXVI are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000153_0002
(XXXXVI) wherein R1, R2, R6, X, and Base are defined herein
Alternatively, the following nucleosides of Formula XXXXVII are prepared, using the appropriate sugar and pyrimidine or purine bases.
Figure imgf000153_0003
(XXXXVII) wherein R ( 11, r R>6°, r R.7', τ τ>1
Figure imgf000153_0004
R.9y, Rι0υ, X, and Base are defined herein. VIII. Biological Assays
A number of assays are available to determine the potency of test compounds against viruses. Several of these biological assays are described in the examples below.
EXAMPLE 25: ANTI-FLAVIVIRUS ORPESTΓVIRTJS 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.
Phosphorylation Assay of Nucleoside to Active Triphosphate To determine the cellular metabolism of the compounds, HepG2 cells are obtained from the American Type Culture Collection (Rockville, MD), and are grown in 225 cm2 tissue culmre flasks in minimal essential medium supplemented with non-essential amino acids, 1% penicillin-streptomycin. The medium is renewed every three days, and the cells are 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 are seeded at a density of 2.5 x 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 are maintained at 37°C under a 5% CO2 atmosphere. At the selected time points, the cells are washed three times with ice-cold phosphate-buffered saline (PBS). Intracellular active compound and its respective metabolites are 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 are then combined, dried under gentle filtered air flow and stored at -20°C until HPLC analysis.
Bioavailability Assay in Cynomolgus Monkeys
Within 1 week prior to the study initiation, the cynomolgus monkey is 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) receives approximately 250 μCi of 3H activity with each dose of active compound 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 is weighed before dosing to gravimetrically determine the quantity of formulation administered. Urine samples are 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 are 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 are analyzed for the maximum concentration (Cmax), time when the maximum concentration is achieved (Tmax), area under the curve (AUC), half life of the dosage concentration (Tι/2), clearance (CL), steady state volume and distribution (Vss) and bioavailability (F). Bone Marrow Toxicity Assay
Human bone manow cells are collected from normal healthy volunteers and the mononuclear population are separated by Ficoll-Hypaque gradient centrifugation as described previously by Sommadossi J-P, Carlisle R. "Toxicity of 3'-azido-3'- deoxythymidine and 9-(l,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 RF, Chu CK, Xie M-Y. "Comparison of cytotoxicity of the (-)- and (+)-enantiomer of 2',3'-dideoxy-3'-thiacytidine in normal human bone manow progenitor cells" Biochemical Pharmacology 1992; 44:1921-1925. The culmre assays for CFU-GM and BFU-E are performed using a bilayer soft agar or methylcellulose method. Drugs are diluted in tissue culmre 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 are counted using an inverted microscope. The results are presented as the percent inhibition of colony formation in the presence of drug compared to solvent control cultures. Mitochondria Toxicity Assay HepG2 cells are 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 VM. "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 are measured using a Boehringer lactic acid assay kit. Lactic acid levels are normalized by cell number as measured by hemocytometer count. Cytotoxicity Assay
Cells are seeded at a rate of between 5 x 103 and 5 x 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 is then added. After incubation for 4 days, cultures are fixed in 50% TCA and stained with sulforhodamineB. The optical density was read at 550 nm. The cytotoxic concenfration was expressed as the concentration required to reduce the cell number by 50% (CC50).
Cell Protection Assay (CPA) The assay is 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) are 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 are added to both infected and uninfected cells in a final concentration of 0.5% DMSO in growth medium. Each dilution is tested in quadruplicate. Cell densities and virus inocula are 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 are fixed with 50% TCA and stained with sulforhodamine B. The optical density of the wells is read in a microplate reader at 550 nm. The 50% effective concentration (EC50) values are defined as the compound concentration that achieved 50% reduction of cytopathic effect of the virus.
Plaque Reduction Assay
For each compound the effective concentration is determined in duplicate 24-well plates by plaque reduction assays. Cell monolayers are 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 are added to the monolayers. Cultures are further incubated at 37°C for 3 days, then fixed with 50% ethanol and 0.8% Crystal Violet, washed and air-dried. Then plaques are counted to determine the concentration to obtain 90% virus suppression. Yield Reduction Assay
For each compound the concenfration to obtain a 6-log reduction in viral load is determined in duplicate 24-well plates by yield reduction assays. The assay is 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 are seeded onto 24-well plates (2 x 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 are added to cells in a final concentration of 0.5% DMSO in growth medium. Each dilution is tested in triplicate. After three days, cell cultures (cell monolayers and supematants) are lysed by three freeze-thaw cycles, and virus yield is quantified by plaque assay. Briefly, MDBK cells are seeded onto 6-well plates (5 x 105 cells per well) 24 h before use. Cells are 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 are fixed with 3.5% formaldehyde and stained with 1% crystal violet (w/v in 50% ethanol) to visualize plaques. The plaques are counted to determine the concentration to obtain a 6-log reduction in viral load.
EXAMPLE 26: IN VITRO ANTI- VIRAL ACTIVITY
In vitro anti-viral activity was tested in the following cell lines: MT-4 for HIV; Vero 76, African green monkey kidney cells for SARS; BHK for Bovine Viral Dianhea Virus;
Sb-1 for poliovirus Sabin type-1; CVB-2, CVB-3, CVB-4, and CVA-9 for Coxsackieviruses B-2, B-3, B-4 and A-9; and REO-1 for double-stranded RNA viruses. Note: BVDV = bovine viral dianhea virus; YFV = yellow fever virus; DENV = dengue virus; WNV = West Nile virus; CVB-2 = Coxsackie B-2 virus; Sb-1 = Sabin type 1 poliomyelitis virus; and REO = double-stranded RNA Reovirus.
CC and EC Test Results for β-D-2'-C-methyl-7-methyl-6-phenyl-3.3a.5.8a-tetrahvdro- 1.3,4,5,7a-penta-aza-.s-indacen-8-one (Compound F)
Figure imgf000157_0001
CC Test Results for β-D-2'-C-methyl-7-methyl-6-phenyl-3.3a,5.8a-tefrahydro-L3A5.7a- penta-aza-.s-indacen-8-one (Compound F)
Figure imgf000158_0001
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

