WO2006091905A1 - Bicyclo (3.1.0) hexane derivatives as antiviral compounds - Google Patents

Bicyclo (3.1.0) hexane derivatives as antiviral compounds Download PDF

Info

Publication number
WO2006091905A1
WO2006091905A1 PCT/US2006/006763 US2006006763W WO2006091905A1 WO 2006091905 A1 WO2006091905 A1 WO 2006091905A1 US 2006006763 W US2006006763 W US 2006006763W WO 2006091905 A1 WO2006091905 A1 WO 2006091905A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
independently
aryl
halo
alkenyl
Prior art date
Application number
PCT/US2006/006763
Other languages
French (fr)
Inventor
Kenneth Alan Jacobson
Richard L Mackman
Bhalchandra V Joshi
Original Assignee
Gilead Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Publication of WO2006091905A1 publication Critical patent/WO2006091905A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/24Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one nitrogen and one sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/06Peri-condensed systems

Definitions

  • HCV Hepatitis C
  • the present invention provides compounds that inhibit one or more viral proteases. Accordingly, the compounds may be useful for treating viruses, such as HCV.
  • the invention provides a compound of the invention, which is a compound of formula I or II:
  • Ri is H, halo, OR a , SR a , or NR b R c ;
  • R 2 is H, halo, (C 1 -C 6 )alkyl , (C 2 -C 6 )alkenyl, or (C 2 -C 6 )alkynyl;
  • R 5 is H, halo, (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, or (C 2 -C 6 )alkynyl; and R 6 is
  • R 9 is a nucleoside base
  • the invention also provides a pharmaceutical composition comprising an effective amount of a compound of the invention, or a pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable excipient.
  • the invention also provides metabolites of compounds of the invention, as well as salts thereof.
  • the invention also provides a method of inhibiting a viral infection in an animal (e.g. a mammal), comprising administering an effective amount a compound of the invention, or a pharmaceutically acceptable salt or prodrug thereof, to the animal.
  • an animal e.g. a mammal
  • the invention also provides a compound of the invention for use in medical therapy (preferably for use in treating a viral infection in an animal), as well as the use of a compound of the invention for the manufacture of a medicament useful for the treatment of a viral infection in an animal (e.g. a mammal).
  • the invention also provides processes and novel intermediates disclosed herein which are useful for preparing compounds of the invention. Some of the compounds of the invention are useful to prepare other compounds of the invention.
  • the invention provides a method for inhibiting a viral infection in a sample comprising treating a sample suspected of containing a virus, with a compound of the invention, or a pharmaceutically acceptable salt or prodrug thereof.
  • Figures 1-4 illustrate synthetic processes and intermediates that can be used to prepare compounds of the invention.
  • Alkyl, alkoxy, alkenyl, alkynyl, etc. denote both straight and branched groups; but reference to an individual radical such as propyl embraces only the straight chain radical, a branched chain isomer such as isopropyl being specifically referred to.
  • Aryl means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Typical aiyl groups include, but are not limited to, radicals derived from benzene, substituted benzene, naphthalene, anthracene, biphenyl, and the like.
  • Carbocycle refers to a saturated, unsaturated or aromatic ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a polycycle.
  • Monocyclic carbocycles have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms.
  • Bicyclic carbocycles have 7 to 12 ring atoms, e.g., arranged as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as a bicyclo [5,6] or [6,6] system.
  • Examples of monocyclic carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1- cyclopent-1-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1- cyclohex-1-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, phenyl, spiryl and naphthyl.
  • Heterocycle as used herein includes, by way of example and not limitation, those heterocycles described in Paquette, Leo A.; Principles of Modem Heterocyclic Chemistry (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; The Chemistry of Heterocyclic Compounds, A Series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem, Soc. (1960) 82:5566.
  • heterocycle includes a “carbocycle” as defined herein, wherein one or more ⁇ e.g., 1, 2, 3, or 4) carbon atoms haVe been replaced with a heteroatom (e.g., O, N, or S).
  • a heteroatom e.g., O, N, or S
  • heterocycles include, by way of example and not limitation, pyridyl, dihydroypyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofuranyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolin
  • carbon bonded heterocycles are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline.
  • carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5- pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyI, 6-pyridazinyl, 2- pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3- pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl.
  • nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3 -imidazoline, pyrazole, pyrazolone, 2-pyrazoline, 3 -pyrazolone, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ -carboline.
  • nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, 1-pyrrolyl, 1-imidazolyl, 1- pyrazolyl, and 1-piperidinyl.
  • Heterocycle encompasses "heteroaryl,” which includes a radical attached via a ring carbon of a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(X) wherein X is absent or is H, O, (C 1 -C 6 )alkyl, phenyl or benzyl, as well as a radical of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms derived therefrom, particularly a benz-derivative or one derived by fusing a propylene, trimethylene, or tetramethylene diradical thereto.
  • phosphonate and “phosphonate group” include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double-bonded to a heteroatom, 3) single-bonded to a heteroatom, and 4) single-bonded to another heteroatom, wherein each heteroatom can be the same or different.
  • phosphonate and “phosphonate group” also include functional groups or moieties that comprise a phosphorous in the same oxidation state as the phosphorous described above, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having the characteristics described above.
  • the terms “phosphonate” and “phosphonate group” include phosphonic acid, phosphonic monoester, phosphonic diester, phosphonamidate, and phosphonthioate functional groups.
  • the terms “phosphonate” and “phosphonate group” include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double- bonded to an oxygen, 3) single-bonded to an oxygen, and 4) single-bonded to another oxygen, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having such characteristics.
  • the terms "phosphonate” and "phosphonate group” include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double-bonded to an oxygen, 3) single-bonded to an oxygen or nitrogen, and 4) single-bonded to another oxygen or nitrogen, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having such characteriatics.
  • treatment or “treating,” to the extent it relates to a disease or condition includes preventing the disease or condition from occurring, inhibiting the disease or condition, eliminating the disease or condition, and/or relieving one or more symptoms of the disease or condition.
  • prodrug refers to any compound that when administered to a biological system generates the drug substance, i.e. active ingredient, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), photolysis, and/or metabolic chemical reaction(s).
  • a prodrug is thus a covalently modified analog or latent form of a therapeutically- active compound.
  • Prodrug moiety refers to a labile functional group which separates from the active inhibitory compound during metabolism, systemically, inside a cell, by hydrolysis, enzymatic cleavage, or by some other process (Bundgaard, Hans, “Design and Application of Prodrugs” in A Textbook of Drug Design and Development (1991), P. Krogsgaard-Larsen and H. Bundgaard, Eds. Harwood Academic Publishers, pp. 113-191).
  • Enzymes which are capable of an enzymatic activation mechanism with a phosphate prodrug of the invention include, but are not limited to, amidases, esterases, microbial enzymes, phospholipases, cholinesterases, and phosphases.
  • Prodrug moieties can serve to modulate (e.g. enhanse) solubility, absorption, lipophilicity, drug delivery, bioavailability, and efficacy.
  • R x is (C 1 -C 6 )alkyl, (C 1 -C 6 )substituted alkyl, C 6 -C 20 aryl or C 6 -C 2O substituted aryl.
  • the acyloxyalkyl ester was first used as a prodrug strategy for carboxylic acids and then applied to phosphates and phosphonates by Farquhar et al. (1983) J. Pharm. ScI 72: 324; also US Patent Nos. 4816570, 4968788, 5663159 and 5792756.
  • acyloxyalkyl ester was used to deliver phosphonic acids across cell membranes and to enhance oral bioavailability.
  • a close variant of the acyloxyalkyl ester, the alkoxycarbonyloxyalkyl ester (carbonate), may also enhance oral bioavailability of a compound of the invention.
  • Aryl esters of phosphorus groups are reported to enhance oral bioavailability (De Lombaert et al. (1994) J Med. Chein. 37: 498). Phenyl esters containing a carboxylic ester ortho to the phosphate have also been described (Khamnei and Torrence, (1996) J Med. Chem. 39:4109- 4115). Benzyl esters are reported to generate the parent phosphonic acid. In some cases, substituents at the ortho-ox jtr ⁇ r ⁇ -position may accelerate the hydrolysis.
  • Benzyl analogs with an acylated phenol or an alkylated phenol may generate the phenolic compound through the action of enzymes, e.g., esterases, oxidases, etc., which in turn undergoes cleavage at the benzylic C-O bond to generate the phosphoric acid and a quinone methide compound.
  • enzymes e.g., esterases, oxidases, etc.
  • Thio-containing prodrugs are reported to be useful for the intracellular delivery of phosphorous containing drugs.
  • Certain of these proesters contain an ethylthio group in which the thiol group is either esterified with an acyl group or combined with another thiol group to form a disulfide. Deesterification or reduction of the disulfide generates the free thio intermediate, which subsequently breaks down to the phosphoric acid and an episulfide (Puech et al. (1993) Antiviral Res., 22: 155-174; Benzaria et al. (1996) J. Med. Chem. 39: 4958).
