US20100291031A2 - Bicyclic nucleosides and nucleotides as therapeutic agents - Google Patents

Bicyclic nucleosides and nucleotides as therapeutic agents Download PDF

Info

Publication number
US20100291031A2
US20100291031A2 US11/908,019 US90801906A US2010291031A2 US 20100291031 A2 US20100291031 A2 US 20100291031A2 US 90801906 A US90801906 A US 90801906A US 2010291031 A2 US2010291031 A2 US 2010291031A2
Authority
US
United States
Prior art keywords
optionally substituted
formula
bond
compound
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/908,019
Other versions
US20090004138A1 (en
US8802840B2 (en
Inventor
Paula Francom
Roland Nearn
Alistair Draffan
John Lambert
Silas Bond
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biota Scientific Management Pty Ltd
Original Assignee
Biota Scientific Management Pty Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Biota Scientific Management Pty Ltd filed Critical Biota Scientific Management Pty Ltd
Priority to US11/908,019 priority Critical patent/US8802840B2/en
Assigned to BIOTA SCIENTIFIC MANAGEMENT PTY LTD reassignment BIOTA SCIENTIFIC MANAGEMENT PTY LTD ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NEARN, ROLAND HENRY, BOND, SILAS, LAMBERT, JOHN NICHOLAS, DRAFFAN, ALISTAIR GEORGE, FRANCOM, PAULA
Publication of US20090004138A1 publication Critical patent/US20090004138A1/en
Publication of US20100291031A2 publication Critical patent/US20100291031A2/en
Application granted granted Critical
Publication of US8802840B2 publication Critical patent/US8802840B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

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
    • C07H19/22Pteridine radicals
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/23Heterocyclic radicals containing two or more heterocyclic rings condensed among themselves or condensed with a common carbocyclic ring system, not provided for in groups C07H19/14 - C07H19/22