CLAIMSWhat is claimed is:
1. A compound of Formula (I):
Figure imgf000159_0001
(I) or a pharmaceutically acceptable salt thereof, wherein:
R1, R2 and R3 are independently H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl, CO-aryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO-substituted aryl, sulfonate ester; benzyl, wherein the phenyl group is optionally substituted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; an amino acid residue; a carbohydrate; a peptide; cholesterol; or pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 and/or R3 is independently H or phosphate; wherein at least one of R2 and R3 is not hydrogen; and wherein:
Y1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR4, NH2, NHR4, NR4R5, SH or SR4;
X1 is a straight chained, branched or cyclic optionally substimted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substimted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO-Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; and
X2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substimted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO-Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; and wherein each Y3 is independently H, F, Cl, Br or I; and eeaacchh ! R4 and R5 is independently hydrogen, acyl, alkyl, lower alkyl, alkenyl, alkynyl or cycloalkyl
A compound of Formula (II):
Figure imgf000160_0001
(II) or a pharmaceutically acceptable salt thereof, wherein:
R1, R2 and R3 are independently H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl, CO-aryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO-substituted aryl, sulfonate ester; benzyl, wherein the phenyl group is optionally substituted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; an amino acid residue; a carbohydrate; a peptide; cholesterol; or pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 and/or R3 is independently H or phosphate; wherein at least one of R2 and R3 is not hydrogen; and wherein:
Y1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR4, NH2, NHR4, NR4R5, SH or SR4;
X1 is a straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substimted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO-Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; and
X2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3,
2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substituted alkenyl, optionally substimted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO-Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; and wherein each Y3 is independently H, F, Cl, Br or I; and each R4 and R5 is independently hydrogen, acyl, alkyl, lower alkyl, alkenyl, alkynyl or cycloalkyl.
3. A compound of Formula (III), (IV) or (V):
Figure imgf000161_0001
(III) (IV) (V) or a pharmaceutically acceptable salt thereof, wherein:
R1, R2 and R3 are independently H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl, CO-aryl, CO-alkoxyalkyl, CO-aryloxyalkyl, CO-substituted aryl, sulfonate ester; benzyl, wherein the phenyl group is optionally substituted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; an amino acid residue; a carbohydrate; a peptide; cholesterol; or pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 and/or R3 is independently H or phosphate; wherein at least one of R2 and R3 is not hydrogen; and wherein: Base is selected from the group consisting of
Figure imgf000161_0002
(A) (B)
Figure imgf000162_0001
(C) (D) (E) (F)
Figure imgf000162_0002
(G) (H)
Figure imgf000162_0003
(I) (J) (K) (L)
Figure imgf000162_0004
(M) (N)
Figure imgf000163_0001
(O) (P) (Q) (R)
Figure imgf000163_0002
(S) (T)
Figure imgf000163_0003
(U) (V) (W) (X)
Figure imgf000163_0004
00 (Z)
Figure imgf000164_0001
(AA) (AB) (AC) (AD)
Figure imgf000164_0002
(AE) (AF)
Figure imgf000164_0003
(AG) (AH) (Al) (AJ
Figure imgf000164_0004
(BA) (BB)
Figure imgf000165_0001
(BC) (BD) (BE) (BF)
Figure imgf000165_0002
(BG) (BH)
Figure imgf000165_0003
(Bl) (BJ) (BK) (BL)
Figure imgf000165_0004
(BM) (BN)
Figure imgf000166_0001
(BO) (BP) (BQ) (BR)
Figure imgf000166_0002
(BS) (BT)
Figure imgf000166_0003
(BU) (BV) (BW) (BX)
Figure imgf000166_0004
(BY) (BZ)
Figure imgf000167_0001
(BAA) (BAB) (BAG) (BAD)
Figure imgf000167_0002
(BAE) (BAF)
each R4 and R5 is independently hydrogen, acyl, alkyl, lower alkyl, alkenyl, alkynyl or cycloalkyl; each W1, W2, W3 and W4 is independently N, CH, CF, CI, CBr, CCl, CCN, CCH3, CCF3, CCH2CH3, CC(0)NH2, CC(0)NHR4, CC(0)N(R4)2, CC(0)OH, CC(0)OR4 or CX3; each W* is independently O, S, NH or NR4;
X is O, S, S02, CH2, CH2OH, CHF, CF2, C(Y3)2, CHCN, C(CN)2, CHR4 or C(R4)2;
X* is CH, CF, CY3 or CR4;
X2 is H, straight chained, branched or cyclic optionally substimted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substituted alkenyl, optionally substituted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO-Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; each X3 is independently a straight chained, branched or cyclic optionally substituted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, N3, CN, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, OH, OR4, -O(acyl), -O(lower acyl), -O(alkyl), -O(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), - S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), chloro, bromo, fluoro, iodo, NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), or -N(acyl)2; each Y is independently selected from the group consisting of H, optionally substituted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3,CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH2, (CH2)mCONR2, and (CH2)mCONHR; R is H, alkyl or acyl;
Y1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR4, NH2, NHR4, NR R5, SH or SR4; each Y2 is independently O, S, NH or NR4; and each Y3 is independently H, F, Cl, Br or I; wherein for Base (B), W4 cannot be CH if W1, W2 and W3 are N; wherein for Base (E), (F), (K), (L), (W) and (X), W4 cannot be CH if W1 is N; each R6 is independently an optionally substimted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, -CH2C(O)OH, -CH2C(0)OR4,
-CH2C(0)O(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(0)N(R4)2, -CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(0)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(0)NH2, -C(0)NHR4, -C(0)NH(lower alkyl), -C(0)N(R4)2,
-C(O)N(lower alkyl)2 or cyano; each R7 is independently OH, OR2, optionally substimted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2- Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, optionally substimted carbocycle, optionally substimted heterocycle, optionally substituted heteroaryl, -CH2C(0)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(0)S(lower alkyl), -CH2C(0)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(0)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(0)S(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(0)0(lower alkyl), -C(O)SH, -C(O)SR4, -C(O)S(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, -O(acyl), -O(lower acyl), -O(R4), -O(alkyl), -O(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), NO2, NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo; alternatively, R6 and R7 can come together to form a spiro compound selected from the group consisting of optionally substimted carbocycle or optionally substituted heterocycle; and each m is independently 0, 1 or 2.
4. A compound of Formula (VI) or (VII):
Figure imgf000169_0001
(VI) (VII) or a pharmaceutically acceptable salt thereof, wherein:
R1 is H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl; CO- aryl; CO-alkoxyalkyl; CO-aryloxyalkyl; CO-substituted aryl; sulfonate ester; benzyl, wherein the phenyl group is optionally substimted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; an amino acid residue; a carbohydrate; a peptide; cholesterol; or pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1 is H or phosphate; and wherein:
Base is selected from the group consisting of
Figure imgf000170_0001
(B)
Figure imgf000170_0002
(G) (H)
Figure imgf000170_0003
(I) (J) (K) (L)
Figure imgf000171_0001
(M) (N)
Figure imgf000171_0002
(O) (P) (Q) (R)
Figure imgf000171_0003
(S) (T)
Figure imgf000171_0004
(U) (V) (W) (X)
Figure imgf000172_0001
(Y) (Z)
Figure imgf000172_0002
(AA) (AB) (AC) (AD)
Figure imgf000172_0003
(AE) (AF)
Figure imgf000172_0004
(AG) (AH) (Al) (AJ)
Figure imgf000173_0001
(BA) (BB)
Figure imgf000173_0002
(BC) (BD) (BE) (BF)
Figure imgf000173_0003
(BG) (BH)
Figure imgf000173_0004
(Bl) (BJ) (BK) (BL)
Figure imgf000174_0001
(BM) (BN)
Figure imgf000174_0002
(BO) (BP) (BQ) (BR)
Figure imgf000174_0003
(BU) (BV) (BW) (BX) 2004
Figure imgf000175_0001
(BAA) (BAB) (BAC) (BAD)
Figure imgf000175_0002
(BAE) (BAF)
each R4 and R5 is independently hydrogen, acyl, alkyl, lower alkyl, alkenyl, alkynyl or cycloalkyl; each W1, W2, W3 and W4 is independently N, CH, CF, CI, CBr, CCl, CCN, CCH3, CCF3, CCH2CH3, CC(O)NH2, CC(O)NHR4, CC(O)N(R4)2, CC(O)OH, CC(0)OR4 or CX3; each W* is independently O, S, NH or NR4;
X is O, S, SO2, CH2, CH2OH, CHF, CF2, C(Y3)2, CHCN, C(CN)2, CHR4 or C(R4)2;
X* is CH, CF, CY3 or CR4;
X2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substituted alkenyl, optionally substimted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO-Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; each X3 is independently a straight chained, branched or cyclic optionally substituted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, N3, CN, -C(0)OH, -C(0)OR4, -C(0)0(lower alkyl), -C(0)NH2, -C(0)NHR4, -C(0)NH(lower alkyl), -C(0)N(R4)2, -C(0)N(lower alkyl)2, OH, OR4, -O(acyl), -0(lower acyl), -O(alkyl), -0(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), - S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl),
-S(cycloalkyl), chloro, bromo, fluoro, iodo, NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), or -N(acyl)2; each Y is independently selected from the group consisting of H, optionally substimted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH2, CH2NHCH3,
CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3jCF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH2, (CH2)mCONR2, and (CH2)mCONHR;
R is H, alkyl or acyl; Y1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR4, NH2, NHR4, NR4R5, SH or SR4; each Y2 is independently O, S, NH or NR4; each Y3 is independently H, F, Cl, Br or I; wherein for Base (B), W4 cannot be CH if W1, W2 and W3 are N; wherein for Base (E), (F), (K), (L), (W) and (X), W4 cannot be CH if W1 is N; each R6 is independently an optionally substimted alkyl, CH3, CH2CN, CH N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl,
CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, -CH2C(0)OH, -CH2C(0)OR4, -CH2C(0)0(lower alkyl), -CH2C(0)NH2, -CH2C(0)NHR4, -CH2C(0)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH )mC(0)0(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)raC(0)N(R4)2, -(CH2)mC(0)N(lower alkyl)2, -C(0)OH, -C(0)OR4, -C(0)0(lower alkyl), -C(0)NH2, -C(0)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2 or cyano; each R7 is independently OH, OR2, optionally substimted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2- Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, optionally substimted carbocycle, optionally substituted heterocycle, optionally substimted heteroaryl,
-CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(0)NH2, -CH2C(0)NHR4, -CH2C(0)NH(lower alkyl), -CH2C(0)N(R4)2, -CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(0)OR4 5 -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(O)S(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl),
-(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)SH, -C(0)SR4, -C(O)S(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, -O(acyl), -O(lower acyl), -O(R4), -O(alkyl), -O(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), NO2,
NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo; alternatively, R6 and R7 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle or optionally substimted heterocycle; each R8 and Rπ is independently hydrogen, an optionally substimted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substituted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, -CH2C(0)OH,
-CH2C(0)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(0)OR4, -C(0)0(lower alkyl), -C(0)NH2, -C(0)NHR4, -C(0)NH(lower alkyl),
-C(0)N(R4)2, -C(0)N(lower alkyl)2, cyano, NH-acyl or N(acyl)2; each R9 and R10 are independently hydrogen, OH, OR2, optionally substimted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3) C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substituted alkynyl, haloalkynyl, optionally substituted carbocycle, optionally substimted heterocycle, optionally substimted heteroaryl, -CH2C(0)OH, -CH2C(0)OR4, -CH2C(0)0(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(O)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)SH, -(CH2)mC(O)SR4, -(CH2)mC(0)S(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(0)N(R4)2, -(CH )mC(0)N(lower alkyl)2, -C(0)OH, -C(O)OR4, -C(O)O(lower alkyl), -C(O)SH, -C(O)SR4, -C(O)S(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, -O(acyl), -O(lower acyl), -0(R4), -O(alkyl), -0(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), N02, NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo; each m is independently 0, 1 or 2; and alternatively, R6 and R10, R7 and R9, Rs and R7 or R9 and R11 can come together to form a bridged compound selected from the group consisting of optionally substituted carbocycle or optionally substituted heterocycle or alternatively, R6 and R7 or R9 and R10 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle or optionally substituted heterocycle.
5. A compound of Formula (VIII), (IX) or (X):
Figure imgf000178_0001
(VIII) (IX) (X) or a pharmaceutically acceptable salt thereof, wherein: wherein R1, R2 and R3 are independently H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl; CO-aryl; CO-alkoxyalkyl; CO-aryloxyalkyl; CO-substituted aryl; sulfonate ester; benzyl, wherein the phenyl group is optionally substimted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; an amino acid residue; a carbohydrate; a peptide; cholesterol; or pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 and/or R3 is independently H or phosphate; wherein at least one of R2 and R3 is not hydrogen; X is O, S, S02, CH2, CH2OH, CHF, CF2, C(Y3)2, CHCN, C(CN)2, CHR4 or C(R4)2;
X* is CH, CF, CY3, or CR4; each Y3 is independently H, F, Cl, Br or I; each R4 and R5 is independently hydrogen, acyl, alkyl, lower alkyl, alkenyl or cycloalkyl; Base* is a purine or pyrimidine base; each R12 is independently a substituted alkyl, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, substituted alkenyl, haloalkenyl (but not Br-vinyl), substituted alkynyl, haloalkynyl, -CH2C(0)OH, -CH2C(0)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)2,
-CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(0)OR4, -(CH2)mC(0)0(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(0)N(R4)2, -(CH2)mC(0)N(lower alkyl)2, -C(0)OH, -C(0)OR4, -C(0)NH2, -C(0)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyfh; each R13 is independently substimted alkyl, CH2CN, CH2N3, CH2NH2, CH2NHCH3,
CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, substituted alkenyl, haloalkenyl (but not Br-vinyl), substituted alkynyl, haloalkynyl, optionally substimted carbocycle, optionally substituted heterocycle, optionally substimted heteroaryl, -CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(0)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(0)NH2, -CH2C(0)NHR4,
-CH2C(0)NH(lower alkyl), -CH2C(0)N(R4)2, -CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(0)OR4 5 -(CH2)mC(0)0(lower alkyl), -(CH2)mC(0)SH, -(CH2)mC(0)SR4, -(CH2)mC(0)S(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(0)N(R4)2, -(CH2)mC(0)N(lower alkyl)2, -C(0)OH, -C(0)OR4, -C(0)SH, -C(0)SR4, -C(0)S(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl),
-C(O)N(R4)2, -C(O)N(lower alkyl)2, -O(R4), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), - S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), -NHR4, -NR4R5, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), SCN, OCN, NCO or fluoro; alternatively, R12 and R13 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle or optionally substimted heterocycle; and each m is independently 0, 1 or 2.
6. A compound of Formula (XI) or (XII):
Figure imgf000180_0001
(XI) (XII) or a pharmaceutically acceptable salt thereof, wherein: R1 is H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl; CO- aryl; CO-alkoxyalkyl; CO-aryloxyalkyl; CO-substituted aryl; sulfonate ester; benzyl, wherein the phenyl group is optionally substituted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; an amino acid residue; a carbohydrate; a peptide; cholesterol; or pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R is H or phosphate;
Base is selected from the group consisting of
Figure imgf000180_0002
(A) (B)
Figure imgf000181_0001
(C) (D) (E) (F)
Figure imgf000181_0002
(G) (H)
Figure imgf000181_0003
(I) (J) (K) CL)
Figure imgf000181_0004
(M) (N)
Figure imgf000182_0001
(O) (P) (Q) (R)
Figure imgf000182_0002
(S) (T)
Figure imgf000182_0003
(U) (V) (W) (X)
Figure imgf000182_0004
00 (Z)
Figure imgf000183_0001
(AA) (AB) (AC) (AD)
Figure imgf000183_0002
(AE) (AF)
Figure imgf000183_0003
(AG) (AH) (Al) (AJ)
Figure imgf000183_0004
(BA) (BB)
Figure imgf000184_0001
(BC) (BD) (BE) (BF)
Figure imgf000184_0002
(BG) (BH)
Figure imgf000184_0003
(Bl) (BJ) (BK) (BL)
Figure imgf000184_0004
(BM) (BN)
Figure imgf000185_0001
(BO) (BP) (BQ) (BR)
Figure imgf000185_0002
(BY) (BZ)
Figure imgf000186_0001
(BAA) (BAB) (BAC) (BAD)
Figure imgf000186_0002
(BAE) (BAF)
each W1, W2, W3 and W4 is independently N, CH, CF, CI, CBr, CCl, CCN, CCH3, CCF3, CCH2CH3, CC(0)NH2, CC(0)NHR4, CC(0)N(R4)2, CC(0)OH, CC(0)OR4 or CX3; each W* is independently O, S, NH or NR4;
X is O, S, S02, CH2, CH2OH, CHF, CF2, C(Y3)2, CHCN, C(CN)2, CHR4 or C(R4)2;
X* is CH, CF, CY3 or CR4;