  • Prodrug moities that can be incorporated into compounds of the invention include those described by Mackman, R.L. and Cihlar, Tomas, Annual Reports in Medicinal Chemistry, 2004, 39, 305-321.
  • Other prodrug moities that can be incorporated into compounds of the invention include those described by Peyrottes et. al., Minireviews in Medicinal Chemistry, 2004, 4(4), 395-408; and Meier, Chris, Advances in Antiviral Drug Design, 2004, 4, 147-213.
  • protecting groups include prodrug moieties and chemical protecting groups.
  • Protecting group refers to a moiety of a compound that masks or alters the properties of a functional group or the properties of the compound as a whole.
  • Chemical protecting groups and strategies for protection/deprotection are well known in the art. See e.g., Protective Groups in Organic Chemistry, Theodora W. Greene, John Wiley & Sons, Inc., New York, 1991. Protecting groups are often utilized to mask the reactivity of certain functional groups and to assist in the efficiency of desired chemical reactions, e.g., making and breaking chemical bonds in an ordered and planned fashion.
  • Protection of functional groups of a compound alters other physical properties besides the reactivity of the protected functional group, such as the polarity, lipophilicity (hydrophobicity), and other properties that can be measured by common analytical tools.
  • Chemically protected intermediates may themselves be biologically active or inactive.
  • Protected compounds may also exhibit altered, and in some cases, optimized properties in vitro and in vivo, such as passage through cellular membranes and resistance to enzymatic degradation or sequestration. In this role, protected compounds with intended therapeutic effects are also prodrugs.
  • protecting groups for -OH groups include "ether- or ester- forming groups”.
  • Ether- or ester-forming groups are capable of functioning as chemical protecting groups in the synthetic schemes set forth herein.
  • (C 1 -C 6 )alkyl can be methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl, pentyl, 3-pentyl, or hexyl;
  • (C 3 -C 6 )cycloalkyl can be cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;
  • (C 3 -C 6 )cycloalkyl(C 1 - C 6 )alkyl can be cyclopropylmethyl, cyclobutylmethyl, cyclopeiitylmethyl, cyclohexylmethyl, 2-cyclopropylethyl, 2-cyclobutylethyl, 2-cyclopentylethy
  • -C 6 )alkoxycarbonyl can be methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl, pentoxycarbonyl, or hexyloxycarbonyl;
  • (C 1 -C 6 )alkylthio can be methylthio, ethylthio, propylthio, isopropylthio, butylthio, isobutylthio, pentylthio, or hexylthio;
  • C 2 -C 6 )alkanoyloxy can be acetoxy, propanoyloxy, butanoyloxy, isobutanoyloxy, pentanoyloxy, or hexanoyloxy;
  • aryl can be phenyl, indenyl, or naphthyl; and heteroaryl can be furyl, imidazolyl, triazolyl, triaziny
  • a specific value for R 2 is H.
  • a specific value for R 3 is H.
  • a specific value for R 4 is H or OH.
  • a specific value for R 5 is H.
  • a specific value for R 6 is OH.
  • a specific value for each of R 7 and R 5 is H.
  • a specific value for R 10 is OH.
  • a specific value for R 10 is a phosphonate.
  • R 10 has the formula:
  • Y 2b is O or NH; each R 1 is independently H, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 - C 6 )alkynyl, aryl, (C 1 -C 6 )alkanoyl, or aryl(C 1 -C 6 )alkyl; and each R 2 , is independently (C 1 -C 6 )alkyl, (C 1 -C 6 )alkylthio, (C 1 -C 6 )alkoxy, (C 1 - C 6 )alkanoyl, (C 1 -C 6 )alkanoyloxy, (C 1 -C 6 )alkoxycarbonyl, cyano, halo, hydroxy, nitro, carboxy, (C 3 -C 6 )cycloalkyl, (C 3 -C 6 )cycloalkoxy, trifluoromethoxy, mercapto, or trifluor
  • R 10 has the formula:
  • R 1 Q A specific value for R 1 Q has the formula:
  • R 2 is (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, or (C 2 -C 6 )alkynyl.
  • a specific value for Rio has the formula: wherein: Y 2b is O or N(R 4 ); R 2 is (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, aryl(C 1 -C 6 )alkyl, or (C 2 -C 6 )alkynyl; and R 4 is (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, aryl(C 1 -C 6 )alkyl, or (C 2 -C 6 )alkynyl.
  • the compound of formula I is not (l'5',2 l i?,3 1 i S r ,4 l i?,5'5)-4-(6-amino-purin-9-yl)-1-[hydroxymethyl]bi- cyclo[3.1.0]hexane-2,3-diol.
  • the compound of formula I is not 4-(6-amino-purin-9-yl)-1-[hydroxymethyl]bi- cyclo[3.1.0]hexane-2,3-diol.
  • the compound of formula I is not: (l'S r ,2'i?,3'5,4'i?,5' ⁇ S)-4-(6-amino-9H-purin-9-yl)-1-
  • the compounds of formula I exclude compounds wherein: Ri is H; R 2 is H; R 3 is H; R 4 is OH; R 5 is H; Re is OH; R 7 and Rg are each H; R9 is 6-amino-9H-purin-9-yl or
  • Nucleoside base The invention provides compounds of formula I and II wherein R 9 is a nucleoside base. A multitude of nucleoside base analogs have been reported. The compounds of formula I and II may include any nucleoside base, provided the final compound possesses useful therapeutic (e.g. anti-viral) properties. For example, a specific value for R9 is a heterocycle or