Definitions

  • the present invention relates to the use of bicyclic nucleosides and nucleotides for the treatment of infectious diseases, including viral infections, novel bicyclic nucleosides and nucleotides and methods for their manufacture.
  • BBV blood-borne viruses
  • HCV hepatitis C virus
  • HBV hepatitis B virus
  • HBV human immunodeficiency virus
  • the Flaviviridae is a group of positive single-stranded RNA viruses with a genome size from 9-15 kb.
  • the Flaviviridae consists of various genera including:
  • Flaviviruses This genus includes the Dengue virus, Japanese Tick-Borne and the Yellow Fever virus. Apart from these major groups, there are some additional Flaviviruses that are unclassified.
  • Hepaciviruses This genus contains only one species, the Hepatitis C virus (HCV), which is composed of many genotypes and subtypes.
  • Hepatitis C virus comprises a positive-strand RNA genome enclosed in a nucleocapsid and lipid envelope and consists of approximately 9600 ribonucleotides, which encodes a polyprotein of about 3000 amino acids (Dymock et al. Antiviral Chemistry & Chemotherapy 2000, 11, 79).
  • the reproduction of HCV virus may be prevented through the manipulation of NS5B's polymerase activity.
  • the inhibition of NS5B protein would suppress or prevent the formation of the double-stranded HCV RNA.
  • a nucleoside analog also may be incorporated into the extending RNA strand and act as a chain-terminator.
  • a deteriorating nucleoside analog also may be incorporated into the extending RNA, which may cause mutagenic damage to the viral genome.
  • HBV has acutely infected almost a third of the world's human population, and about 5% of the infected are chronic carriers of the virus (Delaney I V et al., Antiviral Chemistry & Chemotherapy 2001, 12, 1-35). Chronic HBV infection causes liver damage that frequently progresses to cirrhosis and/or liver cancer later in the life. Despite the availability and widespread use of effective vaccines and chemotherapy, the number of chronic carriers approaches 400 million worldwide. Therefore, more effective anti-HBV drugs need to be developed.
  • HIV causes progressive degeneration of the immune system, leading to the development of AIDS.
  • a number of drugs have been used clinically, including reverse transcriptase inhibitors and protease inhibitors.
  • combination therapies are used widely for the treatment of AIDS in order to reduce the drug resistance.
  • AIDS is still one of the leading epidemic diseases.
  • Bacterial infections have long been the sources of many infectious diseases. The widespread use of antibiotics has produced many new strains of life-threatening antibiotic resistant bacteria. Fungal infections are another type of infectious diseases, some of which also can be life-threatening. There is an ever increasing demand for the treatment of bacterial and fungal infections. As such, antimicrobial drugs based on new mechanisms of action are especially important.
  • Nucleoside drugs have been used clinically for decades for the treatment of viral infections and proliferative disorders such as cancer. Most of the nucleoside drugs are classified as antimetabolites. After they enter cells, nucleoside analogs are phosphorylated successively to nucleoside 5′-monophosphates, 5′-diphosphates, and 5′-triphosphates.
  • nucleoside triphosphates e.g., 3′-azido-3′-deoxythymidine triphosphate (AZT, an anti-HIV drug) and arabinosylcytosine triphosphate (cytarabine, an anticancer drug
  • ZT 3′-azido-3′-deoxythymidine triphosphate
  • cytarabine arabinosylcytosine triphosphate
  • nucleoside analogs exert effects at lower phosphate levels.
  • nucleoside analogs themselves may act at the nonphosphate level such as the inhibitors of adenosine kinases and the ligands of adenosine receptors, currently, clinically-useful nucleoside drugs primarily depend on cellular activation by nucleoside kinases and nucleotide kinases.
  • nucleoside antiviral drugs Two criteria are pertinent for nucleoside antiviral drugs: 1) nucleoside analogs should anabolise to nucleotides in cells; and 2) the anabolised nucleotides should target selectively viral enzymes.
  • nucleoside analogs In order to be phosphorylated in cells and selectively target preferred enzymes, nucleoside analogs should have favourable modifications on their sugar and base moieties.
  • a general approach is to generate diverse nucleoside analogs by modifying the base or the sugar, or by modifying both base and sugar moieties. Numerous examples exist in the literature for the synthesis of a variety of modified nucleosides ( Chemistry of Nucleosides and Nucleotides Vol. 1 (1988), Vol.
  • a class of such compounds is bicyclic nucleosides which are not derived from purine bases. Disclosures of bicyclic nucleosides are very limited considering that natural adenine and guanine (purines) based ribonucleotides and deoxy derivatives thereof, have bicyclic base moieties.
  • WO 01/92282 A2 discloses derivatives of purine nucleosides.
  • WO 01/90121 A2 and WO 04/058792 disclose derivatives of purine nucleosides.
  • the present invention discloses that a certain new class of bicyclic nucleosides and nucleotides display biological activity which may be particularly useful for the treatment of infectious diseases, including viral infections.
  • the present invention relates to a particular class of bicyclic nucleosides, nucleotides, and derivatives thereof and their use in the treatment of microbial infections, and specifically viral infections.
  • the present invention provides a method for the treatment of a microbial infection, comprising administering an effective amount of a compound of the formula (I) or a pharmaceutically acceptable salt thereof; wherein:
  • A is O, S, CH 2 , CHF, CF 2 or NR;
  • R 1 , R 2 , R 2′ , R 3 , R 3′ , and R 4 are independently selected from the group consisting of H, halogen, OH, N 3 , CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkynoxy, optionally substituted aryloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted arylalkoxy, optionally substituted heterocycloxy, optionally substituted heteroaryloxy, optionally substituted cycloalkoxy, optionally substituted cycloalkenoxy, optionally substituted amino
  • R 4′ is —CY 2 SH, —CY 2 OH, —CY 2 NH 2 , or -L′-R 5 ;
  • L′ is selected from the group consisting of —CY 2 —, —CY 2 CY 2 —, —CY 2 OCY 2 —, —CY 2 SCY 2 — and —CY 2 NHCY 2 —;
  • each Y is independently selected from the group consisting of H, F, Cl, Br, OR, C 1 -C 6 alkyl, C 2 -C 6 alkenyl and C 2 -C 6 alkynyl, wherein C 1 -C 6 alkyl, C 2 -C 6 alkenyl, and C 2 -C 6 alkynyl may be optionally substituted with one or more groups selected from F, Cl, Br, OH, COOH, COOCH 3 , SH, SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 , CN, NO 2 , C(O)NH 2 , C(O)NHCH 3 , N 3 , C(S)NH 2 , OCH 3 , and OCH 2 CH 3 ;
  • R 5 is selected from the group consisting of OR, NR 2 , monophosphate, diphosphate, and triphosphate, or a mono, di or triphosphate mimic;
  • each R is independently selected from the group consisting of H, CF 3 , optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
  • Z is independently selected from N or C-G; or, if Z is not a participant in a ⁇ bond (double bond), Z is independently selected from O, S, Se, NR, NOR, NNR 2 , CO, CS, CNR, SO, S(O) 2 , SeO, Se(O) 2 or C(G) 2 , wherein each G is independently selected from the group consisting of H, halogen, OR, SR, NR 2 , NROR, N 3 , COOR, CN, CONR 2 , C(S)NR 2 , C( ⁇ NR)NR 2 , and R; and
  • any two adjacent Z are not both selected from O, S, and Se, or not both selected from CO, CS, CNR, SO, S(O) 2 , SeO, and Se(O) 2 ;
  • X is a participant in a ⁇ bond (double bond), X is C; or if X is not a participant in a ⁇ bond (double bond), X is CR or N;
  • R′′ is a participant in a ⁇ bond (double bond)
  • R′′ is O, S, Se, NR, NOR or NNR 2 ; or if R′′ is not a participant in a ⁇ bond (double bond), R′′ is OR, SR, F, Cl, R, or SeR;
  • dashed lines (---) indicate a possible ⁇ or double bond
  • the present invention also further provides the use of a compound of formula (I) or a salt thereof, wherein:
  • A is O, S, CH 2 , CHF, CF 2 or NR;
  • R 1 , R 2 , R 2′ , R 3 , R 3′ , and R 4 are independently selected from the group consisting of H, halogen, OH, N 3 , CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkynoxy, optionally substituted aryloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted arylalkoxy, optionally substituted heterocycloxy, optionally substituted heteroaryloxy, optionally substituted cycloalkoxy, optionally substituted cycloalkenoxy, optionally substituted amino
  • R 4′ is —CY 2 SH, —CY 2 OH, —CY 2 NH 2 , or -L-R 5 ;
  • L′ is selected from the group consisting of —CY 2 —, —CY 2 CY 2 —, —CY 2 OCY 2 —, —CY 2 SCY 2 — and —CY 2 NHCY 2 —;
  • each Y is independently selected from the group consisting of H, F, Cl, Br, OR, C 1 -C 6 alkyl, C 2 -C 6 alkenyl and C 2 -C 6 alkynyl, wherein C 1 -C 6 alkyl, C 2 -C 6 alkenyl, and C 2 -C 6 alkynyl may be optionally substituted with one or more groups selected from F, Cl, Br, OH, COOH, COOCH 3 , SH, SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 , CN, NO 2 , C(O)NH 2 , C(O)NHCH 3 , N 3 , C(S)NH 2 , OCH 3 , and OCH 2 CH 3 ;
  • R 5 is selected from the group consisting of OR, NR 2 , monophosphate, diphosphate, and triphosphate, or a mono, di or triphosphate mimic;
  • each R is independently selected from the group consisting of H, CF 3 , optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
  • Z is independently selected from N or C-G; or, if Z is not a participant in a ⁇ bond (double bond), Z is independently selected from O, S, Se, NR, NOR, NNR 2 , CO, CS, CNR, SO, S(O) 2 , SeO, Se(O) 2 or C(G) 2 , wherein each G is independently selected from the group consisting of H, halogen, OR, SR, NR 2 , NROR, N 3 , COOR, CN, CONR 2 , C(S)NR 2 , C( ⁇ NR)NR 2 , and R; and
  • any two adjacent Z are not both selected from O, S, and Se, or not both selected from CO, CS, CNR, SO, S(O) 2 , SeO, and Se(O) 2 ;
  • X is a participant in a 7 bond (double bond), X is C; or if X is not a participant in a ⁇ bond (double bond), X is CR or N;
  • R′′ is a participant in a ⁇ bond (double bond)
  • R′′ is O, S, Se, NR, NOR or NNR 2 ; or if R′′ is not a participant in a 71 bond (double bond), R′′ is OR, SR, F, Cl, R, or SeR;
  • dashed lines (---) indicate a possible ⁇ or double bond
  • Alkyl refers to monovalent alkyl groups which may be straight chained or branched and preferably have from 1 to 10 carbon atoms or more preferably 1 to 6 carbon atoms. Examples of such alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, n-hexyl, and the like.
  • Aryl refers to an unsaturated aromatic carbocyclic group having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl), preferably having from 6 to 14 carbon atoms.
  • aryl groups include phenyl, naphthyl and the like.
  • Allene refers to a divalent aryl group wherein the aryl group is as described above.
  • Aryloxy refers to the group aryl-O— wherein the aryl group is as described above.
  • Arylalkyl refers to -alkylene-aryl groups preferably having from 1 to 10 carbon atoms in the alkylene moiety and from 6 to 10 carbon atoms in the aryl moiety. Such arylalkyl groups are exemplified by benzyl, phenethyl and the like.
  • Arylalkoxy refers to the group arylalkyl-O— wherein the arylalkyl group are as described above. Such arylalkoxy groups are exemplified by benzyloxy and the like.
  • Alkoxy refers to the group alkyl-O— where the alkyl group is as described above. Examples include, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.
  • Alkenyl refers to a monovalent alkenyl group which may be straight chained or branched and preferably have from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and have at least 1 and preferably from 1-2, carbon to carbon, double bonds. Examples include ethenyl (—CH ⁇ CH 2 ), n-propenyl (—CH 2 CH ⁇ CH 2 ), iso-propenyl (—C(CH 3 ) ⁇ CH 2 ), but-2-enyl (—CH 2 CH ⁇ CHCH 3 ), and the like.
  • Alkenyloxy refers to the group alkenyl-O— wherein the alkenyl group is as described above.
  • Alkynyl refers to alkynyl groups preferably having from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and having at least 1, and preferably from 1-2, carbon to carbon, triple bonds.
  • alkynyl groups include ethynyl (—C ⁇ CH), propargyl (—CH 2 C ⁇ CH), pent-2-ynyl (—CH 2 C ⁇ CCH 2 —CH 3 ), and the like.
  • Alkynyloxy refers to the group alkynyl-O— wherein the alkynyl groups are described above.
  • Acyl refers to groups H—C(O)—, alkyl-C(O)—, cycloalkyl-C(O)—, aryl-C(O)—, heteroaryl-C(O)— and heterocyclyl-C(O)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Oxyacyl refers to groups H—OC(O)—, alkyl-OC(O)—, cycloalkyl-OC(O)—, aryl-OC(O)—, heteroaryl-OC(O)—, and heterocyclyl-OC(O)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Amino refers to the group —NR′′′R′′′ where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is as described herein.
  • Aminoacyl refers to the group —C(O)NR′′′R′′′ where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Acylamino” refers to the group —NR′′′C(O)R′′′ where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • “Acyloxy” refers to the groups —OC(O)—H, —OC(O)-alkyl, —OC(O)-aryl, —C(O)O-heteroaryl, and —C(O)O-heterocyclyl where alkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Aminoacyloxy” refers to the groups —OC(O)NR′′′—H, —OC(O)NR′′′-alkyl, —OC(O)NR′′′-aryl, —OC(O)NR′′′-heteroaryl, and —OC(O)NR′′′-heterocyclyl where R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Oxyacylamino refers to the groups —NR′′′OC(O)OH, —NR′′′OC(O)O-alkyl, —NR′′′OC(O)O-aryl, —NR′′′C(O)O-heteroaryl, and NR′′′OC(O)O-heterocyclyl where R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Oxyacyloxy refers to the groups —OC(O)—OH, —OC(O)O-alkyl, —O—C(O)O-aryl, —OC(O)O-heteroaryl, and —OC(O)O-heterocyclyl where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • Acylimino refers to the groups —C(NR′′′)—R′′′ where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • Acyliminoxy refers to the groups —O—C(NR′′′)—R′′′ where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • Oxyacylimino refers to the groups —C(NR′′′)—OR′′′ where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • Cycloalkyl refers to cyclic alkyl groups having a single cyclic ring or multiple condensed rings, preferably incorporating 3 to 8 carbon atoms.
  • Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl, and the like, or multiple ring structures such as adamantanyl, and the like.
  • Cycloalkenyl refers to cyclic alkenyl groups having a single cyclic ring and at least one point of internal unsaturation, preferably incorporating 4 to 8 carbon atoms.
  • suitable cycloalkenyl groups include, for instance, cyclobut-2-enyl, cyclopent-3-enyl, cyclohex-4-enyl, cyclooct-3-enyl and the like.
  • Halo or “halogen” refers to fluoro, chloro, bromo and iodo.
  • Heteroaryl refers to a monovalent aromatic heterocyclic group which fulfils the Hückel criteria for aromaticity (i.e., contains 4n+2 ⁇ electrons, is planar and conjugated) and preferably has from 2 to 10 carbon atoms and 1 to 4 heteroatoms selected from oxygen, nitrogen, selenium, and sulfur within the ring (and includes oxides of sulfur, selenium and nitrogen).
  • heteroaryl groups can have a single ring (e.g., pyridyl, pyrrolyl or N-oxides thereof or furyl) or multiple condensed rings (e.g., indolizinyl, benzoimidazolyl, coumarinyl, quinolinyl, isoquinolinyl or benzothienyl).
  • a single ring e.g., pyridyl, pyrrolyl or N-oxides thereof or furyl
  • multiple condensed rings e.g., indolizinyl, benzoimidazolyl, coumarinyl, quinolinyl, isoquinolinyl or benzothienyl.
  • Heterocyclyl refers to a monovalent saturated or unsaturated group having a single ring or multiple condensed rings, preferably from 1 to 8 carbon atoms and from 1 to 4 hetero atoms selected from nitrogen, sulfur, oxygen, selenium or phosphorous within the ring. The most preferred heteroatom is nitrogen.
  • heterocyclyl and heteroaryl groups include, but are not limited to, oxazole, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, isothiazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,7
  • Thio refers to groups H—S—, alkyl-S—, cycloalkyl-S—, aryl-S—, heteroaryl-S—, and heterocyclyl-S—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Thioacyl refers to groups H—C(S)—, alkyl-C(S)—, cycloalkyl-C(S)—, aryl-C(S)—, heteroaryl-C(S)—, and heterocyclyl-C(S)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Oxythioacyl refers to groups HO—C(S)—, alkylO—C(S)—, cycloalkylO—C(S)—, arylO—C(S)—, heteroarylO—C(S)—, and heterocyclylO—C(S)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Oxythioacyloxy refers to groups HO—C(S)—O—, alkylO—C(S)—O—, cycloalkylO—C(S)—O—, arylO—C(S)—O—, heteroarylO—C(S)—O—, and heterocyclylO—C(S)—O—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Phosphorylamino refers to the groups —NR′′′—P(O)(R′′′′)(OR′′′′′) where R′′′ represents H, alkyl, cycloalkyl, alkenyl, or aryl, R′′′′ represents OR′′′′′ or is hydroxy or amino and R′′′′′ is alkyl, cycloalkyl, aryl or arylalkyl, where alkyl, amino, alkenyl, aryl, cycloalkyl, and arylalkyl are as described herein.
  • Thioacyloxy refers to groups H—C(S)—O—, alkyl-C(S)—O—, cycloalkyl-C(S)—O—, aryl-C(S)—O—, heteroaryl-C(S)—O—, and heterocyclyl-C(S)—O—, where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • “Sulfinyl” refers to groups H—S(O)—, alkyl-S(O)—, cycloalkyl-S(O)—, aryl-S(O)—, heteroaryl-S(O)—, and heterocyclyl-S(O)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Sulfonyl” refers to groups H—S(O) 2 —, alkyl-S(O) 2 —, cycloalkyl-S(O) 2 —, aryl-S(O) 2 —, heteroaryl-S(O) 2 —, and heterocyclyl-S(O) 2 —, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Sulfinylamino” refers to groups H—S(O)—NR′′′—, alkyl-S(O)—NR′′′—, cycloalkyl-S(O)—NR′′′—, aryl-S(O)—NR′′′—, heteroaryl-S(O)—NR′′′—, and heterocyclyl-S(O)—NR′′′—, where R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Sulfonylamino” refers to groups H—S(O) 2 —NR′′′—, alkyl-S(O) 2 —NR′′′—, cycloalkyl-S(O) 2 —NR′′′—, aryl-S(O) 2 —NR′′′—, heteroaryl-S(O) 2 —NR′′′—, and heterocyclyl-S(O) 2 —NR′′′—, where R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Oxysulfinylamino refers to groups HO—S(O)—NR′′′—, alkylO—S(O)—NR′′′—, cycloalkylO—S(O)—NR′′′—, arylO—S(O)—NR′′′—, heteroarylO—S(O)—NR′′′—, and heterocyclylO—S(O)—NR′′′—, where R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Oxysulfonylamino refers to groups HO—S(O) 2 —NR′′′—, alkylO—S(O) 2 —NR′′′—, cycloalkylO—S(O) 2 —NR′′′—, arylO—S(O) 2 —NR′′′—, heteroarylO—S(O) 2 —NR′′′—, and heterocyclylO—S(O) 2 —NR′′′—, where R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Aminothioacyl refers to groups R′′′R′′′N—C(S)—, where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, where heterocyclic and alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Thioacylamino refers to groups H—C(S)—NR′′′—, alkyl-C(S)—NR′′′—, cycloalkyl-C(S)—NR′′′—, aryl-C(S)—NR′′′—, heteroaryl-C(S)—NR′′′—, and heterocyclyl-C(S)—NR′′′—, where R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and where heterocyclyl and alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Aminosulfinyl refers to groups R′′′R′′′N—S(O)—, where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclic and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • Aminosulfonyl refers to groups R′′′R′′′N—S(O) 2 —, where each R′′′ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclic and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • a group may or may not be further substituted or fused (so as to form a condensed polycyclic group) with one or more groups selected from hydroxyl, acyl, alkyl, alkoxy, alkenyl, alkenyloxy, alkynyl, alkynyloxy, amino, aminoacyl, thio, arylalkyl, arylalkoxy, aryl, aryloxy, acylamino, cyano, halogen, nitro, sulfo, phosphono, phosphorylamino, phosphinyl, heteroaryl, heteroaryloxy, heterocyclyl, heterocycloxy, oxyacyl, acyloxy, oxime, oxime ether, hydrazone, oxyacylamino, aminoacyloxy, trihalomethyl, trialkylsilyl, pentafluoroethyl, trifluoromethoxy, difluorome
  • base refers to the base moiety of a nucleoside or nucleotide.
  • the base moiety is the nitrogen-heterocycle portion of a nucleoside or nucleotide.
  • the base moiety of a nucleotide of formula (I) is a bicyclic heterocycle represented by formula (II) and designated “B”.
  • the nucleoside base is attached to the sugar moiety of a nucleoside in such ways that both ⁇ and ⁇ anomers of D or L nucleosides can be produced. This is denoted by use of the bond which links the base to the sugar moiety.
  • sugar refers to the furanose portion of a nucleoside.
  • the sugar moiety of formula (I) nucleosides, nucleotides and nucleotides mimics and/or prodrugs thereof may contain one or more substituents at their C1-, C2-, C3- and C4-position of the furanose. Substituents may be directed to either the ⁇ - or ⁇ -face of the furanose.
  • the nucleoside base can be considered as a substituent at the C-1 position of the furanose and is preferably directed to the ⁇ -face of the sugar.
  • the ⁇ -face is the side of a furanose on which a purine or pyrimidine base of natural ⁇ -D-nucleosides is present.
  • the ⁇ -face is the side of the sugar opposite to the ⁇ -face.
  • Examples of a “protecting group” for O, S, or N moieties such as hydroxy or NH 2 includes acyl groups, silyl groups, and the like. Suitable protecting groups for these and other moieties are described by T. W., Greene and P. G. M. Wuts; Protecting Groups in Organic Synthesis, 3 rd Ed, John Wiley & Sons, Inc. (1999), incorporated herein by reference.
  • infection refers to the infection caused by an infectious agent or microbe, such as bacteria, parasite (including protozoan), virus or fungus (including unicellular and multicellular).
  • infectious agent or microbe such as bacteria, parasite (including protozoan), virus or fungus (including unicellular and multicellular).
  • microbes that cause such infection include: Acanthamoeba, African Sleeping Sickness (Trypanosomiasis), amebiasis, American Trypanosomiasis (Chagas Disease), Bilharzia (Schistosomiasis), cryptosporidiosis (diarrheal disease, Cryptosporidium Parvum ), Giardiasis (diarrheal disease, Giardia lamblia ), hepatitis A, B, C, D, E, leishmaniasis (skin sores and visceral), malaria ( Plasmodium falciparum ), Salmonella enteritides infection (stomach cramps, diarrhea and fever), tuber
  • Common infections caused by microbes are further outlined in the following chart: Infection Bacteria Fungus Protozoa Virus AIDS X Athlete's Foot X Chicken Pox X Common Cold X Diarrheal Disease X X X Dengue X Flu X Genital Herpes X Malaria X X Meningitis X Pneumonia X X Sinusitis X X Skin Disease X X X X Strep Throat X Tuberculosis X Urinary Tract Infections X Vaginal Infections X X Viral Hepatitis X
  • the compounds of formula (I) may be particularly useful for treating a microbial infection which is a viral infection caused by an RNA virus, such as a virus belonging to group Flaviviridae, for instance Flaviviruses or HCV, or a DNA or retrovirus such as HBV or HIV.
  • a viral infection caused by an RNA virus of the group Flaviviridae and in particular HCV treats a viral infection caused by an RNA virus of the group Flaviviridae and in particular HCV.
  • a therapeutic effective amount is intended to include at least partially attaining the desired effect, or delaying the onset of, or inhibiting the progression of, or halting or reversing altogether the onset or progression of the particular disease of condition being treated.
  • the term “effective amount” relates to an amount of compound which, when administered according to a desired dosing regimen, provides the desired therapeutic activity. Dosing may occur at intervals of minutes, hours, days, weeks, months or years or continuously over any one of these periods. Suitable dosages lie within the range of about 0.1 ng per kg of body weight to 10 g per kg of body weight per dosage.
  • the dosage may be in the range of 1 ⁇ g to 10 g per kg of body weight per dosage, such as is in the range of 1 mg to 10 g per kg of body weight per dosage. In one embodiment, the dosage may be in the range of 1 mg to 500 mg per kg of body weight per dosage.
  • the dosage may be in the range of 1 mg to 250 mg per kg of body weight per dosage. In yet another preferred embodiment, the dosage may be in the range of 1 mg to 100 mg per kg of body weight per dosage, such as up to 50 mg per body weight per dosage.
  • Suitable dosage amounts and dosing regimens can be determined by the attending physician and may depend on the particular condition being treated, the severity of the condition as well as the general age, health and weight of the subject.
  • the active ingredient may be administered in a single dose or a series of doses. While it is possible for the active ingredient to be administered alone, it is preferable to present it as a composition, preferably as a pharmaceutical composition.
  • the formulation of such compositions is well known to those skilled in the art.
  • the composition may contain any suitable carriers, diluents or excipients. These include all conventional solvents, dispersion media, fillers, solid carriers, coatings, further antifungal and antibacterial agents, dermal penetration agents, surfactants, isotonic and absorption agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
  • compositions include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parental (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • compositions suitable for oral administration may be presented as discrete units such as capsules, sachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a tablet may be made by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g., inert diluent, preservative disintegrant (e.g., sodium starch glycolate, cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethyl cellulose) surface-active or dispersing agent.
  • a binder e.g., inert diluent, preservative disintegrant (e.g., sodium starch glycolate, cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethyl cellulose) surface-active or dispersing agent.
  • Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • compositions suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured base, usually sucrose and acacia or tragacanth gum; pastilles comprising the active ingredient in an inert basis such as gelatine and glycerin, or sucrose and acacia gum; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • compositions suitable for topical administration to the skin may comprise the compounds dissolved or suspended in any suitable carrier or base and may be in the form of lotions, gel, creams, pastes, ointments and the like.
  • suitable carriers include mineral oil, propylene glycol, polyoxyethylene, polyoxypropylene, emulsifying wax, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • Transdermal patches may also be used to administer the compounds of the invention.
  • compositions for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter, glycerin, gelatine or polyethylene glycol.
  • compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • compositions suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bactericides and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • formulations of these compositions in dry powder form for delivery by a dry powder inhaler offer yet another means of administration. This overcomes many of the disadvantages of the oral and intravenous routes.
  • Preferred unit dosage compositions are those containing a daily dose or unit, daily sub-dose, as herein above described, or an appropriate fraction thereof, of the active ingredient.
  • compositions may include other agents conventional in the art having regard to the type of composition in question, for example, those suitable for oral administration may include such further agents as binders, sweeteners, thickeners, flavouring agents disintegrating agents, coating agents, preservatives, lubricants and/or time delay agents.
  • suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine.
  • Suitable disintegrating agents include cornstarch, methylcellulose, polyvinylpyrrolidone, xanthan gum, bentonite, alginic acid or agar.
  • Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring.
  • Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten.
  • Suitable preservatives include sodium benzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite.
  • Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc.
  • Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
  • the present invention provides novel compounds, pharmaceutical compositions of said novel compounds and therapeutic applications of said novel compounds for the treatment of microbial infections.
  • the present invention provides a compound of the formula (I) or a salt thereof; wherein:
  • A is O, S, CH 2 , CHF, CF 2 or NR;
  • R 1 , R 2 , R 2′ , R 3 , R 3′ , and R 4 are independently selected from the group consisting of H, halogen, OH, N 3 , CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkynoxy, optionally substituted aryloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted arylalkoxy, optionally substituted heterocycloxy, optionally substituted heteroaryloxy, optionally substituted cycloalkoxy, optionally substituted cycloalkenoxy, optionally substituted amino
  • R 4′ is —CY 2 SH, —CY 2 OH, —CY 2 NH 2 , or -L′-R 5 ;
  • L′ is selected from the group consisting of —CY 2 —, —CY 2 CY 2 —, —CY 2 OCY 2 —, —CY 2 SCY 2 — and —CY 2 NHCY 2 —;
  • each Y is independently selected from the group consisting of H, F, Cl, Br, OR, C 1 -C 6 alkyl, C 2 -C 6 alkenyl and C 2 -C 6 alkynyl, wherein C 1 -C 6 alkyl, C 2 -C 6 alkenyl, and C 2 -C 6 alkynyl may be optionally substituted with one or more groups selected from F, Cl, Br, OH, COOH, COOCH 3 , SH, SCH 3 , NH 2 , NHCH 3 , N(CH 3 ) 2 , CN, NO 2 , C(O)NH 2 , C(O)NHCH 3 , N 3 , C(S)NH 2 , OCH 3 , and OCH 2 CH 3 ;
  • R 5 is selected from the group consisting of OR, NR 2 , monophosphate, diphosphate, and triphosphate, or a mono, di or triphosphate mimic;
  • each R is independently selected from the group consisting of H, CF 3 , optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
  • Z is independently selected from N or C-G; or, if Z is not a participant in a ⁇ bond (double bond), Z is independently selected from O, S, Se, NR, NOR, NNR 2 , CO, CS, CNR, SO, S(O) 2 , SeO, Se(O) 2 or C(G) 2 , wherein each G is independently selected from the group consisting of H, halogen, OR, SR, NR 2 , NROR, N 3 , COOR, CN, CONR 2 , C(S)NR 2 , C( ⁇ NR)NR 2 , NCONR 2 , NCSNR 2 and R; and
  • any two adjacent Z are not both selected from O, S, and Se, or not both selected from CO, CS, CNR, SO, S(O) 2 , SeO, and Se(O) 2 ;
  • X is a participant in a ⁇ bond (double bond), X is C; or if X is not a participant in a ⁇ bond (double bond), X is CR or N;
  • R′′ is a participant in a ⁇ bond (double bond)
  • R′′ is O, S, Se, NR, NOR, and NNR 2 ; or if R′′ is not a participant in a 71 bond (double bond), R′′ is OR, SR, F, Cl, R, or SeR;
  • R 2′ , R 3′ , and R 5 are OH or OC(O)CH 3
  • L′ is CH 2
  • A is O
  • R 1 , R 2 , R 3 and R 4 are H
  • B is not the group of formula (III), formula (IV), formula (V), formula (VI), or formula (VII)
  • R 2 and R 2′ are not both H
  • R 2′ is F
  • R 3′ is OH
  • R 5 is triphosphate
  • L′ is CH 2
  • A is O
  • R 1 , R 2 , R 3 and R 4 are H
  • B is not a group of formula (X)
  • the bicyclic base structure B of formula II may have one or more ring double bonds and, in some instances, may have two or more ring double bonds.
  • the base structure has at least two double bonds and more preferably three or more double bonds.
  • the base structure B is selected form the following formulae (XI) to (XXI) wherein Z, X and R′′ are as defined above.
  • X is N.
  • the structure of formula II is represented by the structure of formula IIa wherein each Z′ is independently N (if a participant in a ⁇ bond) or NR (if not a participant in a ⁇ bond), and R′′, R and Z are as defined above.
  • the structure of formula II is represented by the structure of formula IIb wherein R′′ and Z′ are as defined above and each Z is independently CG (if a participant is ⁇ bond) or >C(G) 2 (if not a participant in a ⁇ bond).
  • formula II is represented by the structures of formulae IIc, IId and IIe wherein R and G are as defined above.
  • each R is independently selected from H, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl and each G is independently selected from H, halogen, CF 3 , optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl.
  • L′ is —CH 2 —.
  • R 4′ is selected from —CH 2 —OH, —CF 2 OH, —CCl 2 —OH, —C(CH 3 )(CH) 3 OH, —CH(CH 3 )OH, —CH 2 —CH 2 —P(O)(OH) 2 , —CH 2 —CH 2 —P(O)(OH) 2 , —CH 2 SP(O)(OH) 2 , —CH 2 SH, —CF 2 SH, and —CH 2 —O—P(O)(OPh)(NHCH(CH 3 )(CO 2 Me)).
  • R 4′ is —CH 2 —OH.
  • R 1 , R 2 , R 2′ , R 3 , R 3′ , and R 4 are independently selected from the group consisting of H, F, Cl, Br, I, OH, SH, NH 2 , NHOH, NHNH 2 , N 3 , COOH, CN, CONH 2 , C(S)NH 2 , COOR, R, OR, SR, SSR, NHR, and NR 2 wherein at least one of R 2 or R 2′ is a substituent other than H, and wherein R is selected from the group consisting of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, and optionally substituted arylalkyl.
  • the sugar moiety is selected from the following formulae: or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • At least one of R 2 and R 2′ is methyl, hydroxyl or F.
  • the sugar moiety is selected from the following formulae: or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • the compound of formula (I) is selected from the following formulae, or salts thereof: wherein:
  • each R on the sugar moiety is independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl and optionally substituted arylalkyl;
  • each R on the base moiety is independently selected from H, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
  • each G is independently selected from H, halogen, CF 3 , optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl; and
  • nucleosides incorporating a variety of sugar moieties have been found to be useful for the inhibition of viral polymerases, in the case of the Flaviviridae, and in particular, the Hepatitis C virus, 2′-C-methyl ribnucleosides have been found to be particularly useful (see Eldrup, A. B. et al., J. Med. Chem. 2004, 47(21), 5284-97, which is incorporated herein by reference).
  • the sugar moiety is represented by the following formula: or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • the sugar moiety is represented by the following formula: or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • the bicyclic nucleosides of the present invention also include derivatives such as nucleotides, and nucleotide mimics and/or prodrugs thereof.
  • nucleotide mimics of the compounds of the present invention of formula (I) discussed above include a compound in which R 5 is a monophosphate or monophosphate mimic of formula (XXII) or (XXIII): where X 1′ X 4′ , and X 6′ , independently are O, S, NR; X 2′ , X 3′ , and X 5′ are selected independently from the group consisting of H, F, NROR, N 3 , CN, (BH 2 G) ⁇ M + , (BH 3 ) ⁇ M + , R, OR, SR, and NR 2 .
  • the substituents (BH 2 G) ⁇ M + and (BH 3 ) ⁇ M + are ion pairs, which are linked to phosphorus through the negatively charged boron.
  • M + is a cation, preferably a pharmaceutically acceptable cation such as Ca 2+ , ammonium, trialkylammonium or tertaalkylammonium, e.g., NH 4 + , Et 3 NH + , Bu 3 NH + , and Bu 4 N + .
  • nucleotide mimics of the compounds of formula (I) as discussed above include di- and triphosphates and di- and tri-phosphate mimics including a compound in which R 5 is a di- or tri-phosphate moiety of formula (XXIV):
  • X 2 , X 3 , and X 4 are selected independently from the group consisting of O, S, Se, and NR;
  • X 5 and X 6 are selected independently from the group consisting of —O—, —S—, —Se—, —CY 2 C(O)—, —CH(OH)—, —C(OH) 2 —, —CH 2 O—, —CH 2 CH 2 —, —CH 2 CH(NH 2 )—, —CH 2 CH 2 CH(NH 2 )—, —CY 2 OCY 2 —, —CY 2 —, —CRY—, —CY 2 CY 2 —, —CHR—, —C ⁇ C—, —HC ⁇ CH—, —NH—, —NR—, >NOH, >NOR, >NNH 2 , and >NNHR;
  • X 7 , X 8 , X 9 , and X 10 are selected independently from the group consisting of H, F, OR, SR, NR 2 , NROR, NRNR 2 , CN, N 3 , (BH 3 ) ⁇ M + , (BH 2 G) ⁇ M + , R and SeR;
  • n 0 or 1.
  • nucleotide phosphate mimics and methods of making the phosphate mimics appropriate for the compounds of the present invention are described, inter alia, in WO 2003/072757 and WO 2003/073989, the entire contents of which are incorporated herein by reference. Many of the nucleotide mimics discussed herein can be prepared by similar approaches as published or by using well-known knowledge of organophosphorous chemistry. Generally, phosphate mimics of the nucleosides and nucleotides of the present invention can inhibit enzyme function without phosphorylation and/or have enhanced nuclease stability relative to nucleotides with unmodified phosphate.
  • phosphate mimic refers to a phosphate analog, including, but not limited to, phosphonate, phosphothiolate, phosphoselenoate, selenophosphate, thiophosphate, P-boranophosphate, phosphoramidate, sulfamate, sulfonate, and sulfonamide and/or a combination thereof.
  • Preferred embodiments of the phosphate mimics include phosphonate, phosphoramidate, phosphorothioate, methylphosphonate, fluoromethylphosphonate, difluoromethylphosphonate, vinylphosphonate, phenylphosphonate, sulfonate, fluorophosphate, dithiophosphorothioate, 5′-methylenephosphonate, 5′-difluoromethylenephosphonate, 5′-deoxyphosphonate, 5′-aminophosphoramidate, and 5′-thiophosphate. More preferred is phosphonate and phosphoramidate.
  • diphosphate mimic and triphosphate mimic specifically refer to a diphosphate analog and a triphosphate analog, respectively, which comprises at least one of the phosphate mimics, one of the modifications at the bridging site of diphosphate and triphosphate (e.g., X 5 , X 6 and X 10 ), and/or replacements of non-bridging phosphate oxygens (e.g., X 4 , X 3 and X 2 ).
  • the ⁇ -P, ⁇ -P, and ⁇ -P in the mono, di- and triphosphate mimics may independently adopt either R or S configurations when chiral.
  • the invention encompasses enantiomers, or stereoisomers and mixtures thereof, such as enantiomerically enriched mixtures.
  • the base moieties of the present invention may exist as rapidly interconvertible mixtures of isomers. Isomerism of this kind is known in the art as tautomerism. Individual isomers are called tautomers. Where tautomerism is possible the present invention covers all possible tautomers of the compounds of formula (I).
  • the compounds of the present invention can be administered to a subject as a pharmaceutically acceptable salt thereof. It will be appreciated however that non-pharmaceutically acceptable salts also fall within the scope of the present invention since these may be useful as intermediates in the preparation of pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts include, but are not limited to salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, maleic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, toluenesulphonic, benezenesulphonic, salicyclic sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.
  • pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric
  • Base salts include, but are not limited to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylammonium.
  • the present invention includes within its scope cationic salts eg sodium or potassium salts, or alkyl esters (eg methyl, ethyl) of the phosphate group.
  • Basic nitrogen-containing groups may be quarternised with such agents as lower alkyl halide, such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl and diethyl sulfate; and others.
  • lower alkyl halide such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl and diethyl sulfate; and others.
  • the bicyclic nucleosides and nucleotides of the present invention also include their prodrug derivatives.
  • prodrug is used in its broadest sense and encompasses those derivatives that are converted in vivo to the compounds of the invention.
  • Prodrugs of the compounds of the present invention may be prepared by modification of the sugar moiety or of the phosphate or phosphate mimic to include a prodrug substituent. Such prodrug modification is generally performed to enhance drug absorption and/or drug delivery into cells.
  • Prodrugs substituents include, but are not limited to residues of: proteins; antibiotics; D- and L-amino acids which may be attached to a phosphate moiety or a phosphate mimic moiety via a carbon atom (phosphonates), a nitrogen atom (phosphoamidates), or an oxygen atom (phosphoesters) or may be attached to the sugar moiety through any one or more of the R 1 -R 5 groups; peptides (preferably up to 10 amino acids) attached to a phosphate moiety or a phosphate mimic moiety via a carbon atom (phosphonates), a nitrogen atom (phosphoamidates), or an oxygen atom (phosphoesters), or may be attached to the sugar moiety through any one or more of the R 1 -R 5 groups; drug moieties attached to a phosphate moiety or a phosphate mimic moiety via a carbon atom (phosphonates), a nitrogen atom (phosphoamidates), or an oxygen atom (phosphoesters), or may be attached to the
  • prodrug derivatives of nucleosides, nucleotides and nucleotide phosphate mimics and methods of making the prodrugs appropriate for use in the present invention are described, inter alia, in PCT Publications WO 2003/072757 and WO 2003/073989.
  • the prodrug of a nucleoside 5′-monophosphate mimic can mask the negative charges of the phosphate mimic moiety entirely or partially, or mask the negative charges of the di-phosphate mimic or tri-phosphate mimic moiety or phosphate moiety in order to enhance drug absorption and/or drug delivery into cells.
  • a combination of prodrug substituents may be attached to one or more X 2′ , X 3′ and X 5′ positions on a nucleoside mono-phosphate mimic or to one or more X 7 -X 10 positions on a nucleoside di- or tri-phosphate mimic.
  • Preferred prodrug substituents in positions X 2′ , X 3′ or X 5′ position include 2,3-O-diacylglyceryloxy, 2,3-O-dialkylglyceryloxy, 1-O-alkyl-2-O-acylglyceryloxy, 1-O-acyl-2-O-alkylglyceryloxy, 1-S-alkyl-2-O-acyl-1-thioglyceryloxy, acyloxymethoxy, S-acyl-2-thioethoxy, S-pivaloyl-2-thioethoxy, acyloxymethoxy, pivaloyloxymethoxy, alkoxycarbonyloxymethoxy, S-alkyldithio-S′-ethyoxy acyloxymethoxy, S-acyl-2-thioethoxy, S-pivaloyl-2-thioethoxy, pivaloyloxymethoxy, alkoxycarbonyloxymethoxy, S
  • the prodrug substituent is a substituent on a hydroxyl group of the sugar moiety (that is, for instance, any one of R 1 -R 5 ).
  • the modification results in the formation of an ester and in this regard the preferred prodrug substituents are C 1 -C 6 acyl groups for example, acetyl, propionyl, pivaloyl, etc.
  • substituted C 1 -C 6 acyl moieties for example, fluoroacetyl, difluoroacetyl, etc.
  • the substituted C 1 -C 6 acyl group is represented as a residue of an L or D amino acid consisting of alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, aspartic acid, glutamic acid, arginine, histidine, and lysine.
  • the prodrug substituent is an amino acid residue of D or L-valine.
  • the prodrug can be activated either by cellular enzymes such as lipases, esterases, reductases, oxidases, nucleases or by chemical cleavage such as hydrolysis to release (liberate) either the nucleoside, nucleotide or nucleotide mimic after the prodrug enters cells.
  • cellular enzymes such as lipases, esterases, reductases, oxidases, nucleases or by chemical cleavage such as hydrolysis to release (liberate) either the nucleoside, nucleotide or nucleotide mimic after the prodrug enters cells.
  • the delivery of the nucleosides and nucleotides may be assisted by using a therapeutically acceptable carrier such as liposomal suspensions, cationic lipids, and polyimines.
  • a therapeutically acceptable carrier such as liposomal suspensions, cationic lipids, and polyimines.
  • novel nucleosides, nucleotides, nucleotide mimics and prodrugs thereof, of the present invention can be prepared by those who are skilful in synthetic organic and nucleoside chemistry using established synthetic methodology ( Chemistry of Nucleosides and Nucleotides Vol. 1 (1988), Vol. 2 (1991), Vol. 3 (1994), edited by L. B. Townsend, Plenum Press; Handbook of Nucleoside Synthesis by H. Vorbrüggen and C. Ruh-Pohlenz, John Wiley & Sons, Inc., 2001 ; The Organic Chemistry of Nucleic Acids by Y. Mizuno, Elsevier, 1986).
  • the nucleosides of the present invention can be converted to their corresponding monophosphate, diphosphate, and triphosphate nucleosides by established phosphorylation procedures.
  • known methods in the art can be used to synthesise the nucleotide and phosphate mimics and prodrugs.
  • the following schemes and description serve as representative syntheses of the nucleosides and nucleotides of the present invention.
  • other compounds such as those having —CY 2 SH, —CY 2 OH or -L′-R 5 groups other than CH 2 R 5 may similarly be made.
  • the bicyclic nucleosides of the present invention may be prepared by modification of optionally protected and functionalised cytosine, uracil and other base analogues followed by Stille, Heck, Sonogashira or other metal-mediated cross coupling chemistry to introduce an ⁇ , ⁇ -unsaturated ester, alkyne or other functional group.
  • Such processes allow for stereoselective synthesis of an intermediate capable of efficient cyclisation to form the bicyclic compounds of the present invention. Cyclisation and optional deprotection of the product delivers the target bicyclic nucleoside.
  • any compound capable of metal-mediated cross coupling may be used, such as a tin derivative like trialkyltin. More preferably tributyltin.
  • the reactions are carried out using a palladium based coupling agent.
  • Suitable coupling agents are known in the art and include Pd(PPh 3 ) 2 Cl 2 , Pd(PPh 3 ) 4 , Pd(dibenzylideneacetone), and PdCl 2 (CH 3 CN) 2 .
  • the palladium catalysed coupling reactions may also include a co-catalyst, for instance, CuI which may be in the presence of a suitable non-nucleophilic base such as a trialkylamine.
  • Coupling reactions are generally performed at temperatures around room temperature. Elevated temperatures such as temperatures between 30-80° C. can be employed to effect coupling and cyclisation in a single step. It is also preferred that such reactions are carried out under an inert atmosphere of either nitrogen or argon.
  • Suitable solvents include ether solvents such as THF and diethylether or polar solvents such as DMF.
  • Schemes 1 and 2 illustrates some preferred cyclisation methods for forming the 6-membered ring portions of the bicyclic bases (B) of the compounds of the present invention.
  • Methodologies other than metal-mediated cross-coupling and cyclisation can also be used to prepare the bicyclic nucleosides.
  • the final process in scheme 2 depicts how a bifunctional two atom unit (such as an ⁇ -halocarboxylic ester or glyoxal) can be used to form the second 6-membered ring portion of a suitable nucleoside intermediate.
  • each of Z 1 -Z 4 is independently Z.
  • A is O, CH 2 or optionally protected N;
  • Y is halogen or other appropriate group such as trifluoromethanesulfonate;
  • W 4 is H or trialkyltin;
  • Z 1 and Z 2 are each independently C, CH, C-halogen, C-alkyl, C-aryl, C-heteroaryl, C—O-alkyl or C—S-alkyl;
  • Z 3 is CH, C-alkyl, C-halogen, N, CNHR, CNH 2 , CNR 2 , C ⁇ O, or C ⁇ S;
  • Z 4 is CH, C-halogen, C-alkyl, C-aryl, C-heteroaryl, C—O-alkyl, C—S-alkyl, C—OH, C—NH 2 , C—NHR, C—NR 2 or C ⁇ O;
  • R 1 , R 2 , R 2′ R 3 , R 3′ , R 4′ are each independently H, halogen, al
  • the bicyclic bases of the present invention can be further modified either prior to being added to the sugar moiety or once attached. Such modification may involve transformations through functionalisation, defunctionalisation, or functional group interconversion. Modifications may include esterification, the preparation of enol ethers, o-alkylation, bromination, hydrogenation, dihydroxylation, epoxidation, oximation, and amination.
  • the compounds described herein can also be converted into their corresponding mono-, di- and triphosphates using well established methods.
  • prodrugs of mono-, di- and triphosphates can be prepared in order to optimise the biological efficacy of these phosphorylated compounds. Methods for preparing such prodrugs are well known in the art (see Wagner, C. R., et al. Med. Res. Rev., 2000, 20, 417-451).
  • A is O, CH 2 or optionally protected N;
  • R 1 , R 2 , R 2′ , R 3 , R 3′ , R 4 are each independently H, halogen, alkyl, O-alkyl, OH, optionally protected O, or methyl and B is as described herein.
  • phosphate mimics As discussed earlier an alternative to the use of phosphates is the use of phosphate mimics and their prodrugs.
  • One such phosphate mimic is shown below and this can be prepared using appropriately protected nucleosides and known conditions.
  • A is O, CH 2 or optionally protected N;
  • R 1 , R 2 , R 2′ , R 3 , R 3′ , R 4 are each independently H, halogen, alkyl, O-alkyl, OH, optionally protected O, methyl or F;
  • X′ is O, S, NH, CF 2 , CHF, CClH, CBr 2 or CHBr;
  • B is as described herein.
  • the bicyclic compounds of the present invention may be tested for biological activity using well known procedures.
  • Antiviral assays are conducted according to published, widely used protocols. In order to obtain the therapeutic index, compound-induced cytotoxicity to host cells is also measured in parallel with antiviral activities. To determine the mode of action of antiviral nucleosides the corresponding nucleoside triphosphates are subject to enzyme-based assays for the inhibition of viral polymerases according to known protocols (Ranjith-Kumar et al. J. Virol. 2001, 75, 8615; Dhanak et al. J. Biol. Chem. 2002, 277, 38322-38327). Some compounds of the present invention showed K i values of less than 1 ⁇ M against HCV NS5B.
  • HCV replicon-containing cell lines (Randall and Rice, Current Opinion in Infectious Diseases 2001, 14, 743) are used for the identification of potential anti-HCV compounds. Among them is a widely used subgenomic replicon system developed by Lohmann et al. ( Science 1999, 285, 110 ; J. General Virol. 2000, 81, 1631 ; J. Virol. 2001, 75, 1437, 2002, 76, 4008). Some compounds of the present invention showed potent anti-HCV activity with EC 50 values of low ⁇ M.
  • Anti-HIV assays can be conducted according to the protocols developed by Schinazi et al. ( Antimiromobial Agents Chemother. 1990, 34, 1061; 1992, 36, 2423; 1993, 37, 875) or other widely used protocols (Kimpton et al J. Virol. 1992, 66, 2232; Chan et al. J. Med. Chem. 2001, 44, 1866).
  • Preferred nucleoside triphosphates of the present invention may act as potent inhibitors of the non-structural position SB (NS5B) which is HCV's RNA-dependent RNA polymerase. Accordingly, such compounds are preferably suited to treat and/or prevent HCV. Also, as the preferred novel compounds of the present invention are expected to exhibit novel profiles of activity they may provide the artisan with an alternative to treating viruses which display drug resistance to conventional drugs. Other advantages which may be exhibited by the preferred novel compounds of the present invention include:
  • nucleosides, nucleotide, nucleotide mimics and/or their prodrugs of the present invention may be useful for the inhibition of a variety of enzymes including, but not limited to, DNA or RNA polymerases, helicases, ribonucleotide reductases, protein kinases, and telomerases and for the modulation of G-proteins, P2 purinergic receptors and the allosteric sites of a variety of enzymes.
  • the novel nucleosides, nucleotides, nucleotide mimics and/or prodrugs of the present invention are used to treat viral infections caused by the RNV viruses of the group Flaviviridae and, in particular, HCV.
  • novel nucleosides, nucleotides, nucleotide mimics and/or their prodrugs of the present invention are useful for the treatment of infectious diseases caused by infectious agents such as parasites, bacteria and fungi.
  • novel nucleosides, nucleotide mimics and/or their prodrugs that display potent cytotoxicities to fast-dividing cancerous cells may be useful for the treatment of proliferative disorders, including, but not limited to, lung cancer, liver cancer, prostate cancer, colon cancer, breast cancer, ovarian cancer, melanoma, and leukemia.
  • novel nucleosides, nucleotides, nucleotide mimics and/or their prodrugs of the present invention may also be useful for the treatment of a wide range of other diseases and disorders such as inflammatory diseases, autoimmune diseases, Type 2 diabetes, and cardiovascular diseases.
  • nucleosides, nucleotides, nucleotide mimics and/or their prodrugs of the present invention may be therapeutically administered as a single drug, or alternatively may be administered in combination with one or more other active chemical entities to form a combination therapy.
  • the other active chemical entities may be a small molecule, a polypeptide, or a polynucleotide.
  • compounds of this invention may be particularly useful when used in combination with other agents known to exert an antiviral effect.
  • combination with immunomodulatory/antiviral agents such as interferons, interferon derivatives and other large or small molecules known to modulate host immune responses may be beneficial.
  • combinations of compounds of this invention with IMPDH inhibitors e.g., ribavirin
  • antiviral nucleosides e.g., antiviral non-nucleosides
  • antiviral non-nucleosides e.g., polymerase inhibitors, protease inhibitors
  • N,O-Bis(trimethylsilyl)acetamide (7.17 mL) was added to 5-iodocytidine (2.32 g) in dry acetonitrile (15 mL) and the mixture was heated under argon at 80° C. for 30 mins.
  • SnCl 4 (1.71 mL) was added cautiously and heating continued for 2 hrs.
  • Example 5 (10.5 mg) was prepared directly from commercially available 5-Iodo-2′-O-methylcytidine using the method described for intermediate B in scheme 1.
  • N,O-Bis(trimethylsilyl)acetamide (6.45 mL) and uracil (0.986 g) in dry acetonitrile (20 mL) were heated under argon at 80° C. for 30 mins.
  • a suspension of commercially available 1,2,3,5-tetra-O-benzoyl-2-C-methyl-alpha/beta-D-ribofuranose in dry acetonitrile (80 mL) was added and heating continued for 1 hr.
  • SnCl 4 was added cautiously and heating continued for 5 hrs.
  • the reaction was cooled to ambient temperature, poured into an ice cold solution of saturated sodium bicarbonate (100 mL) and extracted with EtOAc ( ⁇ 3).
  • 1,2,4-triazole (207 mg) was dissolved in dry acetonitrile (5 mL) under argon, cooled to 0° C. and treated with POCl 3 (75 ⁇ L). After 5 mins, triethylamine (0.60 mL) was added and stirring continued for 1 h at ambient temperature before addition of Intermediate F in dry acetonitrile (5 mL). The reaction was diluted with EtOAc after 90 mins and washed successively with saturated NaHCO 3 and brine (each ⁇ 3). The organic layer was dried (MgSO 4 ) and evaporated in-vacuo to provide the crude triazole intermediate as a yellow gum (127 mg).
  • the crude triazole intermediate (621 mg) was prepared using the method described for intermediate G. This gum was suspended in dry ethanol (10 mL) before addition of benzylamine (164 ⁇ l). After stirring for 1 hr at ambient temperature the solvent was removed in-vacuo and the residue purified by radial chromatography on silica eluting with 3%-5% MeOH/DCM to give the required product as an oil (424 mg). MS m/z ([M+H] + ) 660.4, 661.4.
  • Example 6 (20 mg) was dissolved in methanol (3 mL) and NH 4 Cl (10 mg) in water (0.2 mL) was added followed by NaBH 4 (10 mg). After 10 mins the reaction was evaporated to dryness in-vacuo and the residue dissolved in water (3 mL) and washed with DCM. The aqueous extract was then purified by preparative HPLC to afford the product as an oil (4.23 mg).
  • Example 7 (20 mg) yielded Example 10 (7.0 mg).
  • Example 11 The general method for Example 11 was applied to the preparation of Examples 12-14.
  • Example 1 (31 mg) was dried overnight under high vacuum over P 2 O 5 and then stirred with trimethyl phosphate (1.7 mL) at ambient temperature with oven-dried molecular sieves (4 ⁇ ) for 16 hrs under argon. The reaction was cooled to 0° C., POCl 3 (31 ⁇ l) added and stirring continued for 2 hours before addition of Bu 3 N (72 ⁇ l) followed by acetonitrile (0.5 mL) and tributylammonium pyrophosphate (190 mg). After a further 2 hrs at 0° C.
  • reaction was quenched by pouring into ice-cold 1M triethylammonium bicarbonate buffer (10 mL, pH 8.5) and the aqueous layer washed with Et 2 O (3 ⁇ 10 mL). The aqueous material was then lyophilised to give a white solid which was purified by preparative HPLC to afford the product (1.67 mg).
  • test compounds were assayed in the stable HCV RNA-replicating cell line, AVA5, derived by transfection of the human hepatoblastoma cell line, Huh7 (Blight, et al., 2000, Sci. 290:1972). Concentrations of compounds were added to dividing cultures once daily for three days and intracellular HCV RNA levels and cytotoxicity assessed 24 hours after the last dose of compound.
  • Intracellular HCV RNA levels were measured using standard blot hybridization techniques using triplicate cultures and levels of ⁇ -actin RNA were used to normalize HCV RNA levels in each sample. Cytotoxicity was measured using an established neutral red dye uptake assay (B. E. Korba and J. L. Gerin. 1992 . Antivir. Res. 19, 55-70) and the 50% effective antiviral concentrations (EC 50 ) and cytotoxic concentrations (CC 50 ) were calculated using a computer program for curve fitting.
  • Examples such as 1-14 were typically active in the replicon assay in the range 1 to >1000 ⁇ M and cytotoxic in the range 30 to >100 ⁇ M.
  • RNA template was synthesized, gel purified and quantified by spectrophotometry.
  • the kinetic constant, K m was determined for the RNA template and for each GTP, CTP, ATP and UTP using a non-linear least square fit of initial rates as a function of substrate concentration assuming Michaelis-Menten kinetics.
  • Standard RdRp assays consisted of 30 nM RNA template and 25 nM HCV NS5b ⁇ 21 (genotype 1b) (Replizyme Ltd) in a 50 L reaction mixture containing 20 mM Tris-HCl, 5 mM MgCl 2 , 3 mM DTT, 0.05% BSA, 22 nM GTP, 600 nM ATP, 15 nM CTP, 30 nM UTP and 3 nM [ ⁇ - 33 P]GTP. Elongation reactions were initiated by the addition of NTPs and proceeded for 30 mins at 25° C. Reactions were quenched by the addition of 0.2 M EDTA and product formation was collected by filtration through Multiscreen plates (Millipore). Quantification of product formation was performed using TopCount (Perkin Elmer).
  • the inhibitor concentration at which the enzyme catalyzed rate is reduced by half (IC 50 ) was determined using a computer program for curve fitting
  • Examples such as 15 were typically inhibitory of NS5b in the range 100 to >1000 ⁇ M.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Engineering & Computer Science (AREA)
  • Communicable Diseases (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)