X2 is H, straight chained, branched or cyclic optionally substituted alkyl, CH3, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, CH2OH, optionally substimted alkenyl, optionally substimted alkynyl, COOH, COOR4, COO-alkyl, COO-aryl, CO-Oalkoxyalkyl, CONH2, CONHR4, CON(R4)2, chloro, bromo, fluoro, iodo, CN, N3, OH, OR4, NH2, NHR4, NR4R5, SH or SR5; each X3 is independently a straight chained, branched or cyclic optionally substimted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, N3, CN, -C(0)OH, -C(0)OR4, -C(0)O(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, OH, OR4, -O(acyl), -0(lower acyl), -O(alkyl), -O(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), - S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), chloro, bromo, fluoro, iodo, NH2, -NH(lower alkyl), -NHR4, -NR4R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), or -N(acyl)2; each Y is independently selected from the group consisting of H, optionally substituted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3,CF2CF3, CH2C02R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH2, (CH2)mCONR2, and (CH2)mCONHR; R is H, alkyl or acyl; Y1 is hydrogen, bromo, chloro, fluoro, iodo, CN, OH, OR4, NH2, NHR4, NR4R5, SH or SR4; each Y2 is independently O, S, NH or NR4; each Y3 is independently H, F, Cl, Br or I; wherein for Base (B), W4 cannot be CH if W1, W2 and W3 are N; wherein for Base (E), (F), (K), (L), (W) and (X), W4 cannot be CH if W1 is N; each R4 and R5 is independently hydrogen, acyl, alkyl, lower alkyl, alkenyl, alkynyl or cycloalkyl; each R12 is independently a substimted alkyl, CH CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, substimted alkenyl, haloalkenyl (but not Br-vinyl), substimted alkynyl, haloalkynyl, -CH2C(0)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(O)N(R4)2, -CH2C(O)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(0)OR4, -(CH2)mC(0)0(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(0)N(R4)2, -(CH2)mC(0)N(lower alkyl)2, -C(0)OH, -C(0)OR4, -C(0)NH2,
-C(0)NHR4, -C(0)NH(lower alkyl), or -C(0)N(R4)2, -C(0)N(lower alkyl)2; each R13 is independently substimted alkyl, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, substimted alkenyl, haloalkenyl (but not Br-vinyl), substituted alkynyl, haloalkynyl, optionally substituted carbocycle, optionally substimted heterocycle, optionally substituted heteroaryl, -CH2C(O)OH, -CH2C(0)OR4, -CH2C(0)0(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(0)S(lower alkyl), -CH2C(0)NH2, -CH2C(0)NHR4, -CH2C(0)NH(lower alkyl), -CH2C(0)N(R4)2, -CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(0)0R4, -(CH2)mC(0)0(lower alkyl), -(CH2)mC(0)SH, -(CH2)mC(0)SR4, -(CH2)mC(0)S(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(0)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(O)SH, -C(O)SR4, -C(O)S(lower alkyl), -C(O)NH2, -C(O)NHR4, -C(O)NH(lower alkyl), -C(O)N(R4)2, -C(O)N(lower alkyl)2, -O(R4), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), -S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), -NHR4, -NR4R5, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), SCN, OCN, NCO or fluoro; and alternatively, R12 and R13 can come together to form a spiro compound selected from the group consisting of optionally substituted carbocycle or optionally substimted heterocycle; each R8 and R11 is independently hydrogen, an optionally substimted alkyl (including lower alkyl), CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH N(CH3)2, CH2OH, halogenated alkyl (including halogenated lower alkyl), CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, -CH2C(0)OH, -CH2C(0)OR4,
-CH2C(0)0(lower alkyl), -CH2C(O)NH2, -CH2C(O)NHR4, -CH2C(O)NH(lower alkyl), -CH2C(0)N(R4)2, -CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(O)OR4, -(CH2)mC(O)O(lower alkyl), -(CH2)mC(O)NH2, -(CH2)mC(O)NHR4, -(CH2)mC(O)NH(lower alkyl), -(CH2)mC(O)N(R4)2, -(CH2)mC(O)N(lower alkyl)2, -C(O)OH, -C(O)OR4, -C(0)0(lower alkyl), -C(O)NH2, -C(0)NHR4, -C(0)NH(lower alkyl), -C(0)N(R4)2,
-C(0)N(lower alkyl)2, cyano, NH-acyl or N(acyl)2; each R9 and R10 are independently hydrogen, OH, OR2, optionally substituted alkyl, CH3, CH2CN, CH2N3, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2OH, halogenated alkyl, CF3, C(Y3)3, 2-Br-ethyl, CH2F, CH2C1, CH2CF3, CF2CF3, C(Y3)2C(Y3)3, optionally substimted alkenyl, haloalkenyl, Br-vinyl, optionally substimted alkynyl, haloalkynyl, optionally substimted carbocycle, optionally substituted heterocycle, optionally substituted heteroaryl, -CH2C(O)OH, -CH2C(O)OR4, -CH2C(O)O(lower alkyl), -CH2C(O)SH, -CH2C(O)SR4, -CH2C(O)S(lower alkyl), -CH2C(O)NH2, -CH2C(0)NHR4, -CH2C(0)NH(lower alkyl), -CH2C(0)N(R4)2, -CH2C(0)N(lower alkyl)2, -(CH2)mC(0)OH, -(CH2)mC(0)OR4, -(CH2)mC(0)0(lower alkyl), -(CH2)mC(0)SH, -(CH2)mC(0)SR4,
-(CH2)mC(0)S(lower alkyl), -(CH2)mC(0)NH2, -(CH2)mC(0)NHR4, -(CH2)mC(0)NH(lower alkyl), -(CH2)mC(0)N(R4)2, -(CH2)mC(0)N(lower alkyl)2, -C(0)OH, -C(0)OR4, -C(0)0(lower alkyl), -C(0)SH, -C(0)SR4, -C(0)S(lower alkyl), -C(0)NH2, -C(O)NHR4, -C(0)NH(lower alkyl), -C(0)N(R4)2, -C(O)N(lower alkyl)2, -O(acyl), -O(lower acyl), -0(R4), -O(alkyl), -0(lower alkyl), -O(alkenyl), -O(alkynyl), -O(aralkyl), -O(cycloalkyl), - S(acyl), -S(lower acyl), -S(R4), -S(lower alkyl), -S(alkenyl), -S(alkynyl), -S(aralkyl), -S(cycloalkyl), N02, NH2, -NH(lower alkyl), -NHR4, -NR R5, -NH(acyl), -N(lower alkyl)2, -NH(alkenyl), -NH(alkynyl), -NH(aralkyl), -NH(cycloalkyl), -N(acyl)2, azido, cyano, SCN, OCN, NCO or halo; each m is independently 0, 1 or 2; and . alternatively, R8 and R13, R9 and R13, R9 and Rπ or R10 and R12 can come together to form a bridged compound selected from the group consisting of optionally substimted carbocycle or optionally substimted heterocycle; or alternatively, R12 and R13 or R9 and R10 can come together to form a spiro compound selected from the group consisting of optionally substimted or optionally substituted heterocycle.
7. A compound of the Formula (XIII) or (XIV):
Figure imgf000189_0001
or a pharmaceutically acceptable salt thereof, wherein:
R3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; acyl; a sulfonate ester; optionally substimted alkyl sulfonyl; optionally substimted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R3 is independently H, or mono-, di- or triphosphate;
B indicates a spiro compound selected from the group consisting of optionally substituted carbocycle or optionally substimted heterocycle; Base is selected from the group consisting of:
Figure imgf000190_0001
(g) (h) (i)
and
Figure imgf000190_0002
wherein each R', R", R'" and R"" are independently selected from the group consisting of H, OH, substimted or unsubstimted alkyl, substimted or unsubstimted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH2, NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH2, CO2-alkyl, CONH-alkyl, CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mN02 and (CH2)mCONH2; m is 0 or 1;
W is C-R" orN;
T and V independently are CH or N; Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH2, or N;
Qi and Q2 independently are N or C-R;
R is H, alkyl, or acyl; and
Q3, Q4, Q5 and Q6 independently are N or CH.
8. A compound of Formula (XIX), (XX), (XXI) (XXfl) or (XXIII):
Figure imgf000191_0001
(XIX) (XX) (XXI) (XXII)
Figure imgf000191_0002
(XXIII) or a pharmaceutically acceptable salt thereof, wherein:
A is selected from the group consisting of optionally substimted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3,CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH2, (CH2)mCONR2, and (CH2)mCONHR;
Y is selected from the group consisting of H, optionally substimted lower alkyl, cycloalkyl, alkenyl, alkynyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3,CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCONH2, (CH2)mCONR2, and (CH2)mCONHR; R is H, alkyl or acyl;
X is selected from the group consisting of -OH, optionally substimted alkyl, cycloalkyl, alkenyl, alkynyl, -O-alkyl, -O-alkenyl, -O-alkynyl, -O-aryl, -O-aralkyl, -O- cycloalkyl-, O-acyl, F, Cl, Br, I, CN, NC, SCN, OCN, NCO, NO2, NH2, N3, NH-acyl, NH-alkyl, N-dialkyl, NH-alkenyl, NH-alkynyl, NH-aryl, NH-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-alkenyl, S-alkynyl, S-aryl, S-aralkyl, S-acyl, S-cycloalkyl, CO -alkyl,
CONH-alkyl, CON-dialkyl, CONH-alkenyl, CONH-alkynyl, CONH-aralkyl, CONH- cycloalkyl, CH2OH, CH2NH2, CH2NHCH3, CH2N(CH3)2, CH2F, CH2C1, CH2N3, CH2CN, CH2CF3, CF3,CF2CF3, CH2CO2R, (CH2)mCOOH, (CH2)mCOOR, (CH2)mCO- NH2, (CH2)mCONR2, (CH2)mCONHR, an optionally substituted 3-7 membered carbocyclic, and an optionally substimted 3-7 membered heterocyclic ring having O, S and/or N independently as a heteroatom taken alone or in combination; m is 0 or 1; R3 is selected from the group consisting of H; mono-, di-, and tri-phosphate or a stabilized phosphate prodrug; substituted or unsubstimted alkyl; acyl; a sulfonate ester; optionally substimted alkyl sulfonyl; optionally substituted arylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; and a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R3 is independently H, or mono-, di- or triphosphate; and
Base is a non-natural base selected from the group of:
Figure imgf000192_0001
wherein: each R', R", R'" and R"" is independently selected from the group consisting of
H, OH, substimted or unsubstituted alkyl, substituted or unsubstimted alkenyl, substimted or unsubstituted alkynyl, cycloalkyl, Br-vinyl, -O-alkyl, O-alkenyl, O- alkynyl, O-aryl, O-aralkyl, -O-acyl, O-cycloalkyl, NH2, NH-alkyl, N-dialkyl,
NH-acyl, N-aryl, N-aralkyl, NH-cycloalkyl, SH, S-alkyl, S-acyl, S-aryl, S- cycloalkyl, S-aralkyl, F, Cl, Br, I, CN, COOH, CONH2, CO2-alkyl, CONH-alkyl,
CON-dialkyl, OH, CF3, CH2OH, (CH2)mOH, (CH2)mNH2, (CH2)mCOOH,
(CH2)mCN, (CH2)mN02 and (CH2)mCONH2; m is 0 or 1;
W is C-R" orN;
T and V independently are CH or N;
Q is CH, -CCl, -CBr, -CF, -CI, -CCN, -C-COOH, -C-CONH2, orN;
Qi and Q2 independently are N or C-R""; and
Q3, Q4, Q5 and Q6 independently are N or CH; with the proviso that in bases (g) and (i), R', R"" are not H, OH, or NH2; and Q, T, V, Q2, Q5 and Q6 are not N.
9. A compound of Formula (LX):
Figure imgf000193_0001
(IX) or rtable salt thereof, wherein:
R1, R2 and R3 are independently H; phosphate; straight chained, branched or cyclic alkyl; acyl; CO-alkyl; CO-aryl; CO-alkoxyalkyl; CO-aryloxyalkyl; CO-substituted aryl; sulfonate ester; benzyl, wherein the phenyl group is optionally substimted with one or more substituents; alkylsulfonyl; arylsulfonyl; aralkylsulfonyl; a lipid; an amino acid; a carbohydrate; a peptide; cholesterol; or a pharmaceutically acceptable leaving group which when administered in vivo is capable of providing a compound wherein R1, R2 and/or R3 is independently H or phosphate; X is O, S, SO2 or CH2;
Base* is a purine or pyrimidine base;
R12 is C(Y3)3;
Y3 is independently H, F, Cl, Br or I; and
R13 is fluoro.
10. The compound of claim 9, wherein X is O, and Y3 is H.
11. The compound of claim 10, wherein R1, R2 and R3 are H.
12. A method for the treatment of a host infected with a Flaviviridae virus, comprising administering an effective treatment amount of a compound as claimed in any one of claims 1-11, or a pharmaceutically acceptable salt thereof.
13. The method of claim 12, wherein the virus is hepatitis C.
14. The method of claim 12, wherein the compound or pharmaceutically acceptable salt thereof is administered in combination or alternation with a second anti-viral agent.
15. The method of claim 14, wherein the second anti-viral agent is selected from the group consisting of an interferon, a ribavirin, an interleukin, aNS3 protease inhibitor, a cysteine protease inhibitor, a phenan-threnequinone, a thiazolidine derivative, a thiazolidine, a benzanilide, a phenan-threnequinone, a helicase inhibitor, a polymerase inhibitor, a nucleotide analogue, a gliotoxin, a cerulenin, an antisense phosphorothioate oligodeoxynucleotide, an inhibitor of IRES-dependent translation, and a ribozyme.
16. The method of claim 15, wherein the second anti-viral agent is an interferon.
17. The method of claim 16, wherein the second anti-viral agent is selected from the group consisting of pegylated interferon alpha 2a, interferon alphacon-1, natural interferon, albuferon, interferon beta- la, omega interferon, interferon alpha, interferon gamma, interferon tau, interferon delta and interferon gamma- lb.
18. The method of claim 12, wherein the compound or pharmaceutically acceptable salt thereof is in the form of a dosage unit.
19. The method of claim 18 wherein the dosage unit contains 50 to 1000 mg or 0.1 to 50 mg of the compound.
20. The method of claim 18 wherein the dosage unit is a tablet or capsule.
21. The method of claim 12, wherein the host is a human.
22. The method of claim 12, wherein the compound or pharmaceutically acceptable salt thereof is in substantially pure form.
23. The method of claim 12, wherein the compound or pharmaceutically acceptable salt thereof is at least 90% by weight of the β-D-isomer.
24. The method of claim 12, wherein the compound or pharmaceutically acceptable salt thereof is at least 95% by weight of the β-D-isomer.
25. The method of claim 12, wherein the compound is in the form of a pharmaceutically acceptable salt selected from the group consisting of a tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorate, α- ketoglutarate, α-glycerophosphate, formate, fumarate, propionate, glycolate, lactate, pyruvate, oxalate, maleate, salicylate, sulfate, nitrate, bicarbonate, carbonate salts, hydrobromate, hydrochloride, di-hydrochloride, and phosphoric acid salt.
26. The method of claim 25, wherein the pharmaceutically acceptable salt is a hydrochloride salt.
27. A pharmaceutical composition comprising an effective amount to treat a Flaviviridae infection of a compound, or a pharmaceutically acceptable salt thereof, of any of claims 1 to 11 in a pharmaceutically acceptable carrier .
28. The pharmaceutical composition of claim 27, wherein the canier is suitable for oral delivery.
29. The pharmaceutical composition of claim 27 comprising an effective amount of the compound or pharmaceutically acceptable salt thereof to treat a host infected with West Nile Virus, Yellow fever, Denge Virus or BVDV.
30. The composition of claim 27, wherein the Flaviviridae virus is hepatitis C.
31. The pharmaceutical composition of claim 29, wherein the compound or pharmaceutically acceptable salt thereof, is in the form of a dosage unit.
32. The composition of claim 31, wherein the dosage unit contains 0.1 to 50 mg or 50 to 1000 mg of the compound or pharmaceutically acceptable salt thereof.
33. The composition of claim 31 , wherein said dosage unit is a tablet or capsule.
34. The pharmaceutical composition of claim 27, further comprising a second anti-viral agent.
35. The pharmaceutical composition of claim 34, wherein the second anti-viral agent is selected from the group consisting of an interferon, a ribavirin, an interleukin, a
NS3 protease inhibitor, a cysteine protease inhibitor, a phenan-threnequinone, a thiazolidine derivative, a thiazolidine, a benzanilide, a phenan-threnequinone, a helicase inhibitor, a polymerase inhibitor, a nucleotide analogue, a gliotoxin, a cerulenin, an antisense phosphorothioate oligodeoxynucleotide, an inhibitor of IRES-dependent translation, and a ribozyme.
36. The pharmaceutical composition of claim 35, wherein the second anti-viral agent is an interferon.
37. The pharmaceutical composition of claim 36, wherein the second anti-viral agent is selected from the group consisting of pegylated interferon alpha 2a, interferon alphacon-1, natural interferon, albuferon, interferon beta- la, omega interferon, interferon alpha, interferon gamma, interferon tau, interferon delta and interferon gamma- lb.
38. The pharmaceutical composition of claim 27, wherein the compound or pharmaceutically acceptable salt thereof, is in substantially pure form.
39. The pharmaceutical composition of claim 27, wherein the compound or pharmaceutically acceptable salt thereof, is at least 90% by weight of the β-D-isomer.
40. The pharmaceutical composition of claim 27, wherein the compound or pharmaceutically acceptable salt thereof, is at least 95% by weight of the β-D-isomer.
41. The pharmaceutical composition of claim 27 further comprising a pharmaceutically acceptable canier suitable for oral, parenteral, inhalant or intravenous delivery.
42. The pharmaceutical composition of claim 27, wherein the pharmaceutically acceptable salt is selected from the group consisting of atosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorate, α-ketoglutarate, α- glycerophosphate, formate, fumarate, propionate, glycolate, lactate, pyruvate, oxalate, maleate, sahcylate, sulfate, nitrate, bicarbonate, carbonate salts, hydrobromate, hydrochloride, di-hydrochloride, and phosphoric acid salt.
43. The pharmaceutical composition of claim 42, wherein the pharmaceutically acceptable salt is a hydrochloride salt.
44. A compound or a pharmaceutically acceptable salt thereof, of any of claims 1 to 11 for the treatment of a host infected with a Flaviviridae virus.
45. The compound of claim 44, wherein the virus is hepatitis C.
46. The compound of claim 44, wherein the host is a human.
47. The use of a compound or a pharmaceutically acceptable salt thereof, of any of claims 1 to 11 in the manufacture of a medicament for the treatment of a host infected with a Flaviviridae virus.
48. The use of claim 44, wherein the virus is hepatitis C.
49. The use of claim 44, wherein the host is a human.
PCT/IB2003/003246 2002-06-28 2003-06-27 Modified 2' and 3' -nucleoside produgs for treating flaviridae infections WO2004002999A2 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
SI200332358T SI1523489T1 (en) 2002-06-28 2003-06-27 Modified 2' and 3' -nucleoside produgs for treating flaviridae infections
BRPI0312286-7A BR0312286A (en) 2002-06-28 2003-06-27 2 'and 3' prodrugs - modified nucleoside for treatment of flaviviridae infections
CA2490191A CA2490191C (en) 2002-06-28 2003-06-27 Modified 2' and 3' -nucleoside prodrugs for treating flaviviridae infections
ES03761744.6T ES2469569T3 (en) 2002-06-28 2003-06-27 2 'and 3' modified nucleoside prodrugs for the treatment of Flaviviridae infections
AP2005003212A AP2005003212A0 (en) 2002-06-28 2003-06-27 Modified 2' and 3' nucleoside prodrugs for treating flaviviridae infections.
AU2003247084A AU2003247084B9 (en) 2002-06-28 2003-06-27 Modified 2' and 3'-nucleoside prodrugs for treating flaviridae infections
YUP-1140/04A RS114004A (en) 2002-06-28 2003-06-27 Modified 2' and 3'-nucleoside produgs for treating flaviridae infections
DK03761744.6T DK1523489T3 (en) 2002-06-28 2003-06-27 MODIFIED 2 'AND 3' NUCLEOSIDE PRODUCTS TO TREAT FLAVIRIDA INFECTIONS
EP03761744.6A EP1523489B1 (en) 2002-06-28 2003-06-27 Modified 2' and 3' -nucleoside produgs for treating flaviridae infections
JP2004517158A JP2005533817A (en) 2002-06-28 2003-06-27 Modified 2 'and 3'-nucleoside prodrugs for the treatment of Flaviviridae virus infection
MXPA04012709A MXPA04012709A (en) 2002-06-28 2003-06-27 Modified 2' and 3' -nucleoside produgs for treating flaviridae infections.
IL16589304A IL165893A0 (en) 2002-06-28 2004-12-21 Modified 2' and 3'-nucleoside prodrugs for treating flaviridae infections
NO20050465A NO330755B1 (en) 2002-06-28 2005-01-27 Compound, pharmaceutical composition and uses thereof
NO20110343A NO20110343A1 (en) 2002-06-28 2011-03-04 Modified 2 'and 3' nucleoside prodrugs for the treatment of flaviviridae infections
NO2014019C NO2014019I1 (en) 2002-06-28 2014-07-30 Sofosbuvir
CY2014031C CY2014031I2 (en) 2002-06-28 2014-08-12 MODIFIED 2' AND 3'-NUCLEOSITE PRODRUGS FOR THE TREATMENT OF FLAVIVIRIDAE INFECTIONS

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US39235002P 2002-06-28 2002-06-28
US39235102P 2002-06-28 2002-06-28
US60/392,351 2002-06-28
US60/392,350 2002-06-28
US46619403P 2003-04-28 2003-04-28
US60/466,194 2003-04-28
US47094903P 2003-05-14 2003-05-14
US60/470,949 2003-05-14

Publications (3)

Publication Number Publication Date
WO2004002999A2 true WO2004002999A2 (en) 2004-01-08
WO2004002999A3 WO2004002999A3 (en) 2004-08-12
WO2004002999A8 WO2004002999A8 (en) 2005-02-17

Family

ID=30003988

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2003/003246 WO2004002999A2 (en) 2002-06-28 2003-06-27 Modified 2' and 3' -nucleoside produgs for treating flaviridae infections

Country Status (27)

Country Link
EP (3) EP2332952B1 (en)
JP (7) JP2005533817A (en)
KR (1) KR20050048544A (en)
CN (6) CN104193791A (en)
AP (1) AP2005003212A0 (en)
AR (2) AR043101A1 (en)
AU (1) AU2003247084B9 (en)
BR (2) BR0312271A (en)
CA (1) CA2490191C (en)
CY (2) CY1115417T1 (en)
DE (1) DE14169110T1 (en)
DK (1) DK1523489T3 (en)
ES (1) ES2469569T3 (en)
HK (3) HK1204624A1 (en)
IL (1) IL165893A0 (en)
MA (1) MA27375A1 (en)
MX (1) MXPA04012709A (en)
MY (1) MY140819A (en)
NO (3) NO330755B1 (en)
PE (3) PE20040636A1 (en)
PL (1) PL374831A1 (en)
PT (1) PT1523489E (en)
RS (1) RS114004A (en)
RU (1) RU2483075C2 (en)
TW (3) TW200500375A (en)
UY (3) UY27874A1 (en)
WO (1) WO2004002999A2 (en)

Cited By (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005003147A2 (en) 2003-05-30 2005-01-13 Pharmasset, Inc. Modified fluorinated nucleoside analogues
WO2005080388A1 (en) 2004-02-20 2005-09-01 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
WO2005123087A2 (en) 2004-06-15 2005-12-29 Merck & Co., Inc. C-purine nucleoside analogs as inhibitors of rna-dependent rna viral polymerase
US7148349B2 (en) 2002-10-31 2006-12-12 Metabasis Therapeutics, Inc. Cyclic phosphate diesters of 1,3-propane-1-aryl diols and their use in preparing prodrugs
US7157441B2 (en) 2000-05-23 2007-01-02 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus
WO2007113159A1 (en) 2006-04-04 2007-10-11 F. Hoffmann-La Roche Ag 3',5'-di-o-acylated nucleosides for hcv treatment
US7323449B2 (en) 2002-07-24 2008-01-29 Merck & Co., Inc. Thionucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
JP2008514639A (en) * 2004-09-24 2008-05-08 イデニクス(ケイマン)リミテツド Methods and compositions for treating flavivirus, pestivirus and hepacivirus infections
WO2008085508A2 (en) 2007-01-05 2008-07-17 Merck & Co., Inc. Nucleoside aryl phosphoramidates for the treatment of rna-dependent rna viral infection
US7585851B2 (en) 2000-06-15 2009-09-08 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
WO2009129120A2 (en) 2008-04-15 2009-10-22 Rfs Pharma, Llc Nucleoside derivatives for treatment of caliciviridae infections, including norovirus infections
EP2141172A1 (en) 2008-07-01 2010-01-06 Ortho Biotech Products L.P. Cyclopropyl polymerase inhibitors
US7666855B2 (en) 2004-02-13 2010-02-23 Metabasis Therapeutics, Inc. 2′-C-methyl nucleoside derivatives
US7666856B2 (en) 2006-10-10 2010-02-23 Medivir Ab Antiviral nucleosides
WO2010066699A1 (en) * 2008-12-08 2010-06-17 Centocor Ortho Biotech Products L.P. Uracyl cyclopropyl nucleotides
US7754699B2 (en) 2005-12-09 2010-07-13 Roche Palo Alto Llc Antiviral nucleosides
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
WO2010084115A2 (en) 2009-01-20 2010-07-29 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Antiviral agents
US7767660B2 (en) 2006-12-20 2010-08-03 Istituto Di Richerche Di Biologia Molecolare P. Angeletti Spa Antiviral indoles
US7781422B2 (en) 2006-12-20 2010-08-24 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Antiviral indoles
US7786110B2 (en) 2006-11-09 2010-08-31 Roche Palo Alto Llc Thiazole and oxazole-substituted arylamides as P2X3 and P2X2/3 antagonists
WO2010101967A2 (en) 2009-03-04 2010-09-10 Idenix Pharmaceuticals, Inc. Phosphothiophene and phosphothiazole hcv polymerase inhibitors
US7824851B2 (en) 2002-11-15 2010-11-02 Idenix Pharmaceuticals, Inc. 2′-branched nucleosides and Flaviviridae mutation
US7858654B2 (en) 2007-12-17 2010-12-28 Roche Palo Alto Llc Imidazole-substituted arylamides as P2X3 and P2X2/3 antagonists
US7879797B2 (en) 2005-05-02 2011-02-01 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
WO2011014487A1 (en) 2009-07-30 2011-02-03 Merck Sharp & Dohme Corp. Hepatitis c virus ns3 protease inhibitors
WO2011017389A1 (en) 2009-08-05 2011-02-10 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors useful against viral infections, particularly hcv
US7902202B2 (en) 2006-12-28 2011-03-08 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
WO2011058084A1 (en) 2009-11-14 2011-05-19 F. Hoffmann-La Roche Ag Biomarkers for predicting rapid response to hcv treatment
WO2011063076A1 (en) 2009-11-19 2011-05-26 Itherx Pharmaceuticals, Inc. Methods of treating hepatitis c virus with oxoacetamide compounds
WO2011067195A1 (en) 2009-12-02 2011-06-09 F. Hoffmann-La Roche Ag Biomarkers for predicting sustained response to hcv treatment
WO2011075615A1 (en) 2009-12-18 2011-06-23 Idenix Pharmaceuticals, Inc. 5,5-fused arylene or heteroarylene hepatitis c virus inhibitors
US7973040B2 (en) 2008-07-22 2011-07-05 Merck Sharp & Dohme Corp. Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
US7989637B2 (en) 2007-12-17 2011-08-02 Roche Palo Alto Llc Triazole-substituted arylamides as P2X3 and P2X2/3 antagonists
US7989438B2 (en) 2007-07-17 2011-08-02 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Therapeutic compounds
WO2011123586A1 (en) 2010-04-01 2011-10-06 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US8048905B2 (en) 2007-12-17 2011-11-01 Roche Palo Alto Llc Tetrazole-substituted arylamides as P2X3 and P2X2/3 antagonists
US8058260B2 (en) 2006-05-22 2011-11-15 Xenoport, Inc. 2′-C-methyl-ribofuranosyl cytidine prodrugs, pharmaceutical compositions and uses thereof
US8093275B2 (en) 2009-06-22 2012-01-10 Roche Palo Alto Llc Oxazolone and pyrrolidinone-substituted pryidine amides as P2X3 and P2X2/3 antagonists
US8101595B2 (en) 2006-12-20 2012-01-24 Istituto di Ricerche di Biologia Molecolare P. Angletti SpA Antiviral indoles
US8138164B2 (en) 2006-10-24 2012-03-20 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US8178520B2 (en) 2006-05-15 2012-05-15 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Macrocyclic compounds as antiviral agents
WO2012080050A1 (en) 2010-12-14 2012-06-21 F. Hoffmann-La Roche Ag Solid forms of a phenoxybenzenesulfonyl compound
US8216999B2 (en) 2005-07-20 2012-07-10 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
EP2476690A1 (en) 2008-07-02 2012-07-18 IDENIX Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US8227431B2 (en) 2008-03-17 2012-07-24 Hetero Drugs Limited Nucleoside derivatives
WO2012109398A1 (en) 2011-02-10 2012-08-16 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating hcv infections
US8278322B2 (en) 2005-08-01 2012-10-02 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
WO2012135581A1 (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
US8283383B2 (en) 2009-06-22 2012-10-09 Roche Palo Alto Llc Biphenyl amides as P2X3 and P2X2/3 antagonists
WO2012142523A2 (en) 2011-04-13 2012-10-18 Gilead Sciences, Inc. 1'-substituted pyrimidine n-nucleoside analogs for antiviral treatment
US8309540B2 (en) 2006-10-24 2012-11-13 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
WO2012154321A1 (en) 2011-03-31 2012-11-15 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US8314062B2 (en) 2006-06-23 2012-11-20 Instituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Macrocyclic compounds as antiviral agents
US8329664B2 (en) 2006-05-09 2012-12-11 University College Cardiff Consultants Limited Anti-viral pyrimidine nucleoside derivatives
US8377874B2 (en) 2006-10-27 2013-02-19 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US8377873B2 (en) 2006-10-24 2013-02-19 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
WO2013039855A1 (en) 2011-09-12 2013-03-21 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
WO2013039920A1 (en) 2011-09-12 2013-03-21 Idenix Pharmaceuticals, Inc. Substituted carbonyloxymethylphosphoramidate compounds and pharmaceutical compositions for the treatment of viral infections
WO2013056046A1 (en) 2011-10-14 2013-04-18 Idenix Pharmaceuticals, Inc. Substituted 3',5'-cyclic phosphates of purine nucleotide compounds and pharmaceutical compositions for the treatment of viral infections
US8440673B2 (en) 2007-12-17 2013-05-14 Roche Palo Alto Llc Pyrazole-substituted arylamides as P2X3 and P2X2/3 antagonists
WO2013074386A2 (en) 2011-11-15 2013-05-23 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors
US8461107B2 (en) 2008-04-28 2013-06-11 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US8470870B2 (en) 2008-03-27 2013-06-25 Idenix Pharmaceuticals, Inc. Solid forms of an anti-HIV phosphoindole compound
US8476457B2 (en) 2009-06-22 2013-07-02 Roche Palo Alto Llc Indole, indazole and benzimidazole arylamides as P2X3 and P2X2/3 antagonists
US8492539B2 (en) 2004-09-14 2013-07-23 Gilead Pharmasset Llc Preparation of 2′-fluoro-2′-alkyl-substituted or other optionally substituted ribofuranosyl pyrimidines and purines and their derivatives
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]
US8551973B2 (en) 2008-12-23 2013-10-08 Gilead Pharmasset Llc Nucleoside analogs
US8580765B2 (en) 2007-03-30 2013-11-12 Gilead Pharmasset Llc Nucleoside phosphoramidate prodrugs
WO2013177188A1 (en) 2012-05-22 2013-11-28 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
WO2013177195A1 (en) 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. 3',5'-cyclic phosphate prodrugs for hcv infection
US8618076B2 (en) 2009-05-20 2013-12-31 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8623901B2 (en) 2009-03-31 2014-01-07 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
US8623879B2 (en) 2008-04-02 2014-01-07 Boehringer Ingelheim International Gmbh 1-heterocyclyl-1,5-dihydro-pyrazolo[3,4-D] pyrimidin-4-one derivates and their use as PDE9A modulators
US8629263B2 (en) 2009-05-20 2014-01-14 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8629275B2 (en) 2008-09-08 2014-01-14 Merck Sharp & Dohme Corp. AHCY hydrolase inhibitors for treatment of hyper homocysteinemia
US8648085B2 (en) 2007-11-30 2014-02-11 Boehringer Ingelheim International Gmbh 1, 5-dihydro-pyrazolo (3, 4-D) pyrimidin-4-one derivatives and their use as PDE9A mudulators for the treatment of CNS disorders
WO2014058801A1 (en) 2012-10-08 2014-04-17 Idenix Pharmaceuticals, Inc. 2'-chloro nucleoside analogs for hcv infection
WO2014063019A1 (en) 2012-10-19 2014-04-24 Idenix Pharmaceuticals, Inc. Dinucleotide compounds for hcv infection
WO2014066239A1 (en) 2012-10-22 2014-05-01 Idenix Pharmaceuticals, Inc. 2',4'-bridged nucleosides for hcv infection
US8716262B2 (en) 2008-12-23 2014-05-06 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8716263B2 (en) 2008-12-23 2014-05-06 Gilead Pharmasset Llc Synthesis of purine nucleosides
WO2014078436A1 (en) 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. D-alanine ester of sp-nucleoside analog
WO2014078427A1 (en) 2012-11-14 2014-05-22 Idenix Pharmaceuticals, Inc. D-alanine ester of rp-nucleoside analog
WO2014099941A1 (en) 2012-12-19 2014-06-26 Idenix Pharmaceuticals, Inc. 4'-fluoro nucleosides for the treatment of hcv
WO2014123795A2 (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
WO2014123794A1 (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
US8809345B2 (en) 2011-02-15 2014-08-19 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
CN103992360A (en) * 2014-04-21 2014-08-20 中美华世通生物医药科技(武汉)有限公司 Reduction method suitable for industrialized production
WO2014137926A1 (en) 2013-03-04 2014-09-12 Idenix Pharmaceuticals, Inc. 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
WO2014148949A1 (en) 2013-03-22 2014-09-25 Асави, Ллс Alkyl 2-{[(2r,3s,5r)-5-(4-amino-2-oxo-2н-pyrimidin-1-yl)-3-hydroxy- tetrahydro-furan-2-yl-methoxy]-phenoxy-phosphoryl-amino}-propionates, nucleoside inhibitors of hcv ns5b rna-polymerase, and methods for producing and use thereof
WO2014165542A1 (en) 2013-04-01 2014-10-09 Idenix Pharmaceuticals, Inc. 2',4'-fluoro nucleosides for the treatment of hcv
US8859756B2 (en) 2010-03-31 2014-10-14 Gilead Pharmasset Llc Stereoselective synthesis of phosphorus containing actives
US8877731B2 (en) 2010-09-22 2014-11-04 Alios Biopharma, Inc. Azido nucleosides and nucleotide analogs
US8889159B2 (en) 2011-11-29 2014-11-18 Gilead Pharmasset Llc Compositions and methods for treating hepatitis C virus
US8895531B2 (en) 2006-03-23 2014-11-25 Rfs Pharma Llc 2′-fluoronucleoside phosphonates as antiviral agents
WO2014197578A1 (en) 2013-06-05 2014-12-11 Idenix Pharmaceuticals, Inc. 1',4'-thio nucleosides for the treatment of hcv
US8912201B2 (en) 2010-08-12 2014-12-16 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
US8912321B2 (en) 2006-10-10 2014-12-16 Gilead Pharmasset Llc Preparation of nucleosides ribofuranosyl pyrimidines
US8927569B2 (en) 2007-07-19 2015-01-06 Merck Sharp & Dohme Corp. Macrocyclic compounds as antiviral agents
WO2015017713A1 (en) 2013-08-01 2015-02-05 Idenix Pharmaceuticals, Inc. D-amino acid phosphoramidate pronucleotides of halogeno pyrimidine compounds for liver disease
WO2015042375A1 (en) 2013-09-20 2015-03-26 Idenix Pharmaceuticals, Inc. Hepatitis c virus inhibitors
WO2015054465A1 (en) * 2013-10-11 2015-04-16 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
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
WO2015066370A1 (en) 2013-11-01 2015-05-07 Idenix Pharmaceuticals, Inc. D-alanine phosphoramidate pronucleotides of 2'-methyl 2'-fluoro guanosine nucleoside compounds for the treatment of hcv
WO2015081297A1 (en) 2013-11-27 2015-06-04 Idenix Pharmaceuticals, Inc. 2'-dichloro and 2'-fluoro-2'-chloro nucleoside analogues for hcv infection
WO2015095419A1 (en) 2013-12-18 2015-06-25 Idenix Pharmaceuticals, Inc. 4'-or nucleosides for the treatment of hcv
US9067945B2 (en) 2002-08-23 2015-06-30 Boehringer Ingehleim International GmbH Selective phosphodiesterase 9A inhibitors as medicaments for improving cognitive processes
US9073960B2 (en) 2011-12-22 2015-07-07 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US9079905B2 (en) 2008-09-08 2015-07-14 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
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
WO2015134780A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Solid prodrug forms of 2'-chloro-2'-methyl uridine for hcv
WO2015134560A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Solid forms of a flaviviridae virus inhibitor compound and salts thereof
WO2015161137A1 (en) 2014-04-16 2015-10-22 Idenix Pharmaceuticals, Inc. 3'-substituted methyl or alkynyl nucleosides for the treatment of hcv
US9243022B2 (en) 2012-12-21 2016-01-26 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US9447132B2 (en) 2013-04-12 2016-09-20 Achillion Pharmaceuticals, Inc. Highly active nucleoside derivative for the treatment of HCV
US9738661B2 (en) 2006-10-27 2017-08-22 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US9828410B2 (en) 2015-03-06 2017-11-28 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US9845336B2 (en) 2012-05-25 2017-12-19 Janssen Sciences Ireland Uc Uracyl spirooxetane nucleosides
US9968628B2 (en) 2000-05-26 2018-05-15 Idenix Pharmaceuticals Llc Methods and compositions for treating flaviviruses and pestiviruses
US9994600B2 (en) 2014-07-02 2018-06-12 Ligand Pharmaceuticals, Inc. Prodrug compounds and uses therof
US10039779B2 (en) 2013-01-31 2018-08-07 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
WO2018217884A1 (en) * 2017-05-23 2018-11-29 Regents Of The University Of Minnesota Antibacterial agents including histidine kinase inhibitors
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
US10449210B2 (en) 2014-02-13 2019-10-22 Ligand Pharmaceuticals Inc. Prodrug compounds and their uses
US10456414B2 (en) 2011-09-16 2019-10-29 Gilead Pharmasset Llc Methods for treating HCV
US10485815B2 (en) 2012-03-21 2019-11-26 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
USRE48171E1 (en) 2012-03-21 2020-08-25 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
EP3750544A2 (en) 2011-11-30 2020-12-16 Emory University Jak inhibitors for use in the prevention or treatment of viral infection
US10874687B1 (en) 2020-02-27 2020-12-29 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US10946033B2 (en) 2016-09-07 2021-03-16 Atea Pharmaceuticals, Inc. 2′-substituted-N6-substituted purine nucleotides for RNA virus treatment
US11116783B2 (en) 2013-08-27 2021-09-14 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
WO2021221983A1 (en) * 2020-04-28 2021-11-04 Southlake Pharmaceuticals, Inc. Interferon tau as antiviral therapy
US11166973B2 (en) 2013-09-11 2021-11-09 Emory University Substituted nucleotides and nucleosides for treating viral infections
US11690860B2 (en) 2018-04-10 2023-07-04 Atea Pharmaceuticals, Inc. Treatment of HCV infected patients with cirrhosis
US11697666B2 (en) 2021-04-16 2023-07-11 Gilead Sciences, Inc. Methods of preparing carbanucleosides using amides
US11767337B2 (en) 2020-02-18 2023-09-26 Gilead Sciences, Inc. Antiviral compounds