Abstract

The invention relates to the use of bicyclic nucleosides and nucleotides based on formula (II) for the treatment of infectious diseases, and in particular, viral infections.

Description

    FIELD OF THE INVENTION
  • The present invention relates to the use of bicyclic nucleosides and nucleotides for the treatment of infectious diseases, including viral infections, novel bicyclic nucleosides and nucleotides and methods for their manufacture.
  • BACKGROUND OF THE INVENTION
  • Viral infections are a major threat to human health and account for many serious infectious diseases. The most notable viruses are the blood-borne viruses (BBV), which include hepatitis C virus (HCV), hepatitis B virus (HBV) and human immunodeficiency virus (HIV) which are all linked by their mode of transmission, i.e., through blood or bodily fluids.
  • The Flaviviridae is a group of positive single-stranded RNA viruses with a genome size from 9-15 kb. The Flaviviridae consists of various genera including:
  • 1. Flaviviruses: This genus includes the Dengue virus, Japanese Tick-Borne and the Yellow Fever virus. Apart from these major groups, there are some additional Flaviviruses that are unclassified.
  • 2. Hepaciviruses: This genus contains only one species, the Hepatitis C virus (HCV), which is composed of many genotypes and subtypes.
  • HCV is a major cause of viral hepatitis and has infected more than 200 million people worldwide. Current treatment for HCV infection is restricted to immunotherapy with interferon-α alone or in combination with ribavirin, a nucleoside analog. This treatment is effective in only about half the patient population. Therefore, there is an urgent need for new HCV drugs. Hepatitis C virus comprises a positive-strand RNA genome enclosed in a nucleocapsid and lipid envelope and consists of approximately 9600 ribonucleotides, which encodes a polyprotein of about 3000 amino acids (Dymock et al. Antiviral Chemistry & Chemotherapy 2000, 11, 79). A HCV protein, NS5B, released from the polyprotein, possesses polymerase activity and is involved in the synthesis of double-stranded RNA from the single-stranded viral RNA genome that serves as the template. The reproduction of HCV virus may be prevented through the manipulation of NS5B's polymerase activity. The inhibition of NS5B protein would suppress or prevent the formation of the double-stranded HCV RNA. Alternatively, a nucleoside analog also may be incorporated into the extending RNA strand and act as a chain-terminator. Furthermore, a deteriorating nucleoside analog also may be incorporated into the extending RNA, which may cause mutagenic damage to the viral genome. Recently, several PCT patent applications (WO 99/43691, WO 01/32153, WO 01/60315, WO 01/79246, WO 01/90121, WO 01/92282, WO 02/18404, WO 02/057287, WO 02/057425) have described nucleoside analogs as anti-HCV agents in in vitro assays.
  • HBV has acutely infected almost a third of the world's human population, and about 5% of the infected are chronic carriers of the virus (Delaney I V et al., Antiviral Chemistry & Chemotherapy 2001, 12, 1-35). Chronic HBV infection causes liver damage that frequently progresses to cirrhosis and/or liver cancer later in the life. Despite the availability and widespread use of effective vaccines and chemotherapy, the number of chronic carriers approaches 400 million worldwide. Therefore, more effective anti-HBV drugs need to be developed.
  • HIV causes progressive degeneration of the immune system, leading to the development of AIDS. A number of drugs have been used clinically, including reverse transcriptase inhibitors and protease inhibitors. Currently, combination therapies are used widely for the treatment of AIDS in order to reduce the drug resistance. Despite the progress in the development of anti-HIV drugs, AIDS is still one of the leading epidemic diseases.
  • Apart from the BBV's discussed above certain other acute viral infections also impose a great threat to human life, including the HSV, CMV, influenza viruses, West Nile virus, SARS virus, small pox, EBV, VZV and RSV. Accordingly, this highlights the continued need for the development of different antiviral drugs.
  • Bacterial infections have long been the sources of many infectious diseases. The widespread use of antibiotics has produced many new strains of life-threatening antibiotic resistant bacteria. Fungal infections are another type of infectious diseases, some of which also can be life-threatening. There is an ever increasing demand for the treatment of bacterial and fungal infections. As such, antimicrobial drugs based on new mechanisms of action are especially important.
  • Nucleoside drugs have been used clinically for decades for the treatment of viral infections and proliferative disorders such as cancer. Most of the nucleoside drugs are classified as antimetabolites. After they enter cells, nucleoside analogs are phosphorylated successively to nucleoside 5′-monophosphates, 5′-diphosphates, and 5′-triphosphates. In most cases, nucleoside triphosphates, e.g., 3′-azido-3′-deoxythymidine triphosphate (AZT, an anti-HIV drug) and arabinosylcytosine triphosphate (cytarabine, an anticancer drug), are the active chemical entities that inhibit DNA or RNA synthesis, through a competitive inhibition of polymerases and subsequent incorporation of modified nucleotides into DNA or RNA sequences. In a few cases, nucleoside analogs exert effects at lower phosphate levels. For instance, 5-fluoro-2′-deoxyuridine 5′-monophosphate (an anticancer drug) and 2′,2′-difluoro-2′-deoxycytidine 5′-diphosphate (an anticancer drug) have been shown to inhibit thymidylate synthase and ribonucleotide reductase, respectively. Although nucleoside analogs themselves may act at the nonphosphate level such as the inhibitors of adenosine kinases and the ligands of adenosine receptors, currently, clinically-useful nucleoside drugs primarily depend on cellular activation by nucleoside kinases and nucleotide kinases.
  • At least, two criteria are pertinent for nucleoside antiviral drugs: 1) nucleoside analogs should anabolise to nucleotides in cells; and 2) the anabolised nucleotides should target selectively viral enzymes. In order to be phosphorylated in cells and selectively target preferred enzymes, nucleoside analogs should have favourable modifications on their sugar and base moieties. To obtain such favourable nucleoside analogs, a general approach is to generate diverse nucleoside analogs by modifying the base or the sugar, or by modifying both base and sugar moieties. Numerous examples exist in the literature for the synthesis of a variety of modified nucleosides (Chemistry of Nucleosides and Nucleotides Vol. 1 (1988), Vol. 2 (1991), Vol. 3 (1994), edited by L. B. Townsend, Plenum Press; Handbook of Nucleoside Synthesis by H. Vorbrüggen and C. Ruh-Pohlenz, John Wiley & Sons, Inc., 2001; The Organic Chemistry of Nucleic Acids by Y. Mizuno, Elsevier, 1986).
  • However, there are certain classes of nucleoside compounds that were not explored intensively for their antiviral activities before the present invention. A class of such compounds is bicyclic nucleosides which are not derived from purine bases. Disclosures of bicyclic nucleosides are very limited considering that natural adenine and guanine (purines) based ribonucleotides and deoxy derivatives thereof, have bicyclic base moieties. WO 01/92282 A2, WO 01/90121 A2 and WO 04/058792 disclose derivatives of purine nucleosides. In contrast to these publications, the present invention discloses that a certain new class of bicyclic nucleosides and nucleotides display biological activity which may be particularly useful for the treatment of infectious diseases, including viral infections.
  • SUMMARY OF THE INVENTION
  • The present invention relates to a particular class of bicyclic nucleosides, nucleotides, and derivatives thereof and their use in the treatment of microbial infections, and specifically viral infections.
  • In particular, the present invention provides a method for the treatment of a microbial infection, comprising administering an effective amount of a compound of the formula (I) or a pharmaceutically acceptable salt thereof;
    Figure US20100291031A2-20101118-C00001

    wherein:
  • A is O, S, CH2, CHF, CF2 or NR;
  • R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, halogen, OH, N3, CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkynoxy, optionally substituted aryloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted arylalkoxy, optionally substituted heterocycloxy, optionally substituted heteroaryloxy, optionally substituted cycloalkoxy, optionally substituted cycloalkenoxy, optionally substituted amino, optionally substituted aminoacyl, optionally substituted aminoacyloxy, optionally substituted acylamino, optionally substituted oxyacylamino, optionally substituted oxyacyloxy, optionally substituted acylimino, optionally substituted acyliminoxy, optionally substituted oxyacylimino, optionally substituted aminothioacyl, optionally substituted thioacylamino, optionally substituted aminosulfinyl, optionally substituted aminosulfonyl, optionally substituted thio, optionally substituted thioacyl, optionally substituted thioacyloxy, optionally substituted oxythioacyl, optionally substituted oxythioacyloxy, optionally substituted phosphorylamino, optionally substituted sulfinyl, optionally substituted sulfonyl, optionally substituted sulfinylamino, optionally substituted sulfonylamino, optionally substituted oxysulfinylamino, and optionally substituted oxysulfonylamino, or R2 and R2′ together or R3 and R3′ together represents ═O, ═S, or =L-Y′ where L is N, CH, CF, CCl or CBr and Y′ is H, halogen, N3, methyl, ethyl or CN;
  • R4′ is —CY2SH, —CY2OH, —CY2NH2, or -L′-R5;
  • L′ is selected from the group consisting of —CY2—, —CY2CY2—, —CY2OCY2—, —CY2SCY2— and —CY2NHCY2—;
  • each Y is independently selected from the group consisting of H, F, Cl, Br, OR, C1-C6alkyl, C2-C6alkenyl and C2-C6alkynyl, wherein C1-C6alkyl, C2-C6alkenyl, and C2-C6alkynyl may be optionally substituted with one or more groups selected from F, Cl, Br, OH, COOH, COOCH3, SH, SCH3, NH2, NHCH3, N(CH3)2, CN, NO2, C(O)NH2, C(O)NHCH3, N3, C(S)NH2, OCH3, and OCH2CH3;
  • R5 is selected from the group consisting of OR, NR2, monophosphate, diphosphate, and triphosphate, or a mono, di or triphosphate mimic;
  • each R is independently selected from the group consisting of H, CF3, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
  • B is a group of formula (II)
    Figure US20100291031A2-20101118-C00002
  • wherein, if Z is a participant in a π bond (double bond), Z is independently selected from N or C-G; or, if Z is not a participant in a π bond (double bond), Z is independently selected from O, S, Se, NR, NOR, NNR2, CO, CS, CNR, SO, S(O)2, SeO, Se(O)2 or C(G)2, wherein each G is independently selected from the group consisting of H, halogen, OR, SR, NR2, NROR, N3, COOR, CN, CONR2, C(S)NR2, C(═NR)NR2, and R; and
  • where any two adjacent Z are not both selected from O, S, and Se, or not both selected from CO, CS, CNR, SO, S(O)2, SeO, and Se(O)2;
  • wherein, if X is a participant in a π bond (double bond), X is C; or if X is not a participant in a π bond (double bond), X is CR or N;
  • wherein, if R″ is a participant in a π bond (double bond), R″ is O, S, Se, NR, NOR or NNR2; or if R″ is not a participant in a π bond (double bond), R″ is OR, SR, F, Cl, R, or SeR; and
  • dashed lines (---) indicate a possible π or double bond;
  • optionally in combination with one or more antiviral or antibacterial agents.
  • The present invention also further provides the use of a compound of formula (I) or a salt thereof,
    Figure US20100291031A2-20101118-C00003