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104193791A (en) 2002-06-28 2014-12-10 埃迪尼克斯医药公司 Modified 2' and 3'-nucleoside produgs for treating flaviridae infections
GB0317009D0 (en) 2003-07-21 2003-08-27 Univ Cardiff Chemical compounds
ATE478886T1 (en) 2003-07-25 2010-09-15 Idenix Pharmaceuticals Inc PURINE NUCLEOSIDES FOR THE TREATMENT OF DISEASES CAUSED BY FLAVIVIDRAE, INCLUDING HEPATITIS C
JP6069215B2 (en) 2010-11-30 2017-02-01 ギリアド ファーマセット エルエルシー Compound
CA2891125A1 (en) * 2012-11-19 2014-05-22 Merck Sharp & Dohme Corp. 2 -alkynyl substituted nucleoside derivatives for treating viral diseases
JP6728075B2 (en) * 2014-06-24 2020-07-22 ヤンセン バイオファーマ インク. Substituted nucleosides, nucleotides and analogs thereof
CN109748943A (en) * 2017-11-03 2019-05-14 中国科学院上海药物研究所 2 '-C- methyl substituted nucleosides class compounds and its preparation and purposes
CN111995649A (en) * 2020-04-09 2020-11-27 瀚海新拓(杭州)生物医药有限公司 Pteridinone nucleotide analogue and pharmaceutical composition, preparation method and medical application thereof
WO2022008025A1 (en) * 2020-07-05 2022-01-13 Since & Technology Development Fund Authority 2-hydroxyiminopyrimidine nucleosides and derivitives and antiviral uses thereto
CN112979733B (en) * 2021-04-25 2021-09-10 南京颐媛生物医学研究院有限公司 Anti-hepatitis B virus compound and preparation method and application thereof

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989002733A1 (en) 1987-09-22 1989-04-06 The Regents Of The University Of California Liposomal nucleoside analogues for treating aids
EP0350287A2 (en) 1988-07-07 1990-01-10 NeXstar Pharmaceuticals, Inc. Lipid derivatives of antiviral nucleosides, liposomal incorporation and method of use
WO1991016920A1 (en) 1990-05-07 1991-11-14 Vical, Inc. Lipid prodrugs of salicylate and nonsteroidal anti-inflammatory drugs
WO1991018914A1 (en) 1990-05-29 1991-12-12 Vical, Inc. Synthesis of glycerol di- and triphosphate derivatives
WO1991019721A1 (en) 1990-06-13 1991-12-26 Arnold Glazier Phosphorous produgs
US5149794A (en) 1990-11-01 1992-09-22 State Of Oregon Covalent lipid-drug conjugates for drug targeting
WO1993000910A1 (en) 1991-07-12 1993-01-21 Vical, Inc. Antiviral liponucleosides: treatment of hepatitis b
US5194654A (en) 1989-11-22 1993-03-16 Vical, Inc. Lipid derivatives of phosphonoacids for liposomal incorporation and method of use
US5256641A (en) 1990-11-01 1993-10-26 State Of Oregon Covalent polar lipid-peptide conjugates for immunological targeting
WO1994026273A1 (en) 1993-05-12 1994-11-24 Hostetler Karl Y Acyclovir derivatives for topical use
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
US5463092A (en) 1989-11-22 1995-10-31 Vestar, Inc. Lipid derivatives of phosphonacids for liposomal incorporation and method of use
WO1996015132A1 (en) 1994-11-15 1996-05-23 The Regents Of The University Of California Improved antiviral prodrugs
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
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
US5554728A (en) 1991-07-23 1996-09-10 Nexstar Pharmaceuticals, Inc. Lipid conjugates of therapeutic peptides and protease inhibitors
WO1999045016A2 (en) 1998-03-06 1999-09-10 Metabasis Therapeutics, Inc. Novel prodrugs for phosphorus-containing compounds
WO2000052015A2 (en) 1999-03-05 2000-09-08 Metabasis Therapeutics, Inc. Novel phosphorus-containing prodrugs
WO2001018013A1 (en) 1999-09-08 2001-03-15 Metabasis Therapeutics, Inc. Prodrugs for liver specific drug delivery
WO2001047935A2 (en) 1999-12-22 2001-07-05 Metabasis Therapeutics, Inc. Novel bisamidate phosphonate prodrugs
US6312662B1 (en) 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds

Family Cites Families (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
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.
JP2706666B2 (en) * 1987-07-31 1998-01-28 大鵬薬品工業株式会社 2'-deoxy-5-fluorouridine derivatives and salts thereof
GB8719367D0 (en) 1987-08-15 1987-09-23 Wellcome Found Therapeutic compounds
EP0346108A3 (en) * 1988-06-09 1991-04-24 The Wellcome Foundation Limited Anti-infective nucleosides
US5705363A (en) 1989-03-02 1998-01-06 The Women's Research Institute Recombinant production of human interferon τ polypeptides and nucleic acids
JPH0368564A (en) 1989-04-27 1991-03-25 Tsumura & Co Nucleic acid derivative
JPH032193A (en) * 1989-05-30 1991-01-08 Ube Ind Ltd 5-fluorouridine and 2'-deoxy-5-fluorouridine derivative
JPH0383994A (en) * 1989-08-28 1991-04-09 Kuraray Co Ltd 2'-deoxy-5-fluorouridine derivative, salt thereof, production thereof and antitumor agent comprising the same derivative as active ingredient
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
DE69132332T2 (en) 1990-04-06 2000-11-30 Genelabs Tech Inc HEPATITIS C-VIRUS-EPITOPE
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
US5595732A (en) 1991-03-25 1997-01-21 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5157027A (en) 1991-05-13 1992-10-20 E. R. Squibb & Sons, Inc. Bisphosphonate squalene synthetase inhibitors and method
TW224053B (en) 1991-09-13 1994-05-21 Paul B Chretien
JPH0597887A (en) * 1991-10-02 1993-04-20 Tanabe Seiyaku Co Ltd New 2'-deoxy-5-fluorouridine derivative
US5676942A (en) 1992-02-10 1997-10-14 Interferon Sciences, Inc. Composition containing human alpha interferon species proteins and method for use thereof
US5610054A (en) 1992-05-14 1997-03-11 Ribozyme Pharmaceuticals, Inc. Enzymatic RNA molecule targeted against Hepatitis C virus
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
US5496546A (en) 1993-02-24 1996-03-05 Jui H. Wang Compositions and methods of application of reactive antiviral polyadenylic acid derivatives
EP0773029A4 (en) 1993-07-19 1997-09-03 Tokyo Tanabe Co Hepatitis c virus proliferation inhibitor
NZ276943A (en) 1993-11-10 1998-02-26 Schering Corp Substituted For Alpha-interferon conjugated to a non-antigenic polymer (preferably a polyalkylene oxide) and its preparation
US5951974A (en) 1993-11-10 1999-09-14 Enzon, Inc. Interferon polymer conjugates
DE4447588C2 (en) 1994-05-03 1997-11-20 Omer Osama Dr Dr Med Treatment of herpes and hepatitis virus infections and bronchitis
DE4432623A1 (en) 1994-09-14 1996-03-21 Huels Chemische Werke Ag Process for bleaching aqueous surfactant solutions
US5738846A (en) 1994-11-10 1998-04-14 Enzon, Inc. Interferon polymer conjugates and process for preparing the same
JP3786447B2 (en) 1995-03-31 2006-06-14 エーザイ株式会社 Preventive and therapeutic agent for hepatitis C
GB9606264D0 (en) * 1995-07-13 1996-05-29 Hoffmann La Roche Pyrimidine nucleoside derivatives
DE69635864T2 (en) 1995-09-27 2006-10-26 Emory University 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
JP2000506010A (en) 1996-02-29 2000-05-23 イミューソル インコーポレイテッド Hepatitis C virus ribozyme
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
US5990276A (en) 1996-05-10 1999-11-23 Schering Corporation Synthetic inhibitors of hepatitis C virus NS3 protease
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
US5837257A (en) 1996-07-09 1998-11-17 Sage R&D Use of plant extracts for treatment of HIV, HCV and HBV infections
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
CA2266889A1 (en) * 1996-10-16 1998-04-23 Guangyi Wang Purine l-nucleosides, analogs and uses thereof
NZ335276A (en) 1996-10-18 2000-09-29 Vertex Pharma Inhibitors of serine proteases, particularly hepatitis C virus (HCV) NS3 (Non Structural Protein 3) protease
AU735438B2 (en) * 1996-11-12 2001-07-05 Medivir Ab Nucleosides
GB9623908D0 (en) 1996-11-18 1997-01-08 Hoffmann La Roche Amino acid derivatives
IL119833A (en) 1996-12-15 2001-01-11 Lavie David Hypericum perforatum extracts for the preparation of pharmaceutical compositions for the treatment of hepatitis
US6004933A (en) 1997-04-25 1999-12-21 Cortech Inc. Cysteine protease inhibitors
EP1009732B1 (en) 1997-06-30 2003-05-21 MERZ + CO. GmbH &amp; Co. 1-amino-alkylcyclohexane nmda receptor antagonists
ES2234144T3 (en) 1997-08-11 2005-06-16 Boehringer Ingelheim (Canada) Ltd. ANALOGS OF INHIBITING PEPTIDES OF HEPATITIS C.
DE69810822T2 (en) 1997-09-21 2003-11-20 Schering Corp Combination therapy for the removal of detectable HCV-RNA in patients with chronic hepatitis C infection
US5981709A (en) 1997-12-19 1999-11-09 Enzon, Inc. α-interferon-polymer-conjugates having enhanced biological activity and methods of preparing the same
EP2390257A1 (en) 1998-02-25 2011-11-30 Emory University 2'-fluoronucleosides
GB9806815D0 (en) 1998-03-30 1998-05-27 Hoffmann La Roche Amino acid derivatives
ES2172288T3 (en) 1998-05-15 2002-09-16 Schering Corp COMBINATION THERAPY THAT UNDERSTANDS RIBAVIRINA AND INTERFERED ALPHA IN PATIENTS WHO HAVE NOT BEEN SUBJECTED TO ANTIVIRAL TREATMENT AND HAVE CHRONIC HEPATITIS C INFECTION.
SI1087778T1 (en) 1998-06-08 2006-02-28 Hoffmann La Roche Use of peg-ifn-alpha and ribavirin for the treatment of chronic hepatitis c
DK1431304T3 (en) 1998-08-10 2015-03-02 Novartis Ag Beta - L-2'-deoxynucleosides for the treatment of hepatitis B
US6323180B1 (en) 1998-08-10 2001-11-27 Boehringer Ingelheim (Canada) Ltd Hepatitis C inhibitor tri-peptides
FR2784892B1 (en) 1998-10-23 2001-04-06 Smith & Nephew Kinetec Sa FOLDABLE PASSIVE MOBILIZATION BRACKET FOR LOWER MEMBER
CA2354536A1 (en) 1998-12-18 2000-06-29 Schering Corporation Ribavirin-pegylated interferon alfa induction hcv combination therapy
US6566365B1 (en) 1999-11-04 2003-05-20 Biochem Pharma Inc. Method for the treatment of Flaviviridea viral infection using nucleoside analogues
WO2001060315A2 (en) * 2000-02-18 2001-08-23 Shire Biochem Inc. Method for the treatment or prevention of flavivirus infections using nucleoside analogues
KR20100003313A (en) 2000-04-13 2010-01-07 파마셋 인코포레이티드 3'- or 2'-hydroxymethyl substituted nucleoside derivatives for treatment of hepatitis virus infections
WO2001081359A1 (en) 2000-04-20 2001-11-01 Schering Corporation Ribavirin-interferon alfa combination therapy for eradicating detectable hcv-rna in patients having chronic hepatitis c infection
MY164523A (en) * 2000-05-23 2017-12-29 Univ Degli Studi Cagliari Methods and compositions for treating hepatitis c virus
YU92202A (en) * 2000-05-26 2006-01-16 Idenix (Cayman) Limited Methods and compositions for treating flaviviruses and pestiviruses
ES2227203T3 (en) * 2000-05-26 2005-04-01 Idenix (Cayman) Limited METHODS TO TREAT INFECTION WITH THE DELTA HEPATITIS VIRUS WITH BETA-1-2'-DEOXINUCLEOSIDS.
MY141594A (en) 2000-06-15 2010-05-14 Novirio Pharmaceuticals Ltd 3'-PRODRUGS OF 2'-DEOXY-ß-L-NUCLEOSIDES
AR034127A1 (en) 2000-07-21 2004-02-04 Schering Corp IMIDAZOLIDINONES AS INHIBITORS OF NS3-SERINA PROTEASA OF THE HEPATITIS C VIRUS, PHARMACEUTICAL COMPOSITION, A METHOD FOR THEIR PREPARATION, AND THE USE OF THE SAME FOR THE MANUFACTURE OF A MEDICINAL PRODUCT
HUP0303358A3 (en) 2000-07-21 2005-10-28 Schering Corp Novel peptides as ns3-serine protease inhibitors of hepatitis c virus and pharmaceutical compositions containing them
AR029851A1 (en) 2000-07-21 2003-07-16 Dendreon Corp NEW PEPTIDES AS INHIBITORS OF NS3-SERINA PROTEASA DEL VIRUS DE HEPATITIS C
WO2002008251A2 (en) 2000-07-21 2002-01-31 Corvas International, Inc. Peptides as ns3-serine protease inhibitors of hepatitis c virus
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
AU2002213343A1 (en) 2000-10-18 2002-04-29 Schering Corporation Ribavirin-pegylated interferon alfa HCV combination therapy
KR101005299B1 (en) 2000-10-18 2011-01-04 파마셋 인코포레이티드 Modified nucleosides for treatment of viral infections and abnormal cellular proliferation
ES2263687T3 (en) 2000-11-20 2006-12-16 Bristol-Myers Squibb Company TRIPEPTIDIC INHIBITORS OF HEPATITIS C.
EP1343807B1 (en) 2000-12-12 2009-04-29 Schering Corporation Diaryl peptides as ns3-serine protease inhibitors of hepatits c virus
AU2002230764A1 (en) 2000-12-13 2002-06-24 Bristol-Myers Squibb Pharma Company Imidazolidinones and their related derivatives as hepatitis c virus ns3 protease inhibitors
WO2002048116A2 (en) 2000-12-13 2002-06-20 Bristol-Myers Squibb Pharma Company Inhibitors of hepatitis c virus ns3 protease
EP1366055A2 (en) 2000-12-15 2003-12-03 Pharmasset Limited Antiviral agents for treatment of flaviviridae infections
CZ20032005A3 (en) 2001-01-22 2004-04-14 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
WO2003024461A1 (en) 2001-09-20 2003-03-27 Schering Corporation Hcv combination therapy
WO2003068244A1 (en) * 2002-02-13 2003-08-21 Merck & Co., Inc. Methods of inhibiting orthopoxvirus replication with nucleoside compounds
CN1653077A (en) * 2002-05-06 2005-08-10 健亚生物科技公司 Nucleoside derivatives for treating hepatitis C virus infection
AU2003251524A1 (en) * 2002-06-17 2003-12-31 Isis Pharmaceuticals, Inc. Carbocyclic nucleoside analogs as RNA-antivirals
WO2004003000A2 (en) * 2002-06-28 2004-01-08 Idenix (Cayman) Limited 1’-, 2'- and 3'- modified nucleoside derivatives for treating flaviviridae infections
CN104193791A (en) * 2002-06-28 2014-12-10 埃迪尼克斯医药公司 Modified 2' and 3'-nucleoside produgs for treating flaviridae infections
EP3521297B1 (en) 2003-05-30 2021-12-22 Gilead Pharmasset LLC Modified fluorinated nucleoside analogues