    wherein:
  • A is O, S, CH2, CHF, CF2 or NR;
  • R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, halogen, OH, N3, CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkynoxy, optionally substituted aryloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted arylalkoxy, optionally substituted heterocycloxy, optionally substituted heteroaryloxy, optionally substituted cycloalkoxy, optionally substituted cycloalkenoxy, optionally substituted amino, optionally substituted aminoacyl, optionally substituted aminoacyloxy, optionally substituted acylamino, optionally substituted oxyacylamino, optionally substituted oxyacyloxy, optionally substituted acylimino, optionally substituted acyliminoxy, optionally substituted oxyacylimino, optionally substituted aminothioacyl, optionally substituted thioacylamino, optionally substituted aminosulfinyl, optionally substituted aminosulfonyl, optionally substituted thio, optionally substituted thioacyl, optionally substituted thioacyloxy, optionally substituted oxythioacyl, optionally substituted oxythioacyloxy, optionally substituted phosphorylamino, optionally substituted sulfinyl, optionally substituted sulfonyl, optionally substituted sulfinylamino, optionally substituted sulfonylamino, optionally substituted oxysulfinylamino, and optionally substituted oxysulfonylamino, or R2 and R2′ together or R3 and R3′ together represents ═O, ═S, or =L-Y′ where L is N, CH, CF, CCl or CBr and Y′ is H, halogen, N3, methyl, ethyl or CN;
  • R4′ is —CY2SH, —CY2OH, —CY2NH2, or -L-R5;
  • L′ is selected from the group consisting of —CY2—, —CY2CY2—, —CY2OCY2—, —CY2SCY2— and —CY2NHCY2—;
  • each Y is independently selected from the group consisting of H, F, Cl, Br, OR, C1-C6alkyl, C2-C6alkenyl and C2-C6alkynyl, wherein C1-C6alkyl, C2-C6alkenyl, and C2-C6alkynyl may be optionally substituted with one or more groups selected from F, Cl, Br, OH, COOH, COOCH3, SH, SCH3, NH2, NHCH3, N(CH3)2, CN, NO2, C(O)NH2, C(O)NHCH3, N3, C(S)NH2, OCH3, and OCH2CH3;
  • R5 is selected from the group consisting of OR, NR2, monophosphate, diphosphate, and triphosphate, or a mono, di or triphosphate mimic;
  • each R is independently selected from the group consisting of H, CF3, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
  • B is a group of formula (II)
    Figure US20100291031A2-20101118-C00004
  • wherein, if Z is a participant in a π bond (double bond), Z is independently selected from N or C-G; or, if Z is not a participant in a π bond (double bond), Z is independently selected from O, S, Se, NR, NOR, NNR2, CO, CS, CNR, SO, S(O)2, SeO, Se(O)2 or C(G)2, wherein each G is independently selected from the group consisting of H, halogen, OR, SR, NR2, NROR, N3, COOR, CN, CONR2, C(S)NR2, C(═NR)NR2, and R; and
  • where any two adjacent Z are not both selected from O, S, and Se, or not both selected from CO, CS, CNR, SO, S(O)2, SeO, and Se(O)2;
  • wherein, if X is a participant in a 7 bond (double bond), X is C; or if X is not a participant in a π bond (double bond), X is CR or N;
  • wherein, if R″ is a participant in a π bond (double bond), R″ is O, S, Se, NR, NOR or NNR2; or if R″ is not a participant in a 71 bond (double bond), R″ is OR, SR, F, Cl, R, or SeR; and
  • dashed lines (---) indicate a possible π or double bond;
  • in the manufacture of a medicament for the treatment of a microbial infection.
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • “Alkyl” refers to monovalent alkyl groups which may be straight chained or branched and preferably have from 1 to 10 carbon atoms or more preferably 1 to 6 carbon atoms. Examples of such alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, n-hexyl, and the like.
  • “Aryl” refers to an unsaturated aromatic carbocyclic group having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl), preferably having from 6 to 14 carbon atoms. Examples of aryl groups include phenyl, naphthyl and the like.
  • “Arylene” refers to a divalent aryl group wherein the aryl group is as described above.
  • “Aryloxy” refers to the group aryl-O— wherein the aryl group is as described above.
  • “Arylalkyl” refers to -alkylene-aryl groups preferably having from 1 to 10 carbon atoms in the alkylene moiety and from 6 to 10 carbon atoms in the aryl moiety. Such arylalkyl groups are exemplified by benzyl, phenethyl and the like.
  • “Arylalkoxy” refers to the group arylalkyl-O— wherein the arylalkyl group are as described above. Such arylalkoxy groups are exemplified by benzyloxy and the like.
  • “Alkoxy” refers to the group alkyl-O— where the alkyl group is as described above. Examples include, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.
  • “Alkenyl” refers to a monovalent alkenyl group which may be straight chained or branched and preferably have from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and have at least 1 and preferably from 1-2, carbon to carbon, double bonds. Examples include ethenyl (—CH═CH2), n-propenyl (—CH2CH═CH2), iso-propenyl (—C(CH3)═CH2), but-2-enyl (—CH2CH═CHCH3), and the like.
  • “Alkenyloxy” refers to the group alkenyl-O— wherein the alkenyl group is as described above.
  • “Alkynyl” refers to alkynyl groups preferably having from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and having at least 1, and preferably from 1-2, carbon to carbon, triple bonds. Examples of alkynyl groups include ethynyl (—C≡CH), propargyl (—CH2C≡CH), pent-2-ynyl (—CH2C≡CCH2—CH3), and the like.
  • “Alkynyloxy” refers to the group alkynyl-O— wherein the alkynyl groups are described above.
  • “Acyl” refers to groups H—C(O)—, alkyl-C(O)—, cycloalkyl-C(O)—, aryl-C(O)—, heteroaryl-C(O)— and heterocyclyl-C(O)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Oxyacyl” refers to groups H—OC(O)—, alkyl-OC(O)—, cycloalkyl-OC(O)—, aryl-OC(O)—, heteroaryl-OC(O)—, and heterocyclyl-OC(O)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Amino” refers to the group —NR′″R′″ where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl is as described herein.
  • “Aminoacyl” refers to the group —C(O)NR′″R′″ where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Acylamino” refers to the group —NR′″C(O)R′″ where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • “Acyloxy” refers to the groups —OC(O)—H, —OC(O)-alkyl, —OC(O)-aryl, —C(O)O-heteroaryl, and —C(O)O-heterocyclyl where alkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Aminoacyloxy” refers to the groups —OC(O)NR′″—H, —OC(O)NR′″-alkyl, —OC(O)NR′″-aryl, —OC(O)NR′″-heteroaryl, and —OC(O)NR′″-heterocyclyl where R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Oxyacylamino” refers to the groups —NR′″OC(O)OH, —NR′″OC(O)O-alkyl, —NR′″OC(O)O-aryl, —NR′″C(O)O-heteroaryl, and NR′″OC(O)O-heterocyclyl where R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Oxyacyloxy” refers to the groups —OC(O)—OH, —OC(O)O-alkyl, —O—C(O)O-aryl, —OC(O)O-heteroaryl, and —OC(O)O-heterocyclyl where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • “Acylimino” refers to the groups —C(NR′″)—R′″ where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • “Acyliminoxy” refers to the groups —O—C(NR′″)—R′″ where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • “Oxyacylimino” refers to the groups —C(NR′″)—OR′″ where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • “Cycloalkyl” refers to cyclic alkyl groups having a single cyclic ring or multiple condensed rings, preferably incorporating 3 to 8 carbon atoms. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl, and the like, or multiple ring structures such as adamantanyl, and the like.
  • “Cycloalkenyl” refers to cyclic alkenyl groups having a single cyclic ring and at least one point of internal unsaturation, preferably incorporating 4 to 8 carbon atoms. Examples of suitable cycloalkenyl groups include, for instance, cyclobut-2-enyl, cyclopent-3-enyl, cyclohex-4-enyl, cyclooct-3-enyl and the like.
  • “Halo” or “halogen” refers to fluoro, chloro, bromo and iodo.
  • “Heteroaryl” refers to a monovalent aromatic heterocyclic group which fulfils the Hückel criteria for aromaticity (i.e., contains 4n+2π electrons, is planar and conjugated) and preferably has from 2 to 10 carbon atoms and 1 to 4 heteroatoms selected from oxygen, nitrogen, selenium, and sulfur within the ring (and includes oxides of sulfur, selenium and nitrogen). Such heteroaryl groups can have a single ring (e.g., pyridyl, pyrrolyl or N-oxides thereof or furyl) or multiple condensed rings (e.g., indolizinyl, benzoimidazolyl, coumarinyl, quinolinyl, isoquinolinyl or benzothienyl).
  • “Heterocyclyl” refers to a monovalent saturated or unsaturated group having a single ring or multiple condensed rings, preferably from 1 to 8 carbon atoms and from 1 to 4 hetero atoms selected from nitrogen, sulfur, oxygen, selenium or phosphorous within the ring. The most preferred heteroatom is nitrogen.
  • Examples of heterocyclyl and heteroaryl groups include, but are not limited to, oxazole, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, isothiazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,7-tetrahydrobenzo[b]thiophene, thiazole, thiadiazoles, oxadiazole, oxatriazole, tetrazole, thiazolidine, thiophene, benzo[b]thiophene, morpholino, piperidinyl, pyrrolidine, tetrahydrofuranyl, triazole, and the like.
  • “Thio” refers to groups H—S—, alkyl-S—, cycloalkyl-S—, aryl-S—, heteroaryl-S—, and heterocyclyl-S—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Thioacyl” refers to groups H—C(S)—, alkyl-C(S)—, cycloalkyl-C(S)—, aryl-C(S)—, heteroaryl-C(S)—, and heterocyclyl-C(S)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Oxythioacyl” refers to groups HO—C(S)—, alkylO—C(S)—, cycloalkylO—C(S)—, arylO—C(S)—, heteroarylO—C(S)—, and heterocyclylO—C(S)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Oxythioacyloxy” refers to groups HO—C(S)—O—, alkylO—C(S)—O—, cycloalkylO—C(S)—O—, arylO—C(S)—O—, heteroarylO—C(S)—O—, and heterocyclylO—C(S)—O—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Phosphorylamino” refers to the groups —NR′″—P(O)(R″″)(OR′″″) where R′″ represents H, alkyl, cycloalkyl, alkenyl, or aryl, R″″ represents OR′″″ or is hydroxy or amino and R′″″ is alkyl, cycloalkyl, aryl or arylalkyl, where alkyl, amino, alkenyl, aryl, cycloalkyl, and arylalkyl are as described herein.
  • “Thioacyloxy” refers to groups H—C(S)—O—, alkyl-C(S)—O—, cycloalkyl-C(S)—O—, aryl-C(S)—O—, heteroaryl-C(S)—O—, and heterocyclyl-C(S)—O—, where alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl are as described herein.
  • “Sulfinyl” refers to groups H—S(O)—, alkyl-S(O)—, cycloalkyl-S(O)—, aryl-S(O)—, heteroaryl-S(O)—, and heterocyclyl-S(O)—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Sulfonyl” refers to groups H—S(O)2—, alkyl-S(O)2—, cycloalkyl-S(O)2—, aryl-S(O)2—, heteroaryl-S(O)2—, and heterocyclyl-S(O)2—, where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Sulfinylamino” refers to groups H—S(O)—NR′″—, alkyl-S(O)—NR′″—, cycloalkyl-S(O)—NR′″—, aryl-S(O)—NR′″—, heteroaryl-S(O)—NR′″—, and heterocyclyl-S(O)—NR′″—, where R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Sulfonylamino” refers to groups H—S(O)2—NR′″—, alkyl-S(O)2—NR′″—, cycloalkyl-S(O)2—NR′″—, aryl-S(O)2—NR′″—, heteroaryl-S(O)2—NR′″—, and heterocyclyl-S(O)2—NR′″—, where R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Oxysulfinylamino” refers to groups HO—S(O)—NR′″—, alkylO—S(O)—NR′″—, cycloalkylO—S(O)—NR′″—, arylO—S(O)—NR′″—, heteroarylO—S(O)—NR′″—, and heterocyclylO—S(O)—NR′″—, where R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Oxysulfonylamino” refers to groups HO—S(O)2—NR′″—, alkylO—S(O)2—NR′″—, cycloalkylO—S(O)2—NR′″—, arylO—S(O)2—NR′″—, heteroarylO—S(O)2—NR′″—, and heterocyclylO—S(O)2—NR′″—, where R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclyl and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Aminothioacyl” refers to groups R′″R′″N—C(S)—, where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, where heterocyclic and alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Thioacylamino” refers to groups H—C(S)—NR′″—, alkyl-C(S)—NR′″—, cycloalkyl-C(S)—NR′″—, aryl-C(S)—NR′″—, heteroaryl-C(S)—NR′″—, and heterocyclyl-C(S)—NR′″—, where R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and where heterocyclyl and alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Aminosulfinyl” refers to groups R′″R′″N—S(O)—, where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclic and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • “Aminosulfonyl” refers to groups R′″R′″N—S(O)2—, where each R′″ is independently hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclic and where alkyl, cycloalkyl, aryl, heteroaryl and heterocyclyl are as described herein.
  • In this specification “optionally substituted” is taken to mean that a group may or may not be further substituted or fused (so as to form a condensed polycyclic group) with one or more groups selected from hydroxyl, acyl, alkyl, alkoxy, alkenyl, alkenyloxy, alkynyl, alkynyloxy, amino, aminoacyl, thio, arylalkyl, arylalkoxy, aryl, aryloxy, acylamino, cyano, halogen, nitro, sulfo, phosphono, phosphorylamino, phosphinyl, heteroaryl, heteroaryloxy, heterocyclyl, heterocycloxy, oxyacyl, acyloxy, oxime, oxime ether, hydrazone, oxyacylamino, aminoacyloxy, trihalomethyl, trialkylsilyl, pentafluoroethyl, trifluoromethoxy, difluoromethoxy, trifluoromethanethio, trifluoroethenyl, mono- and di-alkylamino, mono- and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di-heteroarylamino, mono- and di-heterocyclyl amino, and unsymmetric di-substituted amines having different substituents selected from alkyl, aryl, heteroaryl and heterocyclyl, and the like. For instance, an “optionally substituted amino” group may include amino acid and peptide residues.
  • The term “base”, unless otherwise specified, refers to the base moiety of a nucleoside or nucleotide. The base moiety is the nitrogen-heterocycle portion of a nucleoside or nucleotide. The base moiety of a nucleotide of formula (I) is a bicyclic heterocycle represented by formula (II) and designated “B”. The nucleoside base is attached to the sugar moiety of a nucleoside in such ways that both α and β anomers of D or L nucleosides can be produced. This is denoted by use of the bond
    Figure US20100291031A2-20101118-P00900
    which links the base to the sugar moiety.
  • The term “sugar” refers to the furanose portion of a nucleoside. The sugar moiety of formula (I) nucleosides, nucleotides and nucleotides mimics and/or prodrugs thereof may contain one or more substituents at their C1-, C2-, C3- and C4-position of the furanose. Substituents may be directed to either the α- or β-face of the furanose. The nucleoside base can be considered as a substituent at the C-1 position of the furanose and is preferably directed to the β-face of the sugar. The β-face is the side of a furanose on which a purine or pyrimidine base of natural β-D-nucleosides is present. The α-face is the side of the sugar opposite to the β-face.
  • Examples of a “protecting group” for O, S, or N moieties such as hydroxy or NH2, includes acyl groups, silyl groups, and the like. Suitable protecting groups for these and other moieties are described by T. W., Greene and P. G. M. Wuts; Protecting Groups in Organic Synthesis, 3rd Ed, John Wiley & Sons, Inc. (1999), incorporated herein by reference.
  • The term “infection” or “microbial infection” refers to the infection caused by an infectious agent or microbe, such as bacteria, parasite (including protozoan), virus or fungus (including unicellular and multicellular). Examples of microbes that cause such infection include: Acanthamoeba, African Sleeping Sickness (Trypanosomiasis), amebiasis, American Trypanosomiasis (Chagas Disease), Bilharzia (Schistosomiasis), cryptosporidiosis (diarrheal disease, Cryptosporidium Parvum), Giardiasis (diarrheal disease, Giardia lamblia), hepatitis A, B, C, D, E, leishmaniasis (skin sores and visceral), malaria (Plasmodium falciparum), Salmonella enteritides infection (stomach cramps, diarrhea and fever), tuberculosis (mycobacterium tuberculosis), varicella (chicken pox), yellow fever, pneumonias, urinary tract infections (Chlamydia and Mycoplasma), meningitis and meningococcal septicemia, skin and soft tissue infections (Staphylococcus aureus), lower respiratory tract infections (bacterial pathogens or viral pathogens).
  • Common infections caused by microbes are further outlined in the following chart:
    Infection Bacteria Fungus Protozoa Virus
    AIDS X
    Athlete's Foot X
    Chicken Pox X
    Common Cold X
    Diarrheal Disease X X X
    Dengue X
    Flu X
    Genital Herpes X
    Malaria X X
    Meningitis X
    Pneumonia X X
    Sinusitis X X
    Skin Disease X X X X
    Strep Throat X
    Tuberculosis X
    Urinary Tract Infections X
    Vaginal Infections X X
    Viral Hepatitis X
  • In relation to the therapeutic methods of the present invention the compounds of formula (I) may be particularly useful for treating a microbial infection which is a viral infection caused by an RNA virus, such as a virus belonging to group Flaviviridae, for instance Flaviviruses or HCV, or a DNA or retrovirus such as HBV or HIV. In a preferred embodiment the method of the present invention treats a viral infection caused by an RNA virus of the group Flaviviridae and in particular HCV.
  • The compounds of formula (I) are administered to the subject in a therapeutic effective amount. As used herein, a therapeutic effective amount is intended to include at least partially attaining the desired effect, or delaying the onset of, or inhibiting the progression of, or halting or reversing altogether the onset or progression of the particular disease of condition being treated.
  • As used herein, the term “effective amount” relates to an amount of compound which, when administered according to a desired dosing regimen, provides the desired therapeutic activity. Dosing may occur at intervals of minutes, hours, days, weeks, months or years or continuously over any one of these periods. Suitable dosages lie within the range of about 0.1 ng per kg of body weight to 10 g per kg of body weight per dosage. The dosage may be in the range of 1 μg to 10 g per kg of body weight per dosage, such as is in the range of 1 mg to 10 g per kg of body weight per dosage. In one embodiment, the dosage may be in the range of 1 mg to 500 mg per kg of body weight per dosage. In another embodiment, the dosage may be in the range of 1 mg to 250 mg per kg of body weight per dosage. In yet another preferred embodiment, the dosage may be in the range of 1 mg to 100 mg per kg of body weight per dosage, such as up to 50 mg per body weight per dosage.
  • Suitable dosage amounts and dosing regimens can be determined by the attending physician and may depend on the particular condition being treated, the severity of the condition as well as the general age, health and weight of the subject.
  • The active ingredient may be administered in a single dose or a series of doses. While it is possible for the active ingredient to be administered alone, it is preferable to present it as a composition, preferably as a pharmaceutical composition. The formulation of such compositions is well known to those skilled in the art. The composition may contain any suitable carriers, diluents or excipients. These include all conventional solvents, dispersion media, fillers, solid carriers, coatings, further antifungal and antibacterial agents, dermal penetration agents, surfactants, isotonic and absorption agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
  • The carrier must be pharmaceutically “acceptable” in the sense of being compatible with the other ingredients of the composition and not injurious to the subject. Compositions include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parental (including subcutaneous, intramuscular, intravenous and intradermal) administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • Compositions suitable for oral administration may be presented as discrete units such as capsules, sachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
  • A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g., inert diluent, preservative disintegrant (e.g., sodium starch glycolate, cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethyl cellulose) surface-active or dispersing agent. Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • Compositions suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured base, usually sucrose and acacia or tragacanth gum; pastilles comprising the active ingredient in an inert basis such as gelatine and glycerin, or sucrose and acacia gum; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Compositions suitable for topical administration to the skin may comprise the compounds dissolved or suspended in any suitable carrier or base and may be in the form of lotions, gel, creams, pastes, ointments and the like. Suitable carriers include mineral oil, propylene glycol, polyoxyethylene, polyoxypropylene, emulsifying wax, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. Transdermal patches may also be used to administer the compounds of the invention.
  • Compositions for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter, glycerin, gelatine or polyethylene glycol.
  • Compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Compositions suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bactericides and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Finally, formulations of these compositions in dry powder form for delivery by a dry powder inhaler offer yet another means of administration. This overcomes many of the disadvantages of the oral and intravenous routes.
  • Preferred unit dosage compositions are those containing a daily dose or unit, daily sub-dose, as herein above described, or an appropriate fraction thereof, of the active ingredient.
  • It should be understood that in addition to the active ingredients particularly mentioned above, the compositions may include other agents conventional in the art having regard to the type of composition in question, for example, those suitable for oral administration may include such further agents as binders, sweeteners, thickeners, flavouring agents disintegrating agents, coating agents, preservatives, lubricants and/or time delay agents. Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine. Suitable disintegrating agents include cornstarch, methylcellulose, polyvinylpyrrolidone, xanthan gum, bentonite, alginic acid or agar. Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring. Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten. Suitable preservatives include sodium benzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite. Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc. Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
  • In a further embodiment, the present invention provides novel compounds, pharmaceutical compositions of said novel compounds and therapeutic applications of said novel compounds for the treatment of microbial infections.
  • Accordingly, in another aspect the present invention provides a compound of the formula (I) or a salt thereof;
    Figure US20100291031A2-20101118-C00005

    wherein:
  • A is O, S, CH2, CHF, CF2 or NR;
  • R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, halogen, OH, N3, CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkynoxy, optionally substituted aryloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted arylalkoxy, optionally substituted heterocycloxy, optionally substituted heteroaryloxy, optionally substituted cycloalkoxy, optionally substituted cycloalkenoxy, optionally substituted amino, optionally substituted aminoacyl, optionally substituted aminoacyloxy, optionally substituted acylamino, optionally substituted oxyacylamino, optionally substituted oxyacyloxy, optionally substituted acylimino, optionally substituted acyliminoxy, optionally substituted oxyacylimino, optionally substituted aminothioacyl, optionally substituted thioacylamino, optionally substituted aminosulfinyl, optionally substituted aminosulfonyl, optionally substituted thio, optionally substituted thioacyl, optionally substituted thioacyloxy, optionally substituted oxythioacyl, optionally substituted oxythioacyloxy, optionally substituted phosphorylamino, optionally substituted sulfinyl, optionally substituted sulfonyl, optionally substituted sulfinylamino, optionally substituted sulfonylamino, optionally substituted oxysulfinylamino, and optionally substituted oxysulfonylamino, or R2 and R2′ together or R3 and R3′ together represents ═O, ═S, or =L-Y′ where L is N, CH, CF, CCl or CBr and Y′ is H, halogen, N3, methyl, ethyl or CN;
  • R4′ is —CY2SH, —CY2OH, —CY2NH2, or -L′-R5;
  • L′ is selected from the group consisting of —CY2—, —CY2CY2—, —CY2OCY2—, —CY2SCY2— and —CY2NHCY2—;
  • each Y is independently selected from the group consisting of H, F, Cl, Br, OR, C1-C6alkyl, C2-C6alkenyl and C2-C6alkynyl, wherein C1-C6alkyl, C2-C6alkenyl, and C2-C6alkynyl may be optionally substituted with one or more groups selected from F, Cl, Br, OH, COOH, COOCH3, SH, SCH3, NH2, NHCH3, N(CH3)2, CN, NO2, C(O)NH2, C(O)NHCH3, N3, C(S)NH2, OCH3, and OCH2CH3;
  • R5 is selected from the group consisting of OR, NR2, monophosphate, diphosphate, and triphosphate, or a mono, di or triphosphate mimic;
  • each R is independently selected from the group consisting of H, CF3, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
  • B is a group of formula (II)
    Figure US20100291031A2-20101118-C00006
  • wherein, if Z is a participant in a π bond (double bond), Z is independently selected from N or C-G; or, if Z is not a participant in a π bond (double bond), Z is independently selected from O, S, Se, NR, NOR, NNR2, CO, CS, CNR, SO, S(O)2, SeO, Se(O)2 or C(G)2, wherein each G is independently selected from the group consisting of H, halogen, OR, SR, NR2, NROR, N3, COOR, CN, CONR2, C(S)NR2, C(═NR)NR2, NCONR2, NCSNR2 and R; and
  • where any two adjacent Z are not both selected from O, S, and Se, or not both selected from CO, CS, CNR, SO, S(O)2, SeO, and Se(O)2;
  • wherein, if X is a participant in a π bond (double bond), X is C; or if X is not a participant in a π bond (double bond), X is CR or N;
  • wherein, if R″ is a participant in a π bond (double bond), R″ is O, S, Se, NR, NOR, and NNR2; or if R″ is not a participant in a 71 bond (double bond), R″ is OR, SR, F, Cl, R, or SeR;
  • dashed lines (---) indicate a possible π or double bond; and
  • wherein when R2′, R3′, and R5 are OH or OC(O)CH3, L′ is CH2, A is O, and R1, R2, R3 and R4 are H, B is not the group of formula (III), formula (IV), formula (V), formula (VI), or formula (VII)
    Figure US20100291031A2-20101118-C00007
  • (where all X* are H, or one of X* is CH3 and the other two X are H);
  • when B is a group of formula (III), formula (VIII), or formula (IX)
    Figure US20100291031A2-20101118-C00008
  • R2 and R2′ are not both H;
  • when R2, R3′ and R5 are OH, L′ is CH2, A is O and R1, R2′, R3 and R4 are H, B is not the group of formula (IV); and
  • when R2′ is F, R3′ is OH, R5 is triphosphate, L′ is CH2, A is O, and R1, R2, R3 and R4 are H, B is not a group of formula (X)
    Figure US20100291031A2-20101118-C00009
  • In some preferred embodiments of the invention one or more of the following definitions apply:
  • The bicyclic base structure B of formula II may have one or more ring double bonds and, in some instances, may have two or more ring double bonds. Preferably, the base structure has at least two double bonds and more preferably three or more double bonds.
  • Preferably the base structure B is selected form the following formulae (XI) to (XXI)
    Figure US20100291031A2-20101118-C00010
    Figure US20100291031A2-20101118-C00011

    wherein Z, X and R″ are as defined above.
  • Examples of preferred base structures (B) are as follows:
    Figure US20100291031A2-20101118-C00012
    Figure US20100291031A2-20101118-C00013
    Figure US20100291031A2-20101118-C00014

    wherein G and R are as defined above.
  • Preferably X is N.
  • More preferably, the structure of formula II is represented by the structure of formula IIa
    Figure US20100291031A2-20101118-C00015

    wherein each Z′ is independently N (if a participant in a π bond) or NR (if not a participant in a π bond), and R″, R and Z are as defined above.
  • Even more preferably, the structure of formula II is represented by the structure of formula IIb
    Figure US20100291031A2-20101118-C00016

    wherein R″ and Z′ are as defined above and each Z is independently CG (if a participant is π bond) or >C(G)2 (if not a participant in a π bond).
  • Most preferably, the structure of formula II is represented by the structures of formulae IIc, IId and IIe
    Figure US20100291031A2-20101118-C00017

    wherein R and G are as defined above.
  • Preferably, for the compounds of formulae IIc, IId and IIe each R is independently selected from H, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl and each G is independently selected from H, halogen, CF3, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl.
  • In a further preferred embodiment L′ is —CH2—.
  • In yet a further preferred embodiment R4′ is selected from —CH2—OH, —CF2OH, —CCl2—OH, —C(CH3)(CH)3OH, —CH(CH3)OH, —CH2—CH2—P(O)(OH)2, —CH2—CH2—P(O)(OH)2, —CH2SP(O)(OH)2, —CH2SH, —CF2SH, and —CH2—O—P(O)(OPh)(NHCH(CH3)(CO2Me)).
  • More preferably R4′ is —CH2—OH.
  • Preferably, R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, halogen (more preferably F), OH, N3, CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted alkyloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted amino, optionally substituted aminoacyl, optionally substituted aminoacyloxy, optionally substituted acylamino, optionally substituted thio, or R2 and R2′ together or R3 and R3′ together represents ═O, ═S, or =L-Y′ where L is N, CH, CF, CCl or CBr and Y′ is H, halogen, N3, methyl, ethyl or CN.
  • Even more preferably R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, F, Cl, Br, I, OH, SH, NH2, NHOH, NHNH2, N3, COOH, CN, CONH2, C(S)NH2, COOR, R, OR, SR, SSR, NHR, and NR2 wherein at least one of R2 or R2′ is a substituent other than H, and wherein R is selected from the group consisting of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, and optionally substituted arylalkyl.
  • In yet another preferred embodiment, the sugar moiety is selected from the following formulae:
    Figure US20100291031A2-20101118-C00018

    or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • In another preferred embodiment, at least one of R2 and R2′ is methyl, hydroxyl or F.
  • More preferably, the sugar moiety is selected from the following formulae:
    Figure US20100291031A2-20101118-C00019

    or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • Accordingly, in a preferred embodiment the compound of formula (I) is selected from the following formulae, or salts thereof:
    Figure US20100291031A2-20101118-C00020
    Figure US20100291031A2-20101118-C00021
    Figure US20100291031A2-20101118-C00022

    wherein:
  • each R on the sugar moiety is independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl and optionally substituted arylalkyl;
  • each R on the base moiety is independently selected from H, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
  • each G is independently selected from H, halogen, CF3, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl; and
  • C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • While nucleosides incorporating a variety of sugar moieties have been found to be useful for the inhibition of viral polymerases, in the case of the Flaviviridae, and in particular, the Hepatitis C virus, 2′-C-methyl ribnucleosides have been found to be particularly useful (see Eldrup, A. B. et al., J. Med. Chem. 2004, 47(21), 5284-97, which is incorporated herein by reference).
  • Accordingly, even more preferably, the sugar moiety is represented by the following formula:
    Figure US20100291031A2-20101118-C00023

    or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • Most preferably, the sugar moiety is represented by the following formula:
    Figure US20100291031A2-20101118-C00024

    or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
  • The bicyclic nucleosides of the present invention also include derivatives such as nucleotides, and nucleotide mimics and/or prodrugs thereof.
  • In some embodiments, nucleotide mimics of the compounds of the present invention of formula (I) discussed above include a compound in which R5 is a monophosphate or monophosphate mimic of formula (XXII) or (XXIII):
    Figure US20100291031A2-20101118-C00025

    where X1′X4′, and X6′, independently are O, S, NR; X2′, X3′, and X5′ are selected independently from the group consisting of H, F, NROR, N3, CN, (BH2G)M+, (BH3)M+, R, OR, SR, and NR2. The substituents (BH2G)M+ and (BH3)M+ are ion pairs, which are linked to phosphorus through the negatively charged boron. M+ is a cation, preferably a pharmaceutically acceptable cation such as Ca2+, ammonium, trialkylammonium or tertaalkylammonium, e.g., NH4 +, Et3NH+, Bu3NH+, and Bu4N+.
  • In some embodiments, nucleotide mimics of the compounds of formula (I) as discussed above include di- and triphosphates and di- and tri-phosphate mimics including a compound in which R5 is a di- or tri-phosphate moiety of formula (XXIV):
    Figure US20100291031A2-20101118-C00026
  • X2, X3, and X4 are selected independently from the group consisting of O, S, Se, and NR;
  • X5 and X6 are selected independently from the group consisting of —O—, —S—, —Se—, —CY2C(O)—, —CH(OH)—, —C(OH)2—, —CH2O—, —CH2CH2—, —CH2CH(NH2)—, —CH2CH2CH(NH2)—, —CY2OCY2—, —CY2—, —CRY—, —CY2CY2—, —CHR—, —C≡C—, —HC═CH—, —NH—, —NR—, >NOH, >NOR, >NNH2, and >NNHR;
  • X7, X8, X9, and X10 are selected independently from the group consisting of H, F, OR, SR, NR2, NROR, NRNR2, CN, N3, (BH3)M+, (BH2G)M+, R and SeR;
  • Y, R, (BH2G)M+ and (BH3)M+ are as defined above; and
  • n is 0 or 1.
  • Additional nucleotide phosphate mimics and methods of making the phosphate mimics appropriate for the compounds of the present invention are described, inter alia, in WO 2003/072757 and WO 2003/073989, the entire contents of which are incorporated herein by reference. Many of the nucleotide mimics discussed herein can be prepared by similar approaches as published or by using well-known knowledge of organophosphorous chemistry. Generally, phosphate mimics of the nucleosides and nucleotides of the present invention can inhibit enzyme function without phosphorylation and/or have enhanced nuclease stability relative to nucleotides with unmodified phosphate.
  • Accordingly, the term “phosphate mimic”, unless otherwise specified, refers to a phosphate analog, including, but not limited to, phosphonate, phosphothiolate, phosphoselenoate, selenophosphate, thiophosphate, P-boranophosphate, phosphoramidate, sulfamate, sulfonate, and sulfonamide and/or a combination thereof. Preferred embodiments of the phosphate mimics include phosphonate, phosphoramidate, phosphorothioate, methylphosphonate, fluoromethylphosphonate, difluoromethylphosphonate, vinylphosphonate, phenylphosphonate, sulfonate, fluorophosphate, dithiophosphorothioate, 5′-methylenephosphonate, 5′-difluoromethylenephosphonate, 5′-deoxyphosphonate, 5′-aminophosphoramidate, and 5′-thiophosphate. More preferred is phosphonate and phosphoramidate.
  • Also, it will be appreciated that the term “diphosphate mimic” and “triphosphate mimic” specifically refer to a diphosphate analog and a triphosphate analog, respectively, which comprises at least one of the phosphate mimics, one of the modifications at the bridging site of diphosphate and triphosphate (e.g., X5, X6 and X10), and/or replacements of non-bridging phosphate oxygens (e.g., X4, X3 and X2).
  • The α-P, β-P, and γ-P in the mono, di- and triphosphate mimics may independently adopt either R or S configurations when chiral.
  • Accordingly, in compounds of formula (I) where a chiral centre is present, the invention encompasses enantiomers, or stereoisomers and mixtures thereof, such as enantiomerically enriched mixtures. It will also be appreciated that the base moieties of the present invention may exist as rapidly interconvertible mixtures of isomers. Isomerism of this kind is known in the art as tautomerism. Individual isomers are called tautomers. Where tautomerism is possible the present invention covers all possible tautomers of the compounds of formula (I).
  • The compounds of the present invention can be administered to a subject as a pharmaceutically acceptable salt thereof. It will be appreciated however that non-pharmaceutically acceptable salts also fall within the scope of the present invention since these may be useful as intermediates in the preparation of pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts include, but are not limited to salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, maleic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, toluenesulphonic, benezenesulphonic, salicyclic sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.
  • Base salts include, but are not limited to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylammonium. In particular, the present invention includes within its scope cationic salts eg sodium or potassium salts, or alkyl esters (eg methyl, ethyl) of the phosphate group.
  • Basic nitrogen-containing groups may be quarternised with such agents as lower alkyl halide, such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl and diethyl sulfate; and others.
  • In some embodiments, the bicyclic nucleosides and nucleotides of the present invention also include their prodrug derivatives. The term “prodrug” is used in its broadest sense and encompasses those derivatives that are converted in vivo to the compounds of the invention. Prodrugs of the compounds of the present invention may be prepared by modification of the sugar moiety or of the phosphate or phosphate mimic to include a prodrug substituent. Such prodrug modification is generally performed to enhance drug absorption and/or drug delivery into cells.
  • Prodrugs substituents include, but are not limited to residues of: proteins; antibiotics; D- and L-amino acids which may be attached to a phosphate moiety or a phosphate mimic moiety via a carbon atom (phosphonates), a nitrogen atom (phosphoamidates), or an oxygen atom (phosphoesters) or may be attached to the sugar moiety through any one or more of the R1-R5 groups; peptides (preferably up to 10 amino acids) attached to a phosphate moiety or a phosphate mimic moiety via a carbon atom (phosphonates), a nitrogen atom (phosphoamidates), or an oxygen atom (phosphoesters), or may be attached to the sugar moiety through any one or more of the R1-R5 groups; drug moieties attached to a phosphate moiety or a phosphate mimic moiety via a carbon atom (phosphonates), a nitrogen atom (phosphoamidates), or an oxygen atom (phosphoesters), or may be attached to the sugar moiety through any one or more of the R1-R5 groups; as well as including steroids; vitamins; polyamines; carbohydrates; polyethylene glycols (PEGs); cyclosaligenyls; substituted 4 to 8-membered rings, with or without heteroatom substitutions, 1,3-phosphoamidate attachments to a terminal phosphate or phosphate mimic moiety (γ or β) or connecting between an α,β or β,γ of a phosphate moiety or phosphate mimic moiety, and so on.
  • In addition to those described herein, prodrug derivatives of nucleosides, nucleotides and nucleotide phosphate mimics and methods of making the prodrugs appropriate for use in the present invention are described, inter alia, in PCT Publications WO 2003/072757 and WO 2003/073989.
  • The prodrug of a nucleoside 5′-monophosphate mimic can mask the negative charges of the phosphate mimic moiety entirely or partially, or mask the negative charges of the di-phosphate mimic or tri-phosphate mimic moiety or phosphate moiety in order to enhance drug absorption and/or drug delivery into cells.
  • In one embodiment a combination of prodrug substituents may be attached to one or more X2′, X3′ and X5′ positions on a nucleoside mono-phosphate mimic or to one or more X7-X10 positions on a nucleoside di- or tri-phosphate mimic. Preferred prodrug substituents in positions X2′, X3′ or X5′ position include 2,3-O-diacylglyceryloxy, 2,3-O-dialkylglyceryloxy, 1-O-alkyl-2-O-acylglyceryloxy, 1-O-acyl-2-O-alkylglyceryloxy, 1-S-alkyl-2-O-acyl-1-thioglyceryloxy, acyloxymethoxy, S-acyl-2-thioethoxy, S-pivaloyl-2-thioethoxy, acyloxymethoxy, pivaloyloxymethoxy, alkoxycarbonyloxymethoxy, S-alkyldithio-S′-ethyoxy acyloxymethoxy, S-acyl-2-thioethoxy, S-pivaloyl-2-thioethoxy, pivaloyloxymethoxy, alkoxycarbonyloxymethoxy, and S-alkyldithio-S′-ethyoxy.
  • In a further embodiment, the prodrug substituent is a substituent on a hydroxyl group of the sugar moiety (that is, for instance, any one of R1-R5). Preferably, the modification results in the formation of an ester and in this regard the preferred prodrug substituents are C1-C6acyl groups for example, acetyl, propionyl, pivaloyl, etc. Also preferred are substituted C1-C6 acyl moieties, for example, fluoroacetyl, difluoroacetyl, etc. More preferably the substituted C1-C6 acyl group is represented as a residue of an L or D amino acid consisting of alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine, aspartic acid, glutamic acid, arginine, histidine, and lysine. Most preferably the prodrug substituent is an amino acid residue of D or L-valine.
  • The prodrug can be activated either by cellular enzymes such as lipases, esterases, reductases, oxidases, nucleases or by chemical cleavage such as hydrolysis to release (liberate) either the nucleoside, nucleotide or nucleotide mimic after the prodrug enters cells.
  • In addition to using prodrug approaches, the delivery of the nucleosides and nucleotides may be assisted by using a therapeutically acceptable carrier such as liposomal suspensions, cationic lipids, and polyimines.
  • The novel nucleosides, nucleotides, nucleotide mimics and prodrugs thereof, of the present invention can be prepared by those who are skilful in synthetic organic and nucleoside chemistry using established synthetic methodology (Chemistry of Nucleosides and Nucleotides Vol. 1 (1988), Vol. 2 (1991), Vol. 3 (1994), edited by L. B. Townsend, Plenum Press; Handbook of Nucleoside Synthesis by H. Vorbrüggen and C. Ruh-Pohlenz, John Wiley & Sons, Inc., 2001; The Organic Chemistry of Nucleic Acids by Y. Mizuno, Elsevier, 1986).
  • The nucleosides of the present invention can be converted to their corresponding monophosphate, diphosphate, and triphosphate nucleosides by established phosphorylation procedures. Similarly, known methods in the art can be used to synthesise the nucleotide and phosphate mimics and prodrugs. The following schemes and description serve as representative syntheses of the nucleosides and nucleotides of the present invention. As such, other compounds such as those having —CY2SH, —CY2OH or -L′-R5 groups other than CH2R5 may similarly be made.
  • The bicyclic nucleosides of the present invention may be prepared by modification of optionally protected and functionalised cytosine, uracil and other base analogues followed by Stille, Heck, Sonogashira or other metal-mediated cross coupling chemistry to introduce an α,β-unsaturated ester, alkyne or other functional group. Such processes allow for stereoselective synthesis of an intermediate capable of efficient cyclisation to form the bicyclic compounds of the present invention. Cyclisation and optional deprotection of the product delivers the target bicyclic nucleoside.
  • Any compound capable of metal-mediated cross coupling may be used, such as a tin derivative like trialkyltin. More preferably tributyltin. Preferably the reactions are carried out using a palladium based coupling agent. Suitable coupling agents are known in the art and include Pd(PPh3)2Cl2, Pd(PPh3)4, Pd(dibenzylideneacetone), and PdCl2(CH3CN)2. Also, preferably the palladium catalysed coupling reactions may also include a co-catalyst, for instance, CuI which may be in the presence of a suitable non-nucleophilic base such as a trialkylamine.
  • Coupling reactions are generally performed at temperatures around room temperature. Elevated temperatures such as temperatures between 30-80° C. can be employed to effect coupling and cyclisation in a single step. It is also preferred that such reactions are carried out under an inert atmosphere of either nitrogen or argon. Suitable solvents include ether solvents such as THF and diethylether or polar solvents such as DMF.
  • For example, Schemes 1 and 2 illustrates some preferred cyclisation methods for forming the 6-membered ring portions of the bicyclic bases (B) of the compounds of the present invention.
    Figure US20100291031A2-20101118-C00027
  • Methodologies other than metal-mediated cross-coupling and cyclisation can also be used to prepare the bicyclic nucleosides. For example, the final process in scheme 2 depicts how a bifunctional two atom unit (such as an α-halocarboxylic ester or glyoxal) can be used to form the second 6-membered ring portion of a suitable nucleoside intermediate.
    Figure US20100291031A2-20101118-C00028
  • In the cyclisation methodology depicted in Schemes 1 and 2 each of Z1-Z4 is independently Z.
  • Preferably, in this process A is O, CH2 or optionally protected N; Y is halogen or other appropriate group such as trifluoromethanesulfonate; W4 is H or trialkyltin; Z1 and Z2 are each independently C, CH, C-halogen, C-alkyl, C-aryl, C-heteroaryl, C—O-alkyl or C—S-alkyl; Z3 is CH, C-alkyl, C-halogen, N, CNHR, CNH2, CNR2, C═O, or C═S; Z4 is CH, C-halogen, C-alkyl, C-aryl, C-heteroaryl, C—O-alkyl, C—S-alkyl, C—OH, C—NH2, C—NHR, C—NR2 or C═O; R1, R2, R2′R3, R3′, R4′ are each independently H, halogen, alkyl, O-alkyl, OH, optionally protected O, methyl, H or F; and R5 is an optionally protected OH or NH2; and (--) denotes an optional double bond.
  • The bicyclic bases of the present invention can be further modified either prior to being added to the sugar moiety or once attached. Such modification may involve transformations through functionalisation, defunctionalisation, or functional group interconversion. Modifications may include esterification, the preparation of enol ethers, o-alkylation, bromination, hydrogenation, dihydroxylation, epoxidation, oximation, and amination.
  • The compounds described herein can also be converted into their corresponding mono-, di- and triphosphates using well established methods. Furthermore, as discussed above prodrugs of mono-, di- and triphosphates can be prepared in order to optimise the biological efficacy of these phosphorylated compounds. Methods for preparing such prodrugs are well known in the art (see Wagner, C. R., et al. Med. Res. Rev., 2000, 20, 417-451).
    Figure US20100291031A2-20101118-C00029
  • In Scheme 3, preferably A is O, CH2 or optionally protected N; R1, R2, R2′, R3, R3′, R4 are each independently H, halogen, alkyl, O-alkyl, OH, optionally protected O, or methyl and B is as described herein.
  • As discussed earlier an alternative to the use of phosphates is the use of phosphate mimics and their prodrugs. One such phosphate mimic is shown below and this can be prepared using appropriately protected nucleosides and known conditions.
    Figure US20100291031A2-20101118-C00030
  • In Scheme 4, preferably A is O, CH2 or optionally protected N; R1, R2, R2′, R3, R3′, R4 are each independently H, halogen, alkyl, O-alkyl, OH, optionally protected O, methyl or F; X′ is O, S, NH, CF2, CHF, CClH, CBr2 or CHBr; B is as described herein.
  • The bicyclic compounds of the present invention may be tested for biological activity using well known procedures.
  • Antiviral assays are conducted according to published, widely used protocols. In order to obtain the therapeutic index, compound-induced cytotoxicity to host cells is also measured in parallel with antiviral activities. To determine the mode of action of antiviral nucleosides the corresponding nucleoside triphosphates are subject to enzyme-based assays for the inhibition of viral polymerases according to known protocols (Ranjith-Kumar et al. J. Virol. 2001, 75, 8615; Dhanak et al. J. Biol. Chem. 2002, 277, 38322-38327). Some compounds of the present invention showed Ki values of less than 1 μM against HCV NS5B.
  • Since the replicon RNA replication mimics the replication of HCV RNA in infected hepatocytes, compounds that have the inhibitory effects in replicon assays are potentially useful as anti-HCV drugs. The HCV replicon-containing cell lines (Randall and Rice, Current Opinion in Infectious Diseases 2001, 14, 743) are used for the identification of potential anti-HCV compounds. Among them is a widely used subgenomic replicon system developed by Lohmann et al. (Science 1999, 285, 110; J. General Virol. 2000, 81, 1631; J. Virol. 2001, 75, 1437, 2002, 76, 4008). Some compounds of the present invention showed potent anti-HCV activity with EC50 values of low μM.
  • Widely used protocols developed by Korba et al. (Antiviral Res. 1992, 19, 55), and Pai et al. (Antimicrobial Agents Chemother. 1996, 40, 380) are useful for the determination of in vitro anti-HBV activity.
  • Anti-HIV assays can be conducted according to the protocols developed by Schinazi et al. (Antimiromobial Agents Chemother. 1990, 34, 1061; 1992, 36, 2423; 1993, 37, 875) or other widely used protocols (Kimpton et al J. Virol. 1992, 66, 2232; Chan et al. J. Med. Chem. 2001, 44, 1866).
  • Preferred nucleoside triphosphates of the present invention may act as potent inhibitors of the non-structural position SB (NS5B) which is HCV's RNA-dependent RNA polymerase. Accordingly, such compounds are preferably suited to treat and/or prevent HCV. Also, as the preferred novel compounds of the present invention are expected to exhibit novel profiles of activity they may provide the artisan with an alternative to treating viruses which display drug resistance to conventional drugs. Other advantages which may be exhibited by the preferred novel compounds of the present invention include:
      • reduced toxicity and tolerability relative to existing therapies and those in development; and/or
      • improved pharmacokinetic properties.
  • Accordingly, nucleosides, nucleotide, nucleotide mimics and/or their prodrugs of the present invention may be useful for the inhibition of a variety of enzymes including, but not limited to, DNA or RNA polymerases, helicases, ribonucleotide reductases, protein kinases, and telomerases and for the modulation of G-proteins, P2 purinergic receptors and the allosteric sites of a variety of enzymes. Preferably, the novel nucleosides, nucleotides, nucleotide mimics and/or prodrugs of the present invention are used to treat viral infections caused by the RNV viruses of the group Flaviviridae and, in particular, HCV.
  • The novel nucleosides, nucleotides, nucleotide mimics and/or their prodrugs of the present invention are useful for the treatment of infectious diseases caused by infectious agents such as parasites, bacteria and fungi.
  • Also, the novel nucleosides, nucleotide mimics and/or their prodrugs that display potent cytotoxicities to fast-dividing cancerous cells may be useful for the treatment of proliferative disorders, including, but not limited to, lung cancer, liver cancer, prostate cancer, colon cancer, breast cancer, ovarian cancer, melanoma, and leukemia.
  • As the ligands of P2 receptors and G-proteins as well as the inhibitors of protein kinases, the novel nucleosides, nucleotides, nucleotide mimics and/or their prodrugs of the present invention may also be useful for the treatment of a wide range of other diseases and disorders such as inflammatory diseases, autoimmune diseases, Type 2 diabetes, and cardiovascular diseases.
  • In order to overcome drug resistance, combination therapies are widely used in the treatment of infectious diseases and proliferative disorders. The nucleosides, nucleotides, nucleotide mimics and/or their prodrugs of the present invention may be therapeutically administered as a single drug, or alternatively may be administered in combination with one or more other active chemical entities to form a combination therapy. The other active chemical entities may be a small molecule, a polypeptide, or a polynucleotide.
  • For instance, compounds of this invention may be particularly useful when used in combination with other agents known to exert an antiviral effect. For example, combination with immunomodulatory/antiviral agents such as interferons, interferon derivatives and other large or small molecules known to modulate host immune responses may be beneficial. Similarly, combinations of compounds of this invention with IMPDH inhibitors (e.g., ribavirin), antiviral nucleosides, antiviral non-nucleosides (e.g., polymerase inhibitors, protease inhibitors) could augment the activity of the bicyclic nucleosides when administered alone.
  • The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to prepare and use the compounds disclosed and claimed herein.
  • All references mentioned herein are incorporated herein by reference in their entirety.
  • The abbreviations that may be used herein, including the Schemes and experimental section are as follows unless indicated otherwise:
  • Bu: n-butyl
  • Bn benzyl
  • Bz: benzoyl
  • DCM: dichloromethane
  • DIEA: diisopropylethylamine
  • DMF: dimethylformamide
  • Et: ethyl
  • EtOAc: ethyl acetate
  • Me: methyl
  • MeOH: methyl alcohol
  • MS: mass spectrometry
  • NMR: nuclear magnetic resonance
  • Ph: phenyl
  • HPLC: high performance liquid chromatography
  • TEA: triethylamine
  • TFA: trifluoroacetic acid
  • THF: tetrahydrofuran
  • The following Examples are offered to illustrate but not to limit the invention.
  • EXAMPLES Synthesis
  • TABLE 1
    Example Structure Molec Formula MWt
    1
    Figure US20100291031A2-20101118-C00031
    C13H15N3O6 309.28
    2
    Figure US20100291031A2-20101118-C00032
    C13H14IN3O6 435.17
    3
    Figure US20100291031A2-20101118-C00033
    C13H14BrN3O6 388.17
    4
    Figure US20100291031A2-20101118-C00034
    C13H14ClN3O6 343.72
    5
    Figure US20100291031A2-20101118-C00035
    C13H15N3O6 309.28
    6
    Figure US20100291031A2-20101118-C00036
    C14H17N3O6 323.30
    7
    Figure US20100291031A2-20101118-C00037
    C20H21N3O6 399.40
    8
    Figure US20100291031A2-20101118-C00038
    C16H21N3O6 351.36
    9
    Figure US20100291031A2-20101118-C00039
    C14H19N3O6 325.32
    10
    Figure US20100291031A2-20101118-C00040
    C20H23N3O6 401.42
    11
    Figure US20100291031A2-20101118-C00041
    C17H24N4O5 364.40
    12
    Figure US20100291031A2-20101118-C00042
    C17H24N4O6 380.40
    13
    Figure US20100291031A2-20101118-C00043
    C18H26N4O5 378.43
    14
    Figure US20100291031A2-20101118-C00044
    C17H25N5O5 379.41
    15
    Figure US20100291031A2-20101118-C00045
    C13H18N3O15P3 549.21
  • Experimental Data
  • 1H and 31P NMR spectra were recorded on either a Bruker Avance DRX 400, AC 200 or AM 300 spectrometer. Spectra were recorded in CDCl3, d6-acetone, CD3OD or d6-DMSO using the residual solvent peak as a reference. Chemical shifts are reported on the δ scale in parts per million (ppm) using the following conventions to assign the multiplicity: s (singlet), d (doublet), t (triplet), q (quartet) m (multiplet) and prefixed b (broad). Mass spectra (ESI) were recorded on a Finnigan LCQ Advantage spectrometer. All microwave reactions were carried out in a CEM Discover microwave reactor. Flash and radial chromatography was performed on 40-63 μm silica gel 60 (Merck No. 9385). Preparative HPLC was carried out using a Gilson 322 pump with a Gilson 215 liquid handler and a HP1100 PDA detector. HPLC systems employed Phenomonex C8(2) columns using either acetonitrile or acetonitrile containing 0.06% TFA in water or water containing 0.1% TFA. Alternatively, a Phenomonex C18 column was used with acetonitrile and aqueous 1M triethylammonium acetate (primarily for phosphate nucleotides).
    Figure US20100291031A2-20101118-C00046