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989002733A1 (en) 1987-09-22 1989-04-06 The Regents Of The University Of California Liposomal nucleoside analogues for treating aids
EP0350287A2 (en) 1988-07-07 1990-01-10 NeXstar Pharmaceuticals, Inc. Lipid derivatives of antiviral nucleosides, liposomal incorporation and method of use
WO1990000555A1 (en) 1988-07-07 1990-01-25 Vical, 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
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
US5463092A (en) 1989-11-22 1995-10-31 Vestar, Inc. Lipid derivatives of phosphonacids for liposomal incorporation and method of use
US5194654A (en) 1989-11-22 1993-03-16 Vical, Inc. Lipid derivatives of phosphonoacids for liposomal incorporation and method of use
WO1991016920A1 (en) 1990-05-07 1991-11-14 Vical, Inc. Lipid prodrugs of salicylate and nonsteroidal anti-inflammatory drugs
WO1991018914A1 (en) 1990-05-29 1991-12-12 Vical, Inc. Synthesis of glycerol di- and triphosphate derivatives
WO1991019721A1 (en) 1990-06-13 1991-12-26 Arnold Glazier Phosphorous produgs
US5149794A (en) 1990-11-01 1992-09-22 State Of Oregon Covalent lipid-drug conjugates for drug targeting
US5256641A (en) 1990-11-01 1993-10-26 State Of Oregon Covalent polar lipid-peptide conjugates for immunological targeting
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
US5543391A (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
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
WO1993000910A1 (en) 1991-07-12 1993-01-21 Vical, Inc. Antiviral liponucleosides: treatment of hepatitis b
US5554728A (en) 1991-07-23 1996-09-10 Nexstar Pharmaceuticals, Inc. Lipid conjugates of therapeutic peptides and protease inhibitors
WO1994026273A1 (en) 1993-05-12 1994-11-24 Hostetler Karl Y Acyclovir derivatives for topical use
WO1996015132A1 (en) 1994-11-15 1996-05-23 The Regents Of The University Of California Improved antiviral prodrugs
WO1999045016A2 (en) 1998-03-06 1999-09-10 Metabasis Therapeutics, Inc. Novel prodrugs for phosphorus-containing compounds
US6312662B1 (en) 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds
WO2000052015A2 (en) 1999-03-05 2000-09-08 Metabasis Therapeutics, Inc. Novel phosphorus-containing prodrugs
WO2001018013A1 (en) 1999-09-08 2001-03-15 Metabasis Therapeutics, Inc. Prodrugs for liver specific drug delivery
WO2001047935A2 (en) 1999-12-22 2001-07-05 Metabasis Therapeutics, Inc. Novel bisamidate phosphonate prodrugs

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
BAGINSKI, S. G., PEVEAR, D. C., SEIPEL, M., SUN, S. C. C., BENETATOS, C. A., CHUNDURU, S. K., RICE, C. M., M. S. COLLETT: "Mechanism of action of a pestivirus antiviral compound", PNAS USA, vol. 97, no. 14, 2000, pages 7981 - 7986
DELAMBERT ET AL., J. MED. CHEM., vol. 37, 1994, pages 498
FIELDS, B. N., KNIPE, D. M., AND HOWLEY, P. M.,: "Fields Virology", 1996, LIPPINCOTT-RAVEN PUBLISHERS
GREENE ET AL.: "Protective Groups in Organic Synthesis, Second Edition,", 1991, JOHN WILEY AND SONS
HOSETLER, K.Y., L.M. ST?HMILLER, H.B. LENTING, H. VAN DEN BOSCH, D.D. RICHMAN: "Synthesis and antiretroviral activity of phospholipid analogs of azidothymine and other antiviral nucleosides", J. BIOL. CHEM., vol. 265, 1990, pages 61127
HOSTELLER, K.Y., D.D. RICHMAN, D.A. CARSON, L.M. STUHMILLER, G.M. T. VAN WIJK, H. VAN DEN BOSCH.: "Greatly enhanced inhibition of human immunodeficiency virus type 1 replication in CEM and HT4-6C cells by 3'-deoxythymine diphosphate dimyristoylglycerol, a lipid prodrug of 3,-deoxythymine", ANTIMICROB. AGENTS CHEMOTHER., vol. 36, 1992, pages 2025 - 2029
HUNSTON ET AL., J. MED. CHEM., vol. 27, 1984, pages 440 - 444
J AM. CHEM. SOC., vol. 121, no. 24, 1999, pages 5661 - 5664
J MED. CHEM., vol. 34, pages 1408 - 1414
J. AM. CHEM. SOC., vol. 121, no. 24, 1999, pages 5661 - 5664
J. FARKAS, COLLECT. CZECH. CHEM. COMMUN., vol. 31, 1966, pages 1535
J. FARKAS, F. SORM: "Nucleic acid components and their analogues. XCIV. Synthesis of 6-amino-9-(1-deoxy-?-D-psicofuranosyl)purine", COLLECT. CZECH. CHEM. COMMUN., vol. 32, 1967, pages 2663 - 2667, XP001016337
KHAMNEI, TORRENCE, J. MED. CHEM., vol. 39, 1996, pages 4109 - 4115
MEIER ET AL., BIOORG. MED. CHEM.
MEIER ET AL., J. MED. CHEM., vol. 22, 1979, pages 811 - 815
MITCHELL ET AL., J. CHEM. SOC. PERKIN TRANS., 1992, pages 12345
R. JONES, N. BISCHOFERGER, ANTIVIRAL RESEARCH, vol. 27, 1995, pages 1 - 17
STARRETT ET AL., J. MED. CHEM., vol. 37, 1994, pages 1857 - 1864
SYNTHETIC COMMUNICATIONS, vol. 8, no. 5, 1978, pages 327 - 333
TOWNSEND: "Chemistry of Nucleosides and Nucleotides", 1994, PLENUM PRESS