    Intermediate A
  • N,O-Bis(trimethylsilyl)acetamide (7.17 mL) was added to 5-iodocytidine (2.32 g) in dry acetonitrile (15 mL) and the mixture was heated under argon at 80° C. for 30 mins. A suspension of commercially available 1,2,3,5-tetra-O-benzoyl-2-C-methyl-alpha/beta-D-ribofuranose (5.16 g) in dry acetonitrile (40 mL) was added and the mixture heated for 1 h. SnCl4 (1.71 mL) was added cautiously and heating continued for 2 hrs. The reaction was cooled to ambient temperature, poured into an ice cold solution of saturated aqueous sodium bicarbonate and extracted with EtOAc (×3). The combined organic extracts were washed with brine, dried and evaporated in-vacuo. The residue was purified by flash chromatography on silica eluting with 3% MeOH/DCM. The product was obtained as a gum (3.36 g). MS m/z ([M+H]+) 695.7; ([2M+H]+) 1390.8.
  • Intermediate B
  • A degassed solution of intermediate A (1.39 g) and (Z)-ethyl-3-(tributylstannyl) propenoate (1.56 g) was stirred in dry DMF under argon for 10 mins. CuI (76 mg) and PdCl2(PPh3)2 (140 mg) were added and the mixture heated at 70° C. under argon for 16 hrs. The reaction was cooled to ambient temperature, diluted with water (100 mL) and filtered. The residue was washed with water, dissolved in EtOAc and washed again with water. The organic solution was dried (MgSO4) and the solvent evaporated in-vacuo to provide a brown gum. The aqueous mother liquor was also extracted with EtOAc (×2) and the combined organics dried (MgSO4), and evaporated in-vacuo. The combined crude extracts were purified by radial chromatography on silica eluting with 3%-5% MeOH/DCM to give the required product as an oil, which, on trituration with ether afforded a yellow crystalline solid (70 mg). MS m/z ([2M+H]+) 1243.5. Further product was available from the mother-liquor.
  • Preparation of Intermediates C, D, E
  • Preparation of Intermediate C is an example of the general method.
  • A mixture of Intermediate B (125 mg) and N-iodosuccinimide (90.6 mg) in dry acetonitrile (1.50 mL) was heated in a microwave reactor at 120° C. for 30 mins (initial power 200 W). The solution was evaporated in-vacuo, dissolved in EtOAc and washed with 10% sodium metabisulphite, brine, and evaporated in-vacuo. The crude material was purified by radial chromatography on silica eluting with 3% MeOH/DCM to yield the required product (120 mg). MS m/z ([2M+H]+) 1494.3.
  • Similarly for Intermediates D and E.
  • Intermediate D
  • Intermediate B (62 mg) yielded intermediate D (45 mg). MS m/z ([2M+H]+) 1400.6, 1401.6, 1403.6.
  • Intermediate E
  • Intermediate B (62 mg) yielded 62 mg intermediate D. MS m/z ([2M+H]+) 1310.6, 1311.7, 1313.7.
  • Preparation of Examples 1-4
  • Preparation of Example 1 is an example of the general method.
  • Example 1
  • Intermediate B (60 mg) was suspended in dry methanol (1 mL) under argon. A freshly prepared solution of 1M sodium methoxide in methanol (0.5 mL) was added and the reaction stirred for 18 hrs. Evaporation of the solvent in-vacuo with minimal heating and purification by radial chromatography on silica eluting with 20% MeOH/DCM yielded the required compound as a crystalline solid.
  • MS m/z ([M+H]+) 309.9. 1H 1H NMR (CD3OD+D2O) δ 9.24 (s, 1H), 7.61 (d, 1H), 6.23 (d, 1H), 6.11 (s, 1H), 4.08-4.00 (m, 2H), 3.93-3.83 (m, 2H), 1.12 (s, 3H).
  • Similarly for Examples 2 and 3.
  • Example 2
  • Intermediate C (12 mg) yielded Example 2 (1.70 mg)
  • MS m/z ([M+H]+) 435.7, ([2M+H]+) 870.4. 1H NMR (CD3OD) δ 9.26 (s, 1H), 8.18 (s, 1H), 6.10 (s, 1H), 4.07-4.00 (m, 2H), 3.95-3.82 (m, 2H), 1.12 (s, 3H).
  • Example 3
  • Intermediate D (21 mg) yielded Example 3 (4.1 mg)
  • MS m/z ([M+H]+) 387.7, 389.7. 1H NMR (CD3OD) δ 9.28 (s, 1H), 7.95 (s, 1H), 6.10 (s, 1H), 4.07-3.99 (m, 2H), 3.89-3.82 (m, 2H), 1.12 (s, 3H).
  • Example 4
  • Intermediate E (28 mg) was stirred with 7M NH3 in MeOH at ambient temperature for 18 h. The reaction was evaporated to dryness in-vacuo, dissolved in water (2 mL) and washed with DCM (×3) and EtOAc. The aqueous phase was evaporated and the residue triturated with ether to yield the required compound as a yellow solid (12 mg).
  • MS m/z ([M+H]+) 343.7, 345.7; ([2M+H]+) 686.6, 688.6. 1H NMR (CD3OD) δ 9.34 (s, 1H), 7.73 (s, 1H), 6.11 (s, 1H), 4.07-3.99 (m, 2H), 3.90-3.82 (m, 2H), 1.12 (s, 3H).
  • Example 5
  • Figure US20100291031A2-20101118-C00047
  • Example 5 (10.5 mg) was prepared directly from commercially available 5-Iodo-2′-O-methylcytidine using the method described for intermediate B in scheme 1.
  • MS m/z ([M+H]+) 309.8; ([2M+H]+) 618.6, ([2M+Na]+) 640.8. 1H NMR (CD3OD+D2O) δ 9.23 (s, 1H), 7.64 (d, 1H), 6.27 (d, 1H), 5.95 (s, 1H), 4.21-4.17 (m, 1H) 4.06-4.02 (m, 2H), 3.95-3.73 (m, 2H), 3.64 (s, 3H).
    Figure US20100291031A2-20101118-C00048
    Figure US20100291031A2-20101118-C00049

    Intermediate F
  • N,O-Bis(trimethylsilyl)acetamide (6.45 mL) and uracil (0.986 g) in dry acetonitrile (20 mL) were heated under argon at 80° C. for 30 mins. A suspension of commercially available 1,2,3,5-tetra-O-benzoyl-2-C-methyl-alpha/beta-D-ribofuranose in dry acetonitrile (80 mL) was added and heating continued for 1 hr. SnCl4 was added cautiously and heating continued for 5 hrs. The reaction was cooled to ambient temperature, poured into an ice cold solution of saturated sodium bicarbonate (100 mL) and extracted with EtOAc (×3). The combined organic extracts were washed with brine, dried (MgSO4) and evaporated in-vacuo to provide crude product as a frothy white gum (4.7 g) which was used in subsequent reactions without further purification. MS m/z ([M+H]+) 571, ([2M+Na]+) 1163.
  • Intermediate G
  • 1,2,4-triazole (207 mg) was dissolved in dry acetonitrile (5 mL) under argon, cooled to 0° C. and treated with POCl3 (75 μL). After 5 mins, triethylamine (0.60 mL) was added and stirring continued for 1 h at ambient temperature before addition of Intermediate F in dry acetonitrile (5 mL). The reaction was diluted with EtOAc after 90 mins and washed successively with saturated NaHCO3 and brine (each ×3). The organic layer was dried (MgSO4) and evaporated in-vacuo to provide the crude triazole intermediate as a yellow gum (127 mg). This gum was suspended in MeOH (5 mL) and 2M MeNH2 in MeOH (0.20 mL) added followed by vigorous stirring for 20 mins. After this time, volatile materials were evaporated in-vacuo and the residue purified by radial chromatography on silica eluting with 2% MeOH/DCM to give the required product as a gum (46 mg). MS m/z ([M+H]+) 583.9, ([2M+H]+) 1166.9.
  • Intermediate H
  • The crude triazole intermediate (621 mg) was prepared using the method described for intermediate G. This gum was suspended in dry ethanol (10 mL) before addition of benzylamine (164 μl). After stirring for 1 hr at ambient temperature the solvent was removed in-vacuo and the residue purified by radial chromatography on silica eluting with 3%-5% MeOH/DCM to give the required product as an oil (424 mg). MS m/z ([M+H]+) 660.4, 661.4.
  • Intermediate I
  • The crude triazole intermediate (249 mg) was prepared using the method described for intermediate G. This gum was suspended in dry methanol (5 mL) before addition of N-propylamine (49 μl). After stirring for 30 mins at ambient temperature the solvent was removed in-vacuo and the residue purified by radial chromatography on silica eluting with 3% MeOH/DCM to give the required product as gum (45 mg). MS m/z ([M+H]+) 612.4, 613.4.
  • Intermediates J, K and L were prepared by the general method described previously for Intermediate C.
  • Intermediate J
  • Intermediate G (200 mg) yielded Intermediate J (87 mg)
  • MS m/z ([M+H]+) 709.7.
  • Intermediate K
  • Intermediate H (188 mg) yielded Intermediate K (110 mg).
  • MS m/z ([M+H]+) 785.9.
  • Intermediate L
  • Intermediate I (130 mg) yielded Intermediate L (106 mg).
  • MS m/z ([M+H]+) 738.2
  • Intermediates M, N and O were prepared by the general method described previously for Intermediate B.
  • Intermediate M
  • A degassed solution of intermediate J (100 mg) and (Z)-ethyl-3-(tributylstannyl) propenoate (110 mg) was stirred in dry DMF under argon for 10 mins. CuI (5.5 mg) and PdCl2(PPh3)2 (10 mg) were added and the mixture heated at 70° C. under argon for 16 hr. The solvent was evaporated in-vacuo and the residue partitioned between EtOAc and water. The aqueous phase was further extracted with EtOAc (×2) and the combined organic extracts dried (MgSO4) and evaporated in-vacuo. The crude product was purified by radial chromatography on silica eluting with 3% MeOH/DCM to provide a yellow gum (56 mg). MS m/z ([M+H]+) 635.8, ([2M+H]+) 1270.7.
  • Intermediate N
  • Intermediate K (120 mg) yielded Intermediate N (19 mg).
  • MS m/z ([M+H]+) 711.7, ([2M+H]+) 1422.5.
  • Intermediate O
  • Intermediate L (105 mg) yielded Intermediate O (19 mg).
  • MS m/z ([M+H]+) 663.9, ([2M+H]+) 1326.9.
  • Example 6
  • Intermediate M (36 mg) was stirred with 7M NH3 in MeOH (3 mL) at ambient temperature for 4 hrs. The reaction was evaporated to dryness in-vacuo and the residue purified by radial chromatography on silica eluting with DCM/MeOH/25% NH3 90:9:1 to yield the required compound as a yellow solid (6 mg).
  • MS m/z ([M+H]+) 323.8. 1H 1H NMR (CD3OD) δ 9.28 (s, 1H), 7.51 (d, 1H), 6.33 (d, 1H), 6.12 (s, 1H), 4.08-3.99 (m, 2H), 3.91-3.86 (m, 2H), 3.53 (s, 3H), 1.12 (s, 3H).
  • Similarly for Examples 7 and 8.
  • Example 7
  • Intermediate N (19 mg) yielded Example 7 (3.9 mg).
  • MS m/z ([M+H]+) 399.9. 1H 1H NMR (CD3OD) δ 9.30 (s, 1H), 7.54 (d, 10H), 7.40-7.35 (m, 2H), 7.26-7.17 (m, 3H), 6.36 (d, 1H), 6.12 (s, 1H), 5.44 (s, 1H), 4.07-3.99 (m, 2H), 3.91-3.81 (m, 2H), 1.12 (s, 3H).
  • Example 8
  • Intermediate O (38 mg) yielded Example 8 (8.0 mg).
  • MS m/z ([M+H]+) 351.8. 1H 1H NMR (CD3OD) δ 9.30 (s, 1H), 7.50 (d, 1H), 6.32 (d, 1H), 6.12 (s, 1H), 4.21-4.16 (m, 2H), 4.08-3.99 (m, 2H), 3.91-3.83 (m, 2H), 1.74-1.61 (m, 2H), 1.12 (s, 3H), 0.93 (t, 3H).
  • Example 9
  • Example 6 (20 mg) was dissolved in methanol (3 mL) and NH4Cl (10 mg) in water (0.2 mL) was added followed by NaBH4 (10 mg). After 10 mins the reaction was evaporated to dryness in-vacuo and the residue dissolved in water (3 mL) and washed with DCM. The aqueous extract was then purified by preparative HPLC to afford the product as an oil (4.23 mg).
  • MS m/z ([M+H]+) 326.0. 1H 1H NMR (CD3OD) δ 7.26 (d, 1H), 6.10 (d, 1H), 5.84 (s, 1H), 4.60 (d, 1H), 3.98 (d, 1H), 3.92-3.68 (m, 4H), 3.50 (s, 3H), 1.22 (s, 3H).
  • Similarly for Example 10.
  • Example 10
  • Example 7 (20 mg) yielded Example 10 (7.0 mg).
  • MS m/z ([M+H]+) 402.0. 1H 1H NMR (CD3OD) δ 7.28-7.18 (m, 6H), 6.08 (d, 1H), 5.75 (s, 1H), 5.45 (d, 1H), 5.38 (d, 1H), 4.62 (d, 1H), 4.01 (d, 1H), 3.93-3.89 (m, 1H), 3.80-3.68 (m, 3H), 1.19 (s, 3H).
    Figure US20100291031A2-20101118-C00050

    Intermediate P
  • A degassed solution of intermediate A (695 mg) and propargyl alcohol (175 μl) was stirred in dry DMF (7 mL) under argon. CuI (38 mg) and PdCl2(PPh3)2 (70 mg) were added followed by TEA (278 μl) and the mixture stirred at ambient temperature for 18 hrs. The solvent was evaporated in-vacuo and the residue partitioned between EtOAc and water. The organic layer was washed with water, dried (MgSO4) and evaporated in-vacuo. The crude product was purified by radial chromatography on silica eluting with DCM then 5-10% MeOH/DCM to provide a gum (257 mg). MS m/z ([M+H]+) 624.0, ([2M+H]+) 1247.1.
  • Intermediate Q
  • Intermediate P (248 mg) was suspended in DCM (10 mL) and treated with Dess-Martin periodinane (338 mg) at ambient temperature. The reaction was diluted with DCM after 2 hrs and washed with saturated aqueous NaHCO3. The organic phase was evaporated in-vacuo and the residue purified by radial chromatography on silica eluting with 5-10% MeOH/DCM to provide the product as a gum (263 mg). MS m/z ([M+H]+) 621.8, ([2M+H]+) 1242.9.
  • Intermediate R
  • Intermediate Q (30 mg) and pyrrolidine (14 mg) in acetonitrile (0.5 mL) were heated in a microwave reactor at 70° C. for 10 mins (initial power 200 W). The solvent was evaporated in-vacuo and the residue dissolved in EtOAc, washed with water then 1M HCl. The organic layer was washed with water, dried (MgSO4) and evaporated in-vacuo to yield crude cyclised product (28 mg) used in the next step without further purification. The material was dissolved in MeOH (2 mL) and NaBH3CN (13 mg) was added. After 1 hr the solvent was evaporated in-vacuo and the residue dissolved in EtOAc. The organic layer was washed successively with saturated aqueous NaHCO3 and water, dried (MgSO4) and evaporated in-vacuo. The residue was purified by radial chromatography on silica eluting with 5% MeOH/DCM to yield the product as a gum (17 mg). MS m/z ([M+H]+) 677.2.
  • Intermediates S, T and U were prepared by the general method described for Intermediate R using the appropriate amine and substituting NaBH4 for NaBH3CN.
  • Intermediate S
  • Intermediate Q (40 mg) yielded Intermediate S (21 mg).
  • MS m/z ([M+H]+) 693.1.
  • Intermediate T
  • Intermediate Q (62 mg) yielded Intermediate T (42 mg).
  • MS m/z ([M+H]+) 691.2, ([2M+H]+) 1381.0
  • Intermediate U
  • Intermediate Q (62 mg) yielded Intermediate U (28 mg).
  • MS m/z ([M+H]+) 692.1.
  • Example 11
  • Intermediate R (10 mg) was stirred with 7M NH3 in MeOH (1 mL) at ambient temperature for 18 hrs. The reaction was evaporated to dryness in-vacuo and the residue dissolved in water and washed with DCM. The aqueous layer was then subjected directly to preparative HPLC. This process afforded the target compound as an oil (1.3 mg).
  • MS m/z ([M+H]+) 365.1, ([2M+H]+) 728.8.
  • 1H 1H NMR (CD3OD) δ 7.69 (d, 1H), 6.34 (d, 1H), 5.92 (s, 1H), 5.20 (d, 1H), 4.31 (d, 1H), 3.98-3.90 (m, 1H), 3.80-3.65 (m, 3H), 3.50-3.35 (m, 4H), 1.98-1.90 (m, 4H), 1.20 (s, 3H).
  • The general method for Example 11 was applied to the preparation of Examples 12-14.
  • Example 12
  • Intermediate S (20 mg) yielded compound 12 (2.4 mg) after purification by preparative HPLC.
  • MS m/z ([M+H]+) 381.1, ([2M+Na]+) 782.8.
  • 1H 1H NMR (CD3OD) δ 7.97 (d, 1H), 6.68 (d, 1H), 5.96 (s, 1H), 4.48 (d, 1H), 4.12 (d, 1H), 3.96-3.71 (m, 8H), 3.27-2.87 (m, 4H), 1.12 (s, 3H).
  • Example 13
  • Intermediate T (42 mg) yielded 4.27 mg compound 13 (4.3 mg) after purification by preparative HPLC.
  • MS m/z ([M+H]+) 379.2, ([2M+H]+) 756.9.
  • 1H 1H NMR (CD3OD) δ 7.93 (d, 1H), 6.65 (d, 1H), 5.97 (s, 1H), 4.42 (d, 1H), 4.07 (d, 1H), 3.93 (d, 1H), 3.82-3.71 (m, 3H), 3.04-2.87 (m, 4H), 1.86-1.60 (m, 6H), 1.13 (s, 3H).
  • Example 14
  • Intermediate U (27 mg) yielded compound 14 (2.1 mg) after purification by preparative HPLC.
  • MS m/z ([M+H]+) 380.1, ([2M+Na]+) 780.8.
  • 1H 1H NMR (CD3OD) δ 8.08 (d, 1H), 6.69 (d, 1H), 5.96, (s, 1H), 4.51 (d, 1H), 4.10 (d, 1H), 3.94-3.51 (m, 6H), 3.27-2.87 (m, 6H), 1.14 (s, 3H).
  • Example 15
  • Example 1 (31 mg) was dried overnight under high vacuum over P2O5 and then stirred with trimethyl phosphate (1.7 mL) at ambient temperature with oven-dried molecular sieves (4 Å) for 16 hrs under argon. The reaction was cooled to 0° C., POCl3 (31 μl) added and stirring continued for 2 hours before addition of Bu3N (72 μl) followed by acetonitrile (0.5 mL) and tributylammonium pyrophosphate (190 mg). After a further 2 hrs at 0° C. the reaction was quenched by pouring into ice-cold 1M triethylammonium bicarbonate buffer (10 mL, pH 8.5) and the aqueous layer washed with Et2O (3×10 mL). The aqueous material was then lyophilised to give a white solid which was purified by preparative HPLC to afford the product (1.67 mg).
  • MS m/z ([M−H]) 547.6, ([M+3TEA+H]+) 852.5.
  • 31P NMR (D2O) δ −8.14 (m, 1P), −10.69 (m, 1P), −21.65 (m, 1P).
  • Biological Data In Vitro Anti-HCV Dose-Response and Cytotoxicity HCV Replicon Assay
  • The antiviral activity of test compounds were assayed in the stable HCV RNA-replicating cell line, AVA5, derived by transfection of the human hepatoblastoma cell line, Huh7 (Blight, et al., 2000, Sci. 290:1972). Concentrations of compounds were added to dividing cultures once daily for three days and intracellular HCV RNA levels and cytotoxicity assessed 24 hours after the last dose of compound.
  • Intracellular HCV RNA levels were measured using standard blot hybridization techniques using triplicate cultures and levels of β-actin RNA were used to normalize HCV RNA levels in each sample. Cytotoxicity was measured using an established neutral red dye uptake assay (B. E. Korba and J. L. Gerin. 1992. Antivir. Res. 19, 55-70) and the 50% effective antiviral concentrations (EC50) and cytotoxic concentrations (CC50) were calculated using a computer program for curve fitting.
  • Examples such as 1-14 were typically active in the replicon assay in the range 1 to >1000 μM and cytotoxic in the range 30 to >100 μM.
  • HCV Polymerase Inhibition Assay
  • HCV 3′UTR RNA template was synthesized, gel purified and quantified by spectrophotometry. The kinetic constant, Km, was determined for the RNA template and for each GTP, CTP, ATP and UTP using a non-linear least square fit of initial rates as a function of substrate concentration assuming Michaelis-Menten kinetics.
  • Standard RdRp assays consisted of 30 nM RNA template and 25 nM HCV NS5bΔ21 (genotype 1b) (Replizyme Ltd) in a 50 L reaction mixture containing 20 mM Tris-HCl, 5 mM MgCl2, 3 mM DTT, 0.05% BSA, 22 nM GTP, 600 nM ATP, 15 nM CTP, 30 nM UTP and 3 nM [α-33P]GTP. Elongation reactions were initiated by the addition of NTPs and proceeded for 30 mins at 25° C. Reactions were quenched by the addition of 0.2 M EDTA and product formation was collected by filtration through Multiscreen plates (Millipore). Quantification of product formation was performed using TopCount (Perkin Elmer).
  • The inhibitor concentration at which the enzyme catalyzed rate is reduced by half (IC50) was determined using a computer program for curve fitting
  • Examples such as 15 were typically inhibitory of NS5b in the range 100 to >1000 μM.
  • The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge.
  • Throughout this specification and the claims which follow, unless the context requires otherwise, the word “comprise”, and variations such as “comprises” and “comprising”, will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.

Claims (54)

1. A method for the treatment of a microbial infection comprising administering an effective amount of a compound of the formula (I) which may be a D- or L-nucleoside or nucleotide, or a pharmaceutically acceptable salt thereof,
Figure US20100291031A2-20101118-C00051
wherein:
A is O, S, CH2, CHF, CF2 or NR;
R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, halogen, OH, N3, CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkynoxy, optionally substituted aryloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted arylalkoxy, optionally substituted heterocycloxy, optionally substituted heteroaryloxy, optionally substituted cycloalkoxy, optionally substituted cycloalkenoxy, optionally substituted amino, optionally substituted aminoacyl, optionally substituted aminoacyloxy, optionally substituted acylamino, optionally substituted oxyacylamino, optionally substituted oxyacyloxy, optionally substituted acylimino, optionally substituted acyliminoxy, optionally substituted oxyacylimino, optionally substituted aminothioacyl, optionally substituted thioacylamino, optionally substituted aminosulfinyl, optionally substituted aminosulfonyl, optionally substituted thio, optionally substituted thioacyl, optionally substituted thioacyloxy, optionally substituted oxythioacyl, optionally substituted oxythioacyloxy, optionally substituted phosphorylamino, optionally substituted sulfinyl, optionally substituted sulfonyl, optionally substituted sulfinylamino, optionally substituted sulfonylamino, optionally substituted oxysulfinylamino, and optionally substituted oxysulfonylamino, or R2 and R2′ together or R3 and R3′ together represents ═O, ═S, or =L-Y′ where L is N, CH, CF, CCl or CBr and Y′ is H, halogen, N3, methyl, ethyl or CN;
R4′ is —CY2SH, —CY2OH, —CY2NH2, or -L-R5;
L′ is selected from the group consisting of —CY2—, —CY2CY2—, —CY2OCY2—, —CY2SCY2— and —CY2NHCY2—;
each Y is independently selected from the group consisting of H, F, Cl, Br, OR, C1-C6alkyl, C2-C6alkenyl and C2-C6alkynyl, wherein C1-C6alkyl, C2-C6alkenyl, and C2-C6alkynyl may be optionally substituted with one or more groups selected from F, Cl, Br, OH, COOH, COOCH3, SH, SCH3, NH2, NHCH3, N(CH3)2, CN, NO2, C(O)NH2, C(O)NHCH3, N3, C(S)NH2, OCH3, and OCH2CH3;
R5 is selected from the group consisting of OR, NR2, monophosphate, diphosphate, and triphosphate, or a mono, di or triphosphate mimic;
each R is independently selected from the group consisting of H, CF3, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
B is selected from a group of formula (I)
Figure US20100291031A2-20101118-C00052
wherein, if Z is a participant in a π bond (double bond), Z is independently selected from N or C-G; or, if Z is not a participant in a π bond (double bond), Z is independently selected from O, S, Se, NR, NOR, NNR2, CO, CS, CNR, SO, S(O)2, SeO, Se(O)2 or C(G)2; each G is independently selected from the group consisting of H, halogen, OR, SR, NR2, NROR, N3, COOR, CN, CONR2, C(S)NR2, C(═NR)NR2, and R; and
where any two adjacent Z are not both selected from O, S, and Se, or not both selected from CO, CS, CNR, SO, S(O)2, SeO, and Se(O)2;
wherein, if X is a participant in a π bond (double bond), X is C; or if X is not a participant in a π bond (double bond), X is CR or N;
wherein, if R″ is a participant in a π bond (double bond), R″ is O, S, Se, NR, NOR or NNR2; or if R″ is not a participant in a π bond (double bond), R″ is OR, SR, F, Cl, R, or SeR; and
dashed lines (---) indicate a possible π or double bond;
optionally in combination with one or more antiviral, antibacterial, or antiproliferative agents.
2. A method according to claim 1 for the treatment of a microbial infection which is a viral infection.
3. A method according to claim 2 wherein the viral infection is caused by an RNA virus.
4. A method according to claim 3 wherein the viral infection is caused by an RNA virus of the group Flaviviridae.
5. A method according to claim 4 wherein the virus is the hepatitis C virus.
6. A method according to claim 1, wherein the compound of formula (I) has a base structure B selected form the following formulae (XI) to (XXI)
Figure US20100291031A2-20101118-C00053
Figure US20100291031A2-20101118-C00054
wherein Z, X and R″ are as defined in claim 1.
7. A method according to claim 6, wherein X is N.
8. A method according to claim 1, wherein the compound of formula (I) has a base structure B selected from the following:
Figure US20100291031A2-20101118-C00055
Figure US20100291031A2-20101118-C00056
Figure US20100291031A2-20101118-C00057
wherein G and R are as defined in claim 1.
9. A method according to claim 1, wherein the compound of formula (I) has a base structure B represented by formula IIa:
Figure US20100291031A2-20101118-C00058
wherein each Z′ is independently N (if a participant in a π bond) or NR (if not a participant in a π bond), and R″, R and Z are as defined in claim 1.
10. A method according to claim 1, wherein the compound of formula (I) has a base structure B represented by the formula IIb:
Figure US20100291031A2-20101118-C00059
wherein each Z′ is independently N (if a participant in a π bond) or NR (if not a participant in a π bond), and each Z is independently CG (if a participant in a π bond) or >C(G)2 (if not a participant in a π bond), wherein R″ and G are as defined in claim 1.
11. A method according to claim 1, wherein the compound of formula (I) has a base structure B represented by formula IIc:
Figure US20100291031A2-20101118-C00060
wherein R and G are as defined in claim 1.
12. A method according to claim 1, wherein the compound of formula (I) has a base structure B represented by the formula IId:
Figure US20100291031A2-20101118-C00061
wherein R and G are as defined in claim 1.
13. A method according to claim 1, wherein the compound of formula (I) has a base structure B represented by the formula IIe:
Figure US20100291031A2-20101118-C00062
wherein R and G are as defined in claim 1.
14. A method according to claim 1, wherein at least one of R2 and R2′ of the compound of formula (I) is methyl, hydroxyl or F.
15. A method according to claim 1, wherein L′ of the compound of formula (I) is —CH2—.
16. A method according to claim 1, wherein R4′ of the compound of formula (I) is selected from —CH2—OH, —CF2OH, —CCl2—OH, —C(CH3)(CH)3OH, —CH(CH3)OH, —CH2—CH2—P(O)(OH)2, —CH2—CH2—P(O)(OH)2, —CH2SP(O)(OH)2, —CH2SH, —CF2SH, and —CH2—O—P(O)(OPh)(NHCH(CH3)(CO2Me)).
17. A method according to claim 16, wherein R4′ is —CH2—OH.
18. A method according to claim 1, wherein the sugar moiety of the compound of formula (I) is selected from the following formulae:
Figure US20100291031A2-20101118-C00063
or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics, wherein R is as defined in claim 1.
19. A method according to claim 1, wherein the sugar moiety of the compound of formula (I) is selected from the following formulae:
Figure US20100291031A2-20101118-C00064
or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics, wherein R is as defined in claim 1.
20. A method according to claim 1, wherein the sugar moiety of the compound of formula (I) is represented by the formula:
Figure US20100291031A2-20101118-C00065
or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics, wherein R is as defined in claim 1.
21. A method according to claim 1, wherein the sugar moiety of the compound of formula (I) is represented by the formula:
Figure US20100291031A2-20101118-C00066
or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
22. A method according to claim 1, wherein the compound of formula (I) is selected from
Figure US20100291031A2-20101118-C00067
Figure US20100291031A2-20101118-C00068
Figure US20100291031A2-20101118-C00069
wherein:
each R on the sugar moiety is independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl and optionally substituted arylalkyl;
each R on the base moiety is independently selected from H, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
each G is independently selected from H, halogen, CF3, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl; and
C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
23. A method according to claim 1, wherein the compound of formula (I) is selected from
Figure US20100291031A2-20101118-C00070
Figure US20100291031A2-20101118-C00071
Figure US20100291031A2-20101118-C00072
24. A method according to claim 1 which includes the administration of a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with one or more antiviral agents.
25. A method according to claim 24, wherein the antiviral agents are selected from interferons and interferon derivatives, IMPDH inhibitors, antiviral nucleosides, polymerase inhibitors and protease inhibitors.
26-48. (canceled)
49. A compound of the formula (I) which may be a D- or L-nucleoside or nucleotide, or a salt thereof;
Figure US20100291031A2-20101118-C00073
wherein:
A is O, S, CH2, CHF, CF2 or NR;
R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, halogen, OH, N3, CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkynoxy, optionally substituted aryloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted arylalkoxy, optionally substituted heterocycloxy, optionally substituted heteroaryloxy, optionally substituted cycloalkoxy, optionally substituted cycloalkenoxy, optionally substituted amino, optionally substituted aminoacyl, optionally substituted aminoacyloxy, optionally substituted acylamino, optionally substituted oxyacylamino, optionally substituted oxyacyloxy, optionally substituted acylimino, optionally substituted acyliminoxy, optionally substituted oxyacylimino, optionally substituted aminothioacyl, optionally substituted thioacylamino, optionally substituted aminosulfinyl, optionally substituted aminosulfonyl, optionally substituted thio, optionally substituted thioacyl, optionally substituted thioacyloxy, optionally substituted oxythioacyl, optionally substituted oxythioacyloxy, optionally substituted phosphorylamino, optionally substituted sulfinyl, optionally substituted sulfonyl, optionally substituted sulfinylamino, optionally substituted sulfonylamino, optionally substituted oxysulfinylamino, and optionally substituted oxysulfonylamino, or R2 and R2′ together or R3 and R3′ together represents ═O, ═S, or =L-Y′ where L is N, CH, CF, CCl or CBr and Y′ is H, halogen, N3, methyl, ethyl or CN;
R4′ is —CY2SH, —CY2OH, —CY2NH2, or -L-R5;
L′ is selected from the group consisting of —CY2—, —CY2CY2—, —CY2OCY2—, —CY2SCY2— and —CY2NHCY2—;
each Y is independently selected from the group consisting of H, F, Cl, Br, OR, C1-C6alkyl, C2-C6alkenyl and C2-C6alkynyl, wherein C1-C6alkyl, C2-C6alkenyl, and C2-C6alkynyl may be optionally substituted with one or more groups selected from F, Cl, Br, OH, COOH, COOCH3, SH, SCH3, NH2, NHCH3, N(CH3)2, CN, NO2, C(O)NH2, C(O)NHCH3, N3, C(S)NH2, OCH3, and OCH2CH3;
R5 is selected from the group consisting of OR, NR2, monophosphate, diphosphate, and triphosphate, or a mono, di or triphosphate mimic;
each R is independently selected from the group consisting of H, CF3, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
B is a group of formula (II)
Figure US20100291031A2-20101118-C00074
wherein, if Z is a participant in a π bond (double bond), Z is independently selected from N or C-G; or, if Z is not a participant in a π bond (double bond), Z is independently selected from O, S, Se, NR, NOR, NNR2, CO, CS, CNR, SO, S(O)2, SeO, Se(O)2 or C(G)2, wherein each G is independently selected from the group consisting of H, halogen, OR, SR, NR2, NROR, N3, COOR, CN, CONR2, C(S)NR2, C(═NR)NR2, and R; and
where any two adjacent Z are not both selected from O, S, and Se, or not both selected from CO, CS, CNR, SO, S(O)2, SeO, and Se(O)2;
wherein, if X is a participant in a π bond (double bond), X is C; or if X is not a participant in a π bond (double bond), X is CR or N;
wherein, if R″ is a participant in a π bond (double bond), R″ is O, S, Se, NR, NOR, and NNR2; or if R″ is not a participant in a π bond (double bond), R″ is OR, SR, F, Cl, R, or SeR;
dashed lines (---) indicate a possible π or double bond; and
wherein when R2′, R3′, and R5 are OH or OC(O)CH3, L′ is CH2, A is O, and R1, R2, R3 and R4 are H, B is not the group of formula (III), formula (IV), formula (V), formula (VI), or formula (VII)
Figure US20100291031A2-20101118-C00075
(where each X* is H or one of X* is CH3 and the other two X* are H);
when B is a group of formula (III), formula (VIII), or formula (IX)
Figure US20100291031A2-20101118-C00076
R2 and R2′ are not both H;
when R2, R3′ and R5 are OH, L′ is CH2, A is O and R1, R2′, R3 and R4 are H, B is not the group of formula (IV); and
when R2′ is F, R3′ is OH, R5 is triphosphate, L′ is CH2, A is O, and R1, R2, R3 and R4 are H, B is not a group of formula (X)
Figure US20100291031A2-20101118-C00077
50. A compound according to claim 49, or a salt thereof, wherein the base structure B is selected from the following formulae (XI) to (XXI)
Figure US20100291031A2-20101118-C00078
Figure US20100291031A2-20101118-C00079
wherein Z, X and R″ are as defined in claim 49.
51. A compound according to claim 49, or a salt thereof, wherein X is N.
52. A compound according to claim 49, or a salt thereof, wherein the base structure B is selected from the following:
Figure US20100291031A2-20101118-C00080
Figure US20100291031A2-20101118-C00081
Figure US20100291031A2-20101118-C00082
wherein G and R are as defined in claim 49.
53. A compound according to claim 49, or a salt thereof, wherein the base structure B is represented by formula IIa:
Figure US20100291031A2-20101118-C00083
wherein each Z′ is independently N (if a participant in a π bond) or NR (if not a participant in a π bond), and R″, R and Z are as defined in claim 49.
54. A compound according to claim 49, or a salt thereof, wherein the base structure B is represented by the formula IIb:
Figure US20100291031A2-20101118-C00084
wherein each Z′ is independently N (if a participant in a π bond) or NR (if not a participant in a π bond), and each Z is independently CG (if a participant in a π bond) or >C(G)2 (if not a participant in a π bond), and wherein R″ and G is as defined in claim 49.
55. A compound according to claim 49, or a salt thereof, wherein the base structure B is represented by formula IIc:
Figure US20100291031A2-20101118-C00085
wherein R and G are as defined in claim 49.
56. A compound according to claim 49, or a salt thereof, wherein the base structure B is represented by the formula IId:
Figure US20100291031A2-20101118-C00086
wherein R and G are as defined in claim 49.
57. A compound according to claim 49, or a salt thereof, wherein the base structure B is represented by the formula IIe:
Figure US20100291031A2-20101118-C00087
wherein R and G are as defined in claim 49.
58. A compound according to claim 49, or a salt thereof, wherein R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, halogen, OH, N3, CN, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, optionally substituted arylalkyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted alkyloxy, optionally substituted acyloxy, optionally substituted oxyacyl, optionally substituted amino, optionally substituted aminoacyl, optionally substituted aminoacyloxy, optionally substituted acylamino, optionally substituted thio, or R2 and R2′ together or R3 and R3′ together represents ═O, ═S, or =L-Y′ where L is N, CH, CF, CCl or CBr and Y′ is H, halogen, N3, methyl, ethyl or CN.
59. A compound according to claim 49, or a salt thereof, wherein R1, R2, R2′, R3, R3′, and R4 are independently selected from the group consisting of H, F, Cl, Br, I, OH, SH, NH2, NHOH, NHNH2, N3, COOH, CN, CONH2, C(S)NH2, COOR, R, OR, SR, SSR, NHR, and NR2 wherein at least one of R2 or R2′ is a substituent other than H, and wherein R is selected from the group consisting of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl, and optionally substituted arylalkyl.
60. A compound according to claim 49, or a salt thereof, wherein at least one of R2 and R2′ is methyl, hydroxyl or F.
61. A compound according to claim 49, or a salt thereof, wherein L′ is —CH2—.
62. A compound according to claim 49, or salt thereof, wherein R4′ is selected from —CH2—OH, —CF2OH, —CCl2—OH, —C(CH3)(CH)3OH, —CH(CH3)OH, —CH2—CH2—P(O)(OH)2, —CH2—CH2—P(O)(OH)2, —CH2SP(O)(OH)2, —CH2SH, —CF2SH, and —CH2—O—P(O)(OPh)(NHCH(CH3)(CO2Me)).
63. A compound according to claim 62 or a salt thereof wherein R4′ is —CH2—OH.
64. A compound according to claim 49, or salt thereof, wherein the sugar moiety is selected from the following formulae:
Figure US20100291031A2-20101118-C00088
or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics, wherein R is as defined in claim 49.
65. A compound according to claim 49, or salt thereof, wherein the sugar moiety is selected from the following formulae:
Figure US20100291031A2-20101118-C00089
or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics, wherein R is as defined in claim 49.
66. A compound of formula (I) selected from the following formulae, or a salt thereof:
Figure US20100291031A2-20101118-C00090
Figure US20100291031A2-20101118-C00091
Figure US20100291031A2-20101118-C00092
wherein:
each R on the sugar moiety is independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted acyl and optionally substituted arylalkyl;
each R on the base moiety is independently selected from H, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl;
each G is independently selected from H, halogen, CF3, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, and optionally substituted arylalkyl; and
C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
67. A compound according to claim 49, or a salt thereof, wherein the sugar moiety is represented by the formula:
Figure US20100291031A2-20101118-C00093
or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics, wherein R is as defined in claim 49.
68. A compound according to claim 49, or a salt thereof, wherein the sugar moiety is represented by the formula:
Figure US20100291031A2-20101118-C00094
or C-5 monophosphate, diphosphate and triphosphate derivatives thereof, or C-5 mono, di or triphosphate mimics.
69. A compound according to claim 49, or a salt thereof, which is a B anomer.
70. A compound according to claim 49, or a salt thereof, selected from
Figure US20100291031A2-20101118-C00095
Figure US20100291031A2-20101118-C00096
Figure US20100291031A2-20101118-C00097
71. A pharmaceutical composition comprising a compound according to claim 49 or a salt thereof, and at least one pharmaceutically acceptable carrier or diluent.
72. A pharmaceutical composition according to claim 71 for use in the treatment of a viral infection.
73. A pharmaceutical composition according to claim 72, wherein the virus is the hepatitis C virus.
74. A pharmaceutical composition according to claim 71 further comprising one or more antiviral or antibacterial agents.
75. A pharmaceutical composition according to claim 74, wherein the antiviral agents are selected from the group selected from interferon and interferon derivatives, IMPDH inhibitors, antiviral nucleosides, polymerase inhibitors and protease inhibitors.
76. A pharmaceutical composition according to claim 75, wherein the composition comprises interferon or ribavirin.
US11/908,019 2005-03-08 2006-03-08 Bicyclic nucleosides and nucleotides as therapeutic agents Expired - Fee Related US8802840B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/908,019 US8802840B2 (en) 2005-03-08 2006-03-08 Bicyclic nucleosides and nucleotides as therapeutic agents