Cited By (241)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7157441B2 (en) 2000-05-23 2007-01-02 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
US10758557B2 (en) 2000-05-23 2020-09-01 Idenix Pharmaceuticals Llc Methods and compositions for treating hepatitis C virus
US7169766B2 (en) 2000-05-23 2007-01-30 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus
US9968628B2 (en) 2000-05-26 2018-05-15 Idenix Pharmaceuticals Llc Methods and compositions for treating flaviviruses and pestiviruses
US7585851B2 (en) 2000-06-15 2009-09-08 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
US7323449B2 (en) 2002-07-24 2008-01-29 Merck & Co., Inc. Thionucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
US9067945B2 (en) 2002-08-23 2015-06-30 Boehringer Ingehleim International GmbH Selective phosphodiesterase 9A inhibitors as medicaments for improving cognitive processes
US7151092B2 (en) 2002-10-31 2006-12-19 Metabasis Therapeutics, Inc. Cytarabine monophosphate prodrugs
US7148349B2 (en) 2002-10-31 2006-12-12 Metabasis Therapeutics, Inc. Cyclic phosphate diesters of 1,3-propane-1-aryl diols and their use in preparing prodrugs
US7553826B2 (en) 2002-10-31 2009-06-30 Metabasis Therapeutics, Inc. Cytarabine monophosphate prodrugs
US7498320B2 (en) 2002-10-31 2009-03-03 Metabasis Therapeutics, Inc. Cyclic phosphate diesters of 1,3-propane-1-aryl diols and their use in preparing prodrugs
US7824851B2 (en) 2002-11-15 2010-11-02 Idenix Pharmaceuticals, Inc. 2′-branched nucleosides and Flaviviridae mutation
US10525072B2 (en) 2002-11-15 2020-01-07 Idenix Pharmaceuticals Llc 2′-branched nucleosides and flaviviridae mutation
EP2604620B1 (en) 2003-05-30 2016-06-29 Gilead Pharmasset LLC Modified fluorinated nucleoside analogues
EP3521297A1 (en) 2003-05-30 2019-08-07 Gilead Pharmasset LLC Modified fluorinated nucleoside analogues
US10287311B2 (en) 2003-05-30 2019-05-14 Gilead Pharmasset Llc Modified fluorinated nucleoside analogues
EP2345657A1 (en) 2003-05-30 2011-07-20 Pharmasset, Inc. Modified fluorinated nucleoside analogues
EP2345659A1 (en) 2003-05-30 2011-07-20 Pharmasset, Inc. Modified fluorinated nucleoside analogues
EP2345658A1 (en) 2003-05-30 2011-07-20 Pharmasset, Inc. Modified fluorinated nucleoside analogues
EP4032897A1 (en) 2003-05-30 2022-07-27 Gilead Pharmasset LLC Modified fluorinated nucleoside analogues
EP2345661A1 (en) 2003-05-30 2011-07-20 Pharmasset, Inc. Modified fluorinated nucleoside analogues
EP1633766A2 (en) 2003-05-30 2006-03-15 Pharmasset, Inc. Modified fluorinated nucleoside analogues
WO2005003147A2 (en) 2003-05-30 2005-01-13 Pharmasset, Inc. Modified fluorinated nucleoside analogues
EP1633766B1 (en) * 2003-05-30 2019-03-06 Gilead Pharmasset LLC Modified fluorinated nucleoside analogues
EP2604620A1 (en) 2003-05-30 2013-06-19 Gilead Pharmasset LLC Modified fluorinated nucleoside analogues
US7666855B2 (en) 2004-02-13 2010-02-23 Metabasis Therapeutics, Inc. 2′-C-methyl nucleoside derivatives
WO2005080388A1 (en) 2004-02-20 2005-09-01 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
EP2626354A1 (en) 2004-02-20 2013-08-14 Boehringer Ingelheim International GmbH Viral polymerase inhibitors
WO2005123087A2 (en) 2004-06-15 2005-12-29 Merck & Co., Inc. C-purine nucleoside analogs as inhibitors of rna-dependent rna viral polymerase
US7534767B2 (en) 2004-06-15 2009-05-19 Merck & Co., Inc. C-purine nucleoside analogs as inhibitors of RNA-dependent RNA viral polymerase
US10577359B2 (en) 2004-09-14 2020-03-03 Gilead Pharmasset Llc Preparation of 2′-fluoro-2′-alkyl-substituted or other optionally substituted ribofuranosyl pyrimidines and purines and their derivatives
US8492539B2 (en) 2004-09-14 2013-07-23 Gilead Pharmasset Llc Preparation of 2′-fluoro-2′-alkyl-substituted or other optionally substituted ribofuranosyl pyrimidines and purines and their derivatives
JP2008514639A (en) * 2004-09-24 2008-05-08 イデニクス(ケイマン)リミテツド Methods and compositions for treating flavivirus, pestivirus and hepacivirus infections
US7879797B2 (en) 2005-05-02 2011-02-01 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US8216999B2 (en) 2005-07-20 2012-07-10 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US8278322B2 (en) 2005-08-01 2012-10-02 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US7754699B2 (en) 2005-12-09 2010-07-13 Roche Palo Alto Llc Antiviral nucleosides
US8895531B2 (en) 2006-03-23 2014-11-25 Rfs Pharma Llc 2′-fluoronucleoside phosphonates as antiviral agents
US7645745B2 (en) 2006-04-04 2010-01-12 Roche Palo Alto Llc Antiviral nucleosides
WO2007113159A1 (en) 2006-04-04 2007-10-11 F. Hoffmann-La Roche Ag 3',5'-di-o-acylated nucleosides for hcv treatment
US8859512B2 (en) 2006-05-09 2014-10-14 University College Cardiff Consultants Limited Anti-viral pyrimidine nucleoside derivatives
US8329664B2 (en) 2006-05-09 2012-12-11 University College Cardiff Consultants Limited Anti-viral pyrimidine nucleoside derivatives
US9427447B2 (en) 2006-05-09 2016-08-30 University College Cardiff Consultants Limited Anti-viral pyrimidine nucleoside derivatives
US8178520B2 (en) 2006-05-15 2012-05-15 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Macrocyclic compounds as antiviral agents
US8058260B2 (en) 2006-05-22 2011-11-15 Xenoport, Inc. 2′-C-methyl-ribofuranosyl cytidine prodrugs, pharmaceutical compositions and uses thereof
US8314062B2 (en) 2006-06-23 2012-11-20 Instituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Macrocyclic compounds as antiviral agents
US7666856B2 (en) 2006-10-10 2010-02-23 Medivir Ab Antiviral nucleosides
US8158779B2 (en) 2006-10-10 2012-04-17 Medivir Ab Antiviral nucleosides
US8912321B2 (en) 2006-10-10 2014-12-16 Gilead Pharmasset Llc Preparation of nucleosides ribofuranosyl pyrimidines
US7825239B2 (en) 2006-10-10 2010-11-02 Medivir Ab Antiviral nucleosides
US7935681B2 (en) 2006-10-10 2011-05-03 Medivir Ab Antiviral nucleosides
EP2361922A1 (en) 2006-10-10 2011-08-31 Medivir AB Intermediate to HCV-Nucleoside Inhibitors
US8138164B2 (en) 2006-10-24 2012-03-20 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US8377873B2 (en) 2006-10-24 2013-02-19 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US8309540B2 (en) 2006-10-24 2012-11-13 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US8377874B2 (en) 2006-10-27 2013-02-19 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US9738661B2 (en) 2006-10-27 2017-08-22 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
US7786110B2 (en) 2006-11-09 2010-08-31 Roche Palo Alto Llc Thiazole and oxazole-substituted arylamides as P2X3 and P2X2/3 antagonists
US7767660B2 (en) 2006-12-20 2010-08-03 Istituto Di Richerche Di Biologia Molecolare P. Angeletti Spa Antiviral indoles
US7781422B2 (en) 2006-12-20 2010-08-24 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Antiviral indoles
US8101595B2 (en) 2006-12-20 2012-01-24 Istituto di Ricerche di Biologia Molecolare P. Angletti SpA Antiviral indoles
US8691788B2 (en) 2006-12-28 2014-04-08 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US7951789B2 (en) 2006-12-28 2011-05-31 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US7902202B2 (en) 2006-12-28 2011-03-08 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
WO2008085508A2 (en) 2007-01-05 2008-07-17 Merck & Co., Inc. Nucleoside aryl phosphoramidates for the treatment of rna-dependent rna viral infection
US10183037B2 (en) 2007-03-30 2019-01-22 Gilead Pharmasset Llc Nucleoside phosphoramidate prodrugs
US8580765B2 (en) 2007-03-30 2013-11-12 Gilead Pharmasset Llc Nucleoside phosphoramidate prodrugs
US9085573B2 (en) 2007-03-30 2015-07-21 Gilead Pharmasset Llc Nucleoside phosphoramidate prodrugs
US11642361B2 (en) 2007-03-30 2023-05-09 Gilead Sciences, Inc. Nucleoside phosphoramidate prodrugs
US8906880B2 (en) 2007-03-30 2014-12-09 Gilead Pharmasset Llc Nucleoside phosphoramidate prodrugs
US9585906B2 (en) 2007-03-30 2017-03-07 Gilead Pharmasset Llc Nucleoside phosphoramidate prodrugs
US8735372B2 (en) 2007-03-30 2014-05-27 Gilead Pharmasset Llc Nucleoside phosphoramidate prodrugs
US7989438B2 (en) 2007-07-17 2011-08-02 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Therapeutic compounds
US8927569B2 (en) 2007-07-19 2015-01-06 Merck Sharp & Dohme Corp. Macrocyclic compounds as antiviral agents
US8648085B2 (en) 2007-11-30 2014-02-11 Boehringer Ingelheim International Gmbh 1, 5-dihydro-pyrazolo (3, 4-D) pyrimidin-4-one derivatives and their use as PDE9A mudulators for the treatment of CNS disorders
US8440673B2 (en) 2007-12-17 2013-05-14 Roche Palo Alto Llc Pyrazole-substituted arylamides as P2X3 and P2X2/3 antagonists
US7858654B2 (en) 2007-12-17 2010-12-28 Roche Palo Alto Llc Imidazole-substituted arylamides as P2X3 and P2X2/3 antagonists
US7989637B2 (en) 2007-12-17 2011-08-02 Roche Palo Alto Llc Triazole-substituted arylamides as P2X3 and P2X2/3 antagonists
US8048905B2 (en) 2007-12-17 2011-11-01 Roche Palo Alto Llc Tetrazole-substituted arylamides as P2X3 and P2X2/3 antagonists
US8227431B2 (en) 2008-03-17 2012-07-24 Hetero Drugs Limited Nucleoside derivatives
US8470870B2 (en) 2008-03-27 2013-06-25 Idenix Pharmaceuticals, Inc. Solid forms of an anti-HIV phosphoindole compound
US8623879B2 (en) 2008-04-02 2014-01-07 Boehringer Ingelheim International Gmbh 1-heterocyclyl-1,5-dihydro-pyrazolo[3,4-D] pyrimidin-4-one derivates and their use as PDE9A modulators
US9096603B2 (en) 2008-04-02 2015-08-04 Boehringer Ingelheim International Gmbh 1-heterocyclyl-1,5-dihydro-pyrazolo[3,4-D] pyrimidin-4-one derivatives and their use as PDE9A modulators
EP2268288A4 (en) * 2008-04-15 2012-05-30 Rfs Pharma Llc Nucleoside derivatives for treatment of caliciviridae infections, including norovirus infections
EP3042660A2 (en) 2008-04-15 2016-07-13 RFS Pharma, LLC. Nucleoside derivatives for treatment of caliciviridae infections, including norovirus infections
US8415321B2 (en) 2008-04-15 2013-04-09 Raymond F. Schinazi Nucleoside derivatives for treatment of Caliciviridae infections, including Norovirus infections
EP2268288A2 (en) * 2008-04-15 2011-01-05 RFS Pharma, LLC. Nucleoside derivatives for treatment of caliciviridae infections, including norovirus infections
WO2009129120A2 (en) 2008-04-15 2009-10-22 Rfs Pharma, Llc Nucleoside derivatives for treatment of caliciviridae infections, including norovirus infections
US8461107B2 (en) 2008-04-28 2013-06-11 Merck Sharp & Dohme Corp. HCV NS3 protease inhibitors
EP2141172A1 (en) 2008-07-01 2010-01-06 Ortho Biotech Products L.P. Cyclopropyl polymerase inhibitors
WO2010000459A1 (en) * 2008-07-01 2010-01-07 Centocor Ortho Biotech Products L.P. Cyclopropyl polymerase inhibitors
EA022754B1 (en) * 2008-07-01 2016-02-29 СЕНТОКОР ОРТО БАЙОТЕК ПРОДАКТС Эл.Пи. Cyclopropyl polymerase inhibitors
CN105693794A (en) * 2008-07-01 2016-06-22 杨森产品有限公司 Cyclopropyl polymerase inhibitors
US20110092460A1 (en) * 2008-07-01 2011-04-21 Centocor Ortho Biotech Products L.P. Cyclopropyl polymerase inhibitors
CN102083845B (en) * 2008-07-01 2016-09-14 森托科尔奥索生物科技产品股份有限公司 Cyclopropyl polymerase inhibitors
US8431588B2 (en) 2008-07-01 2013-04-30 Janssen Products, Lp Cyclopropyl polymerase inhibitors
AU2009266004B2 (en) * 2008-07-01 2014-01-30 Centocor Ortho Biotech Products L.P. Cyclopropyl polymerase inhibitors
EP2476690A1 (en) 2008-07-02 2012-07-18 IDENIX Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
EP2540349A1 (en) 2008-07-22 2013-01-02 Merck Sharp & Dohme Corp. Pharmaceutical compositions comprising a macrocyclic quinoxaline compound which is an HCV NS3 protease inhibitor
EP2540350A1 (en) 2008-07-22 2013-01-02 Merck Sharp & Dohme Corp. Combinations of a macrocyclic quinoxaline compound which is an HCV NS3 protease inhibitors with other HCV agents
US8080654B2 (en) 2008-07-22 2011-12-20 Insituto di Ricerche di Biologia Molecolare P. Angeletti SpA Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
US7973040B2 (en) 2008-07-22 2011-07-05 Merck Sharp & Dohme Corp. Macrocyclic quinoxaline compounds as HCV NS3 protease inhibitors
US9079905B2 (en) 2008-09-08 2015-07-14 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
US8629275B2 (en) 2008-09-08 2014-01-14 Merck Sharp & Dohme Corp. AHCY hydrolase inhibitors for treatment of hyper homocysteinemia
CN102256991A (en) * 2008-12-08 2011-11-23 森托科尔奥索生物科技产品股份有限公司 Uracyl cyclopropyl nucleotides
AU2009326125B2 (en) * 2008-12-08 2015-03-12 Centocor Ortho Biotech Products L.P. Uracyl cyclopropyl nucleotides
EA020244B1 (en) * 2008-12-08 2014-09-30 СЕНТОКОР ОРТО БАЙОТЕК ПРОДАКТС Эл.Пи. Uracyl cyclopropyl nucleotides
US8399429B2 (en) 2008-12-08 2013-03-19 Janssen Products, Lp Uracyl cyclopropyl nucleotides
CN102256991B (en) * 2008-12-08 2015-08-12 森托科尔奥索生物科技产品股份有限公司 Uracyl cyclopropyl nucleotides
WO2010066699A1 (en) * 2008-12-08 2010-06-17 Centocor Ortho Biotech Products L.P. Uracyl cyclopropyl nucleotides
US8957045B2 (en) 2008-12-23 2015-02-17 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8551973B2 (en) 2008-12-23 2013-10-08 Gilead Pharmasset Llc Nucleoside analogs
US8716262B2 (en) 2008-12-23 2014-05-06 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8716263B2 (en) 2008-12-23 2014-05-06 Gilead Pharmasset Llc Synthesis of purine nucleosides
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
WO2010084115A2 (en) 2009-01-20 2010-07-29 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Antiviral agents
WO2010101967A2 (en) 2009-03-04 2010-09-10 Idenix Pharmaceuticals, Inc. Phosphothiophene and phosphothiazole hcv polymerase inhibitors
US8623901B2 (en) 2009-03-31 2014-01-07 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
US9102679B2 (en) 2009-03-31 2015-08-11 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
US8735569B2 (en) 2009-05-20 2014-05-27 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8629263B2 (en) 2009-05-20 2014-01-14 Gilead Pharmasset Llc Nucleoside phosphoramidates
US9284342B2 (en) 2009-05-20 2016-03-15 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8642756B2 (en) 2009-05-20 2014-02-04 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8633309B2 (en) 2009-05-20 2014-01-21 Gilead Pharmasset Llc Nucleoside phosphoramidates
US9206217B2 (en) 2009-05-20 2015-12-08 Gilead Pharmasset Llc Nucleoside phosphoramidates
US9637512B2 (en) 2009-05-20 2017-05-02 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8618076B2 (en) 2009-05-20 2013-12-31 Gilead Pharmasset Llc Nucleoside phosphoramidates
US8093275B2 (en) 2009-06-22 2012-01-10 Roche Palo Alto Llc Oxazolone and pyrrolidinone-substituted pryidine amides as P2X3 and P2X2/3 antagonists
US8283383B2 (en) 2009-06-22 2012-10-09 Roche Palo Alto Llc Biphenyl amides as P2X3 and P2X2/3 antagonists
US8476457B2 (en) 2009-06-22 2013-07-02 Roche Palo Alto Llc Indole, indazole and benzimidazole arylamides as P2X3 and P2X2/3 antagonists
WO2011014487A1 (en) 2009-07-30 2011-02-03 Merck Sharp & Dohme Corp. Hepatitis c virus ns3 protease inhibitors
US8828930B2 (en) 2009-07-30 2014-09-09 Merck Sharp & Dohme Corp. Hepatitis C virus NS3 protease inhibitors
WO2011017389A1 (en) 2009-08-05 2011-02-10 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors useful against viral infections, particularly hcv
WO2011058084A1 (en) 2009-11-14 2011-05-19 F. Hoffmann-La Roche Ag Biomarkers for predicting rapid response to hcv treatment
WO2011063076A1 (en) 2009-11-19 2011-05-26 Itherx Pharmaceuticals, Inc. Methods of treating hepatitis c virus with oxoacetamide compounds
WO2011067195A1 (en) 2009-12-02 2011-06-09 F. Hoffmann-La Roche Ag Biomarkers for predicting sustained response to hcv treatment
WO2011075615A1 (en) 2009-12-18 2011-06-23 Idenix Pharmaceuticals, Inc. 5,5-fused arylene or heteroarylene hepatitis c virus inhibitors
US8859756B2 (en) 2010-03-31 2014-10-14 Gilead Pharmasset Llc Stereoselective synthesis of phosphorus containing actives
WO2011123586A1 (en) 2010-04-01 2011-10-06 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US8680071B2 (en) 2010-04-01 2014-03-25 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US9328120B2 (en) 2010-08-12 2016-05-03 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
US8912201B2 (en) 2010-08-12 2014-12-16 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
US9346848B2 (en) 2010-09-22 2016-05-24 Alios Biopharma, Inc. Azido nucleosides and nucleotide analogs
US8877731B2 (en) 2010-09-22 2014-11-04 Alios Biopharma, Inc. Azido nucleosides and nucleotide analogs
WO2012080050A1 (en) 2010-12-14 2012-06-21 F. Hoffmann-La Roche Ag Solid forms of a phenoxybenzenesulfonyl compound
WO2012109398A1 (en) 2011-02-10 2012-08-16 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating hcv infections
US8809345B2 (en) 2011-02-15 2014-08-19 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
WO2012154321A1 (en) 2011-03-31 2012-11-15 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
WO2012135581A1 (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
WO2012142523A2 (en) 2011-04-13 2012-10-18 Gilead Sciences, Inc. 1'-substituted pyrimidine n-nucleoside analogs for antiviral treatment
WO2013039855A1 (en) 2011-09-12 2013-03-21 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US8951985B2 (en) 2011-09-12 2015-02-10 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
WO2013039920A1 (en) 2011-09-12 2013-03-21 Idenix Pharmaceuticals, Inc. Substituted carbonyloxymethylphosphoramidate compounds and pharmaceutical compositions for the treatment of viral infections
US10456414B2 (en) 2011-09-16 2019-10-29 Gilead Pharmasset Llc Methods for treating HCV
WO2013056046A1 (en) 2011-10-14 2013-04-18 Idenix Pharmaceuticals, Inc. Substituted 3',5'-cyclic phosphates of purine nucleotide compounds and pharmaceutical compositions for the treatment of viral infections
US9328138B2 (en) 2011-11-15 2016-05-03 Msd Italia S.R.L. HCV NS3 protease inhibitors
WO2013074386A2 (en) 2011-11-15 2013-05-23 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
EP3750544A2 (en) 2011-11-30 2020-12-16 Emory University Jak inhibitors for use in the prevention or treatment of viral infection
US9073960B2 (en) 2011-12-22 2015-07-07 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US11021509B2 (en) 2011-12-22 2021-06-01 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US10464965B2 (en) 2011-12-22 2019-11-05 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
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]
US10485815B2 (en) 2012-03-21 2019-11-26 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
USRE48171E1 (en) 2012-03-21 2020-08-25 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
WO2013177188A1 (en) 2012-05-22 2013-11-28 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
WO2013177195A1 (en) 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. 3',5'-cyclic phosphate prodrugs for hcv infection
US9845336B2 (en) 2012-05-25 2017-12-19 Janssen Sciences Ireland Uc Uracyl spirooxetane nucleosides
US10774106B2 (en) 2012-05-25 2020-09-15 Janssen Sciences Ireland Unlimited Company Uracyl spirooxetane nucleosides
US10301347B2 (en) 2012-05-25 2019-05-28 Janssen Sciences Ireland Unlimited Company Uracyl spirooxetane nucleosides
US10544184B2 (en) 2012-05-25 2020-01-28 Janssen Sciences Ireland Unlimited Company Uracyl spirooxetane nucleosides
US10040814B2 (en) 2012-05-25 2018-08-07 Janssen Sciences Ireland Uc Uracyl spirooxetane nucleosides
WO2014058801A1 (en) 2012-10-08 2014-04-17 Idenix Pharmaceuticals, Inc. 2'-chloro nucleoside analogs for hcv infection
WO2014063019A1 (en) 2012-10-19 2014-04-24 Idenix Pharmaceuticals, Inc. Dinucleotide compounds for hcv infection
WO2014066239A1 (en) 2012-10-22 2014-05-01 Idenix Pharmaceuticals, Inc. 2',4'-bridged nucleosides for hcv infection
WO2014078427A1 (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
WO2014099941A1 (en) 2012-12-19 2014-06-26 Idenix Pharmaceuticals, Inc. 4'-fluoro nucleosides for the treatment of hcv
US10683320B2 (en) 2012-12-21 2020-06-16 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US10144755B2 (en) 2012-12-21 2018-12-04 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US10487104B2 (en) 2012-12-21 2019-11-26 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US9243022B2 (en) 2012-12-21 2016-01-26 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US10793591B2 (en) 2012-12-21 2020-10-06 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US10112966B2 (en) 2012-12-21 2018-10-30 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US9249174B2 (en) 2012-12-21 2016-02-02 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US11485753B2 (en) 2012-12-21 2022-11-01 Janssen Pharmaceutica Nv Substituted nucleosides, nucleotides and analogs thereof
US10039779B2 (en) 2013-01-31 2018-08-07 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
WO2014123795A2 (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
WO2014123794A1 (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
WO2014137926A1 (en) 2013-03-04 2014-09-12 Idenix Pharmaceuticals, Inc. 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
WO2014148949A1 (en) 2013-03-22 2014-09-25 Асави, Ллс Alkyl 2-{[(2r,3s,5r)-5-(4-amino-2-oxo-2н-pyrimidin-1-yl)-3-hydroxy- tetrahydro-furan-2-yl-methoxy]-phenoxy-phosphoryl-amino}-propionates, nucleoside inhibitors of hcv ns5b rna-polymerase, and methods for producing and use thereof
WO2014165542A1 (en) 2013-04-01 2014-10-09 Idenix Pharmaceuticals, Inc. 2',4'-fluoro nucleosides for the treatment of hcv
US9447132B2 (en) 2013-04-12 2016-09-20 Achillion Pharmaceuticals, Inc. Highly active nucleoside derivative for the treatment of HCV
WO2014197578A1 (en) 2013-06-05 2014-12-11 Idenix Pharmaceuticals, Inc. 1',4'-thio nucleosides for the treatment of hcv
WO2015017713A1 (en) 2013-08-01 2015-02-05 Idenix Pharmaceuticals, Inc. D-amino acid phosphoramidate pronucleotides of halogeno pyrimidine compounds for liver disease
US11707479B2 (en) 2013-08-27 2023-07-25 Gilead Sciences, Inc. Combination formulation of two antiviral compounds
US11116783B2 (en) 2013-08-27 2021-09-14 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
US11857560B2 (en) 2013-09-11 2024-01-02 Emory University Pharmaceutical compositions comprising substituted nucleotides and nucleosides for treating viral infections
US11166973B2 (en) 2013-09-11 2021-11-09 Emory University Substituted nucleotides and nucleosides for treating viral infections
WO2015042375A1 (en) 2013-09-20 2015-03-26 Idenix Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US9862743B2 (en) 2013-10-11 2018-01-09 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US10370401B2 (en) 2013-10-11 2019-08-06 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
WO2015054465A1 (en) * 2013-10-11 2015-04-16 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
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
WO2015066370A1 (en) 2013-11-01 2015-05-07 Idenix Pharmaceuticals, Inc. D-alanine phosphoramidate pronucleotides of 2'-methyl 2'-fluoro guanosine nucleoside compounds for the treatment of hcv
WO2015081297A1 (en) 2013-11-27 2015-06-04 Idenix Pharmaceuticals, Inc. 2'-dichloro and 2'-fluoro-2'-chloro nucleoside analogues for hcv infection
US10683321B2 (en) 2013-12-18 2020-06-16 Idenix Pharmaceuticals Llc 4′-or nucleosides for the treatment of HCV
WO2015095419A1 (en) 2013-12-18 2015-06-25 Idenix Pharmaceuticals, Inc. 4'-or nucleosides for the treatment of hcv
US11278559B2 (en) 2014-02-13 2022-03-22 Ligand Pharmaceuticals Incorporated Prodrug compounds and their uses
US10449210B2 (en) 2014-02-13 2019-10-22 Ligand Pharmaceuticals Inc. Prodrug compounds and their uses
WO2015134780A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Solid prodrug forms of 2'-chloro-2'-methyl uridine for hcv
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
WO2015134560A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Solid forms of a flaviviridae virus inhibitor compound and salts thereof
WO2015161137A1 (en) 2014-04-16 2015-10-22 Idenix Pharmaceuticals, Inc. 3'-substituted methyl or alkynyl nucleosides for the treatment of hcv
CN103992360A (en) * 2014-04-21 2014-08-20 中美华世通生物医药科技(武汉)有限公司 Reduction method suitable for industrialized production
US9994600B2 (en) 2014-07-02 2018-06-12 Ligand Pharmaceuticals, Inc. Prodrug compounds and uses therof
US10150788B2 (en) 2014-07-02 2018-12-11 Ligand Pharmaceuticals, Inc. Prodrug compounds and uses thereof
US10005811B2 (en) 2015-03-06 2018-06-26 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10875885B2 (en) 2015-03-06 2020-12-29 Atea Pharmaceuticals, Inc. β-d-2′-deoxy-2′-α-fluoro-2′-β-c-substituted-2-modified-n6-substituted purine nucleotides for HCV treatment
US10239911B2 (en) 2015-03-06 2019-03-26 Atea Pharmaceuticals, Inc. Beta-D-2′-deoxy-2′-alpha-fluoro-2′-beta-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10870672B2 (en) 2015-03-06 2020-12-22 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10870673B2 (en) 2015-03-06 2020-12-22 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10815266B2 (en) 2015-03-06 2020-10-27 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US9828410B2 (en) 2015-03-06 2017-11-28 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US10000523B2 (en) 2015-03-06 2018-06-19 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
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
US10946033B2 (en) 2016-09-07 2021-03-16 Atea Pharmaceuticals, Inc. 2′-substituted-N6-substituted purine nucleotides for RNA virus treatment
US11975016B2 (en) 2016-09-07 2024-05-07 Atea Pharmaceuticals, Inc. 2′-substituted-N6-substituted purine nucleotides for RNA virus treatment
US11452714B2 (en) 2017-05-23 2022-09-27 Regents Of The University Of Minnesota Antibacterial agents including histidine kinase inhibitors
WO2018217884A1 (en) * 2017-05-23 2018-11-29 Regents Of The University Of Minnesota Antibacterial agents including histidine kinase inhibitors
US11690860B2 (en) 2018-04-10 2023-07-04 Atea Pharmaceuticals, Inc. Treatment of HCV infected patients with cirrhosis
US11767337B2 (en) 2020-02-18 2023-09-26 Gilead Sciences, Inc. Antiviral compounds
US11707480B2 (en) 2020-02-27 2023-07-25 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US11738038B2 (en) 2020-02-27 2023-08-29 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US11813278B2 (en) 2020-02-27 2023-11-14 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
US10874687B1 (en) 2020-02-27 2020-12-29 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
WO2021221983A1 (en) * 2020-04-28 2021-11-04 Southlake Pharmaceuticals, Inc. Interferon tau as antiviral therapy
US11697666B2 (en) 2021-04-16 2023-07-11 Gilead Sciences, Inc. Methods of preparing carbanucleosides using amides