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US66166505P 2005-03-08 2005-03-08
US11/908,019 US8802840B2 (en) 2005-03-08 2006-03-08 Bicyclic nucleosides and nucleotides as therapeutic agents
PCT/AU2006/000303 WO2006094347A1 (en) 2005-03-08 2006-03-08 Bicyclic nucleosides and nucleotides as therapeutic agents

Publications (3)

Publication Number Publication Date
US20090004138A1 US20090004138A1 (en) 2009-01-01
US20100291031A2 true US20100291031A2 (en) 2010-11-18
US8802840B2 US8802840B2 (en) 2014-08-12

Family

ID=36952869

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/908,019 Expired - Fee Related US8802840B2 (en) 2005-03-08 2006-03-08 Bicyclic nucleosides and nucleotides as therapeutic agents

Country Status (6)

Country Link
US (1) US8802840B2 (en)
EP (1) EP1858889A1 (en)
JP (1) JP2008532950A (en)
AU (1) AU2006222563A1 (en)
CA (1) CA2600886A1 (en)
WO (1) WO2006094347A1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8853171B2 (en) 2008-04-23 2014-10-07 Gilead Sciences, Inc. 1′-substituted carba-nucleoside analogs for antiviral treatment
US9090642B2 (en) 2010-07-19 2015-07-28 Gilead Sciences, Inc. Methods for the preparation of diasteromerically pure phosphoramidate prodrugs
US9724360B2 (en) 2014-10-29 2017-08-08 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
US10065958B2 (en) 2010-07-22 2018-09-04 Gilead Sciences, Inc. Methods and compounds for treating Paramyxoviridae virus infections
US10251904B2 (en) 2015-09-16 2019-04-09 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
US10675296B2 (en) 2017-07-11 2020-06-09 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
US10682368B2 (en) 2017-03-14 2020-06-16 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
US10836787B2 (en) 2017-05-01 2020-11-17 Gilead Sciences, Inc. Crystalline forms of (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5- (4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy) phosphoryl)amino)propanoate
US10988498B2 (en) 2009-09-21 2021-04-27 Gilead Sciences, Inc. Processes and intermediates for the preparation of 1′-substituted carba-nucleoside analogs
US11491169B2 (en) 2020-05-29 2022-11-08 Gilead Sciences, Inc. Remdesivir treatment methods
US11613553B2 (en) 2020-03-12 2023-03-28 Gilead Sciences, Inc. Methods of preparing 1′-cyano nucleosides
US11660307B2 (en) 2020-01-27 2023-05-30 Gilead Sciences, Inc. Methods for treating SARS CoV-2 infections
US11701372B2 (en) 2020-04-06 2023-07-18 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carba-nucleoside analogs
US11780844B2 (en) 2022-03-02 2023-10-10 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11814406B2 (en) 2020-08-27 2023-11-14 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11939347B2 (en) 2020-06-24 2024-03-26 Gilead Sciences, Inc. 1′-cyano nucleoside analogs and uses thereof

Families Citing this family (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080261913A1 (en) 2006-12-28 2008-10-23 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of liver disorders
AU2008338273B2 (en) 2007-12-19 2014-10-02 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US8173621B2 (en) 2008-06-11 2012-05-08 Gilead Pharmasset Llc Nucleoside cyclicphosphates
PA8852101A1 (en) * 2008-12-08 2010-07-27 Medivir Ab URACIL CYCLOPROPYLL NUCLEOTIDES
CL2009002206A1 (en) 2008-12-23 2011-08-26 Gilead Pharmasset Llc Compounds derived from pyrrolo - (2-3-d] -pyrimidin-7 (6h) -tetrahydrofuran-2-yl phosphonamidate, pharmaceutical composition; and its use in the treatment of viral diseases.
SG172848A1 (en) 2009-01-07 2011-08-29 Scynexis Inc Cyclosporine derivative for use in the treatment of hcv and hiv infection
EA201200218A1 (en) * 2009-08-06 2012-09-28 Мерк Патент Гмбх NEW BICYCLIC UREA COMPOUNDS
US8563530B2 (en) 2010-03-31 2013-10-22 Gilead Pharmassel LLC Purine nucleoside phosphoramidate
CA2795054A1 (en) 2010-04-01 2011-10-06 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
BR112013005872A2 (en) 2010-09-22 2019-09-24 Alios Biopharma Inc compounds, pharmaceutical composition and their uses
ES2701020T3 (en) 2010-09-22 2019-02-20 Alios Biopharma Inc Azido nucleosides and nucleotide analogs
TW201242974A (en) 2010-11-30 2012-11-01 Gilead Pharmasset Llc Compounds
US9243025B2 (en) 2011-03-31 2016-01-26 Idenix Pharmaceuticals, Llc Compounds and pharmaceutical compositions for the treatment of viral infections
US9403863B2 (en) 2011-09-12 2016-08-02 Idenix Pharmaceuticals Llc Substituted carbonyloxymethylphosphoramidate compounds and pharmaceutical compositions for the treatment of viral infections
WO2013096680A1 (en) 2011-12-22 2013-06-27 Alios Biopharma, Inc. Substituted phosphorothioate nucleotide analogs
UA117095C2 (en) 2011-12-22 2018-06-25 Аліос Біофарма, Інк. Substituted nucleosides, nucleotides and analogs thereof
USRE48171E1 (en) 2012-03-21 2020-08-25 Janssen Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
US9441007B2 (en) 2012-03-21 2016-09-13 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
NZ631601A (en) 2012-03-21 2016-06-24 Alios Biopharma Inc Solid forms of a thiophosphoramidate nucleotide prodrug
US9012427B2 (en) 2012-03-22 2015-04-21 Alios Biopharma, Inc. Pharmaceutical combinations comprising a thionucleotide analog
EP2852604B1 (en) 2012-05-22 2017-04-12 Idenix Pharmaceuticals LLC 3',5'-cyclic phosphoramidate prodrugs for hcv infection
US9296778B2 (en) 2012-05-22 2016-03-29 Idenix Pharmaceuticals, Inc. 3′,5′-cyclic phosphate prodrugs for HCV infection
AU2013266393B2 (en) 2012-05-22 2017-09-28 Idenix Pharmaceuticals Llc D-amino acid compounds for liver disease
MX2014014323A (en) 2012-05-25 2015-02-12 Janssen R & D Ireland Uracyl spirooxetane nucleosides.
US9192621B2 (en) 2012-09-27 2015-11-24 Idenix Pharmaceuticals Llc Esters and malonates of SATE prodrugs
MD20150036A2 (en) 2012-10-08 2015-08-31 Idenix Pharmaceuticals, Inc. 2'-Chloro nucleoside analogs for HCV infection
EP2935304A1 (en) 2012-12-19 2015-10-28 IDENIX Pharmaceuticals, Inc. 4'-fluoro nucleosides for the treatment of hcv
DK2935303T3 (en) 2012-12-21 2021-05-03 Janssen Biopharma Inc 4`-fluoronucleosides, 4`-fluoronucleotides and their analogues for the treatment of HCV
UY35338A (en) 2013-02-21 2014-08-29 Bristol Myers Squibb Company Una Corporación Del Estado De Delaware BICYCLIC COMPOUNDS MODULATING THE ACTIVITY OF S1P1 AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
WO2014137930A1 (en) 2013-03-04 2014-09-12 Idenix Pharmaceuticals, Inc. Thiophosphate nucleosides for the treatment of hcv
WO2014137926A1 (en) 2013-03-04 2014-09-12 Idenix Pharmaceuticals, Inc. 3'-deoxy nucleosides for the treatment of hcv
WO2014160484A1 (en) 2013-03-13 2014-10-02 Idenix Pharmaceuticals, Inc. Amino acid phosphoramidate pronucleotides of 2'-cyano, azido and amino nucleosides for the treatment of hcv
US9187515B2 (en) 2013-04-01 2015-11-17 Idenix Pharmaceuticals Llc 2′,4′-fluoro nucleosides for the treatment of HCV
US10005779B2 (en) 2013-06-05 2018-06-26 Idenix Pharmaceuticals Llc 1′,4′-thio nucleosides for the treatment of HCV
US20150037282A1 (en) 2013-08-01 2015-02-05 Idenix Pharmaceuticals, Inc. D-amino acid phosphoramidate pronucleotides of halogeno pyrimidine compounds for liver disease
US9862743B2 (en) 2013-10-11 2018-01-09 Alios Biopharma, Inc. Substituted nucleosides, nucleotides and analogs thereof
WO2015161137A1 (en) 2014-04-16 2015-10-22 Idenix Pharmaceuticals, Inc. 3'-substituted methyl or alkynyl nucleosides for the treatment of hcv
TWI689487B (en) 2014-08-20 2020-04-01 美商必治妥美雅史谷比公司 Substituted bicyclic compounds
AU2016229966B2 (en) 2015-03-06 2018-09-27 Atea Pharmaceuticals, Inc. Beta-D-2'-deoxy-2'alpha-fluoro-2'-beta-C-substituted-2-modified-N6-substituted purine nucleotides for HCV treatment
US9902703B2 (en) 2015-07-01 2018-02-27 Crinetics Pharmaceuticals, Inc. Somatostatin modulators and uses thereof
US10202412B2 (en) 2016-07-08 2019-02-12 Atea Pharmaceuticals, Inc. β-D-2′-deoxy-2′-substituted-4′-substituted-2-substituted-N6-substituted-6-aminopurinenucleotides for the treatment of paramyxovirus and orthomyxovirus infections
US10711029B2 (en) 2016-07-14 2020-07-14 Atea Pharmaceuticals, Inc. Beta-d-2′-deoxy-2′-alpha-fluoro-2′-beta-c-substituted-4′fluoro-n6-substituted-6-amino-2-substituted purine nucleotides for the treatment of hepatitis c virus infection
MY197236A (en) 2016-09-07 2023-06-07 Atea Pharmaceuticals Inc 2'-substituted-n6-substituted purine nucleotides for rna virus treatment
EP3577124A4 (en) 2017-02-01 2021-01-06 ATEA Pharmaceuticals, Inc. Nucleotide hemi-sulfate salt for the treatment of hepatitis c virus
EP3658560A4 (en) 2017-07-25 2021-01-06 Crinetics Pharmaceuticals, Inc. Somatostatin modulators and uses thereof
EP3773753A4 (en) 2018-04-10 2021-12-22 ATEA Pharmaceuticals, Inc. Treatment of hcv infected patients with cirrhosis
CA3134613A1 (en) 2019-04-02 2020-10-08 Aligos Therapeutics, Inc. Compounds targeting prmt5
CA3164134A1 (en) 2019-12-06 2021-06-10 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
TWI794742B (en) 2020-02-18 2023-03-01 美商基利科學股份有限公司 Antiviral compounds
US10874687B1 (en) 2020-02-27 2020-12-29 Atea Pharmaceuticals, Inc. Highly active compounds against COVID-19
CN117120444A (en) 2021-04-16 2023-11-24 吉利德科学公司 Method for preparing carbanucleoside using amide
UY39800A (en) 2021-06-04 2023-01-31 Vertex Pharma N–(HYDROXYALKYL (HETERO)ARYL) TETRAHYDROFURAN CARBOXAMIDES AS SODIUM CHANNEL MODULATORS

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4965350A (en) * 1985-09-09 1990-10-23 Teijin Limited Pyridopyrimidine nucleotide compounds
US6063628A (en) * 1996-10-28 2000-05-16 University Of Washington Induction of viral mutation by incorporation of miscoding ribonucleoside analogs into viral RNA
US20030130226A1 (en) * 2001-11-07 2003-07-10 Medical Research Council Inhibition of viruses
US20030144502A1 (en) * 2000-07-06 2003-07-31 Zbigniew Pietrzkowski Pyrido[2,3-d]pyrimidine and pyrimido[4,5-d]pyrimidine nucleosides

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH01143895A (en) * 1987-11-30 1989-06-06 Meiji Seika Kaisha Ltd Pyrimidine derivative
GB9716231D0 (en) 1997-07-31 1997-10-08 Amersham Int Ltd Base analogues
US6348587B1 (en) 1998-02-25 2002-02-19 Emory University 2′-Fluoronucleosides
JP2004512810A (en) 1999-08-31 2004-04-30 サーナ・セラピューティクス・インコーポレイテッド Modulators of gene expression based on nucleic acids
WO2001032153A2 (en) 1999-11-04 2001-05-10 Shire Biochem Inc. Method for the treatment or prevention of flaviviridae viral infection using nucleoside analogues
EP1296690A2 (en) 2000-02-18 2003-04-02 Shire Biochem Inc. METHOD FOR THE TREATMENT OR PREVENTION OF i FLAVIVIRUS /i INFECTIONS USING NUCLEOSIDE ANALOGUES
CN100457118C (en) 2000-04-13 2009-02-04 法玛塞特有限公司 3'-or 2'-hydroxymethyl substd. nucleoside derivs. for treatment of hepatitis virus infections
MY164523A (en) 2000-05-23 2017-12-29 Univ Degli Studi Cagliari Methods and compositions for treating hepatitis c virus
EP1736478B1 (en) 2000-05-26 2015-07-22 IDENIX Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
US20030008841A1 (en) 2000-08-30 2003-01-09 Rene Devos Anti-HCV nucleoside derivatives
MY134070A (en) 2001-01-22 2007-11-30 Isis Pharmaceuticals Inc Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
WO2002062816A1 (en) 2001-02-07 2002-08-15 Celltech R & D Limited Non-natural nucleotides and dinucleotides
JP2005525358A (en) 2002-02-28 2005-08-25 ビオタ インコーポレーティッド Nucleotide mimetics and their prodrugs
WO2003073989A2 (en) 2002-02-28 2003-09-12 Biota, Inc. Nucleoside 5'-monophosphate mimics and their prodrugs
JP2004175708A (en) * 2002-11-26 2004-06-24 Geneticlab Co Ltd Bicyclic naphthyridine nucleoside
EP1575971A4 (en) 2002-12-23 2008-03-05 Idenix Cayman Ltd Process for the production of 3 -nucleoside prodrugs
JP2005015395A (en) * 2003-06-26 2005-01-20 Japan Science & Technology Agency New pyrimidopyrimidine nucleoside and its structural analog

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4965350A (en) * 1985-09-09 1990-10-23 Teijin Limited Pyridopyrimidine nucleotide compounds
US6063628A (en) * 1996-10-28 2000-05-16 University Of Washington Induction of viral mutation by incorporation of miscoding ribonucleoside analogs into viral RNA
US20030144502A1 (en) * 2000-07-06 2003-07-31 Zbigniew Pietrzkowski Pyrido[2,3-d]pyrimidine and pyrimido[4,5-d]pyrimidine nucleosides
US20030130226A1 (en) * 2001-11-07 2003-07-10 Medical Research Council Inhibition of viruses
US20050043268A1 (en) * 2001-11-07 2005-02-24 David Loakes Inhibition of viruses

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8853171B2 (en) 2008-04-23 2014-10-07 Gilead Sciences, Inc. 1′-substituted carba-nucleoside analogs for antiviral treatment
USRE46762E1 (en) 2008-04-23 2018-03-27 Gilead Sciences, Inc 1′-substituted carba-nucleoside analogs for antiviral treatment
US10988498B2 (en) 2009-09-21 2021-04-27 Gilead Sciences, Inc. Processes and intermediates for the preparation of 1′-substituted carba-nucleoside analogs
US9090642B2 (en) 2010-07-19 2015-07-28 Gilead Sciences, Inc. Methods for the preparation of diasteromerically pure phosphoramidate prodrugs
US9487544B2 (en) 2010-07-19 2016-11-08 Gilead Sciences, Inc. Methods for the preparation of diasteromerically pure phosphoramidate prodrugs
US11492353B2 (en) 2010-07-22 2022-11-08 Gilead Sciences, Inc. Methods and compounds for treating Paramyxoviridae virus infections
US10065958B2 (en) 2010-07-22 2018-09-04 Gilead Sciences, Inc. Methods and compounds for treating Paramyxoviridae virus infections
US10696679B2 (en) 2010-07-22 2020-06-30 Gilead Sciences, Inc. Methods and compounds for treating paramyxoviridae virus infections
US9949994B2 (en) 2014-10-29 2018-04-24 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
US11266666B2 (en) 2014-10-29 2022-03-08 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
US11344565B2 (en) 2014-10-29 2022-05-31 Gilead Sciences, Inc. Methods for the preparation of ribosides
US10251898B2 (en) 2014-10-29 2019-04-09 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
US9724360B2 (en) 2014-10-29 2017-08-08 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
US10695357B2 (en) 2014-10-29 2020-06-30 Gilead Sciences, Inc. Methods for treating filoviridae virus infections
US10251904B2 (en) 2015-09-16 2019-04-09 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
US11007208B2 (en) 2015-09-16 2021-05-18 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
US10695361B2 (en) 2015-09-16 2020-06-30 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
US11382926B2 (en) 2015-09-16 2022-07-12 Gilead Sciences, Inc. Methods for treating Arenaviridae and Coronaviridae virus infections
US11260070B2 (en) 2017-03-14 2022-03-01 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
US10682368B2 (en) 2017-03-14 2020-06-16 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
US10836787B2 (en) 2017-05-01 2020-11-17 Gilead Sciences, Inc. Crystalline forms of (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5- (4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy) phosphoryl)amino)propanoate
US11597742B2 (en) 2017-05-01 2023-03-07 Gilead Sciences, Inc. Crystalline forms of (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy) (phenoxy) phosphoryl)amino)propanoate
US10675296B2 (en) 2017-07-11 2020-06-09 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
US11975017B2 (en) 2017-07-11 2024-05-07 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
US11266681B2 (en) 2017-07-11 2022-03-08 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
US11660307B2 (en) 2020-01-27 2023-05-30 Gilead Sciences, Inc. Methods for treating SARS CoV-2 infections
US11613553B2 (en) 2020-03-12 2023-03-28 Gilead Sciences, Inc. Methods of preparing 1′-cyano nucleosides
US11701372B2 (en) 2020-04-06 2023-07-18 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carba-nucleoside analogs
US11903953B2 (en) 2020-05-29 2024-02-20 Gilead Sciences, Inc. Remdesivir treatment methods
US11491169B2 (en) 2020-05-29 2022-11-08 Gilead Sciences, Inc. Remdesivir treatment methods
US11975012B2 (en) 2020-05-29 2024-05-07 Gilead Sciences, Inc. Remdesivir treatment methods
US11939347B2 (en) 2020-06-24 2024-03-26 Gilead Sciences, Inc. 1′-cyano nucleoside analogs and uses thereof
US11814406B2 (en) 2020-08-27 2023-11-14 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11926645B2 (en) 2020-08-27 2024-03-12 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11845755B2 (en) 2022-03-02 2023-12-19 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11851438B2 (en) 2022-03-02 2023-12-26 Gilead Sciences, Inc. 1′-cyano nucleoside analogs and methods for treatment of viral infections
US11780844B2 (en) 2022-03-02 2023-10-10 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections

Also Published As

Publication number Publication date
EP1858889A1 (en) 2007-11-28
JP2008532950A (en) 2008-08-21
US20090004138A1 (en) 2009-01-01
US8802840B2 (en) 2014-08-12
AU2006222563A1 (en) 2006-09-14
WO2006094347A1 (en) 2006-09-14
CA2600886A1 (en) 2006-09-14

Similar Documents

Publication Publication Date Title
US8802840B2 (en) Bicyclic nucleosides and nucleotides as therapeutic agents
US7268119B2 (en) Tricyclic nucleosides or nucleotides as therapeutic agents
US8501699B2 (en) Bicyclic nucleosides and nucleotides as therapeutic agents
US9394331B2 (en) 2′-spiro-nucleosides and derivatives thereof useful for treating hepatitis C virus and dengue virus infections
KR101806314B1 (en) Compositions and methods for treating viral infections
CN101321775B (en) ODCASE inhibitors for the treatment of malaria
US20030176370A1 (en) Anti-viral pyrimidine nucleoside analogues
TW200524606A (en) Nucleoside compounds for treating viral infections
CZ20032005A3 (en) Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
HUT73659A (en) New antiviral nucleoside derivatives
EP2166016A1 (en) Phosphoramidate Derivatives of Nucleosides

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIOTA SCIENTIFIC MANAGEMENT PTY LTD, AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FRANCOM, PAULA;NEARN, ROLAND HENRY;DRAFFAN, ALISTAIR GEORGE;AND OTHERS;REEL/FRAME:020778/0922;SIGNING DATES FROM 20070927 TO 20071023

Owner name: BIOTA SCIENTIFIC MANAGEMENT PTY LTD, AUSTRALIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FRANCOM, PAULA;NEARN, ROLAND HENRY;DRAFFAN, ALISTAIR GEORGE;AND OTHERS;SIGNING DATES FROM 20070927 TO 20071023;REEL/FRAME:020778/0922

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.)

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20180812