Also Published As

Publication number Publication date
CN103319554A (en) 2013-09-25
UY27875A1 (en) 2003-12-31
EP2332952B1 (en) 2015-04-29
WO2004002999A8 (en) 2005-02-17
PL374831A1 (en) 2005-10-31
JP2010280660A (en) 2010-12-16
IL165893A0 (en) 2006-01-15
CY2014031I1 (en) 2016-06-22
DK1523489T3 (en) 2014-06-16
NO330755B1 (en) 2011-07-04
HK1204625A1 (en) 2015-11-27
AU2003247084B2 (en) 2010-02-11
BR0312286A (en) 2007-06-19
AU2003247084B9 (en) 2018-07-26
CY2014031I2 (en) 2016-06-22
WO2004002999A3 (en) 2004-08-12
CN102424698A (en) 2012-04-25
NO2014019I1 (en) 2014-07-30
AR043101A1 (en) 2005-07-20
UY27874A1 (en) 2003-12-31
TW200500373A (en) 2005-01-01
JP2015205885A (en) 2015-11-19
PE20040634A1 (en) 2004-12-03
ES2469569T3 (en) 2014-06-18
TW200500375A (en) 2005-01-01
EP2799442A1 (en) 2014-11-05
JP2018076342A (en) 2018-05-17
PE20040636A1 (en) 2004-12-03
EP2332952A1 (en) 2011-06-15
KR20050048544A (en) 2005-05-24
UY27876A1 (en) 2003-12-31
CA2490191A1 (en) 2004-01-08
PE20040635A1 (en) 2004-12-03
JP6069410B2 (en) 2017-02-01
RS114004A (en) 2007-02-05
RU2011133741A (en) 2013-02-20
CN100348607C (en) 2007-11-14
HK1204624A1 (en) 2015-11-27
CA2490191C (en) 2010-08-03
JP2011251983A (en) 2011-12-15
CN104193791A (en) 2014-12-10
JP6069169B2 (en) 2017-02-01
JP2017061480A (en) 2017-03-30
AR043298A1 (en) 2005-07-27
AU2003247084B8 (en) 2010-03-11
JP2005533817A (en) 2005-11-10
NO20050465L (en) 2005-01-27
PT1523489E (en) 2014-06-24
DE14169110T1 (en) 2022-05-12
JP2014058537A (en) 2014-04-03
EP1523489A2 (en) 2005-04-20
AP2005003212A0 (en) 2005-03-31
MXPA04012709A (en) 2005-09-30
BR0312271A (en) 2007-11-06
JP6309590B2 (en) 2018-04-11
EP1523489B1 (en) 2014-03-12
MY140819A (en) 2010-01-29
CN104163841B (en) 2016-08-24
NO20110343A1 (en) 2011-03-04
MA27375A1 (en) 2005-06-01
CN1678621A (en) 2005-10-05
CN103275159A (en) 2013-09-04
TW200500374A (en) 2005-01-01
AU2003247084A1 (en) 2004-01-19
RU2483075C2 (en) 2013-05-27
HK1206361A1 (en) 2016-01-08
CN104163841A (en) 2014-11-26
CY1115417T1 (en) 2016-06-22
JP5463332B2 (en) 2014-04-09

Similar Documents

Publication Publication Date Title
US20210024565A1 (en) Modified 2&#39; and 3&#39;-nucleoside prodrugs for treating flaviviridae infections
AU2003247084B8 (en) Modified 2&#39; and 3&#39;-nucleoside prodrugs for treating flaviridae infections
WO2004003000A2 (en) 1’-, 2&#39;- and 3&#39;- modified nucleoside derivatives for treating flaviviridae infections
RU2437892C2 (en) Modified 2- and 3-nucleosides and use thereof to produce medicinal agent, having hepatitis c inhibiting activity

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: P-1140/04

Country of ref document: YU

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2490191

Country of ref document: CA

Ref document number: PA/a/2004/012709

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 200410249

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2004517158

Country of ref document: JP

Ref document number: 1020047021286

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 374831

Country of ref document: PL

WWE Wipo information: entry into national phase

Ref document number: 537663

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2003761744

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2003247084

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1-2005-500173

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2005102013

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 343/DELNP/2005

Country of ref document: IN

Ref document number: 1200500107

Country of ref document: VN

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 02/2004 UNDER (71, 72) DELETE "CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE ¢FR/FR!; 3, RUE MICHEL-ANGE, F-75794 PARIS CéDEX 16 (FR)."; UNDER (71) ADD "CENTRE NATIONAL DE LA RECHERCHE SCIENTIFIQUE ¢FR/FR!; 3, RUEMICHEL-ANGE, F-75794 PARIS CéDEX 16 (FR)"

WWE Wipo information: entry into national phase

Ref document number: 20038205017

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2003761744

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020047021286

Country of ref document: KR

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: PI0312286

Country of ref document: BR

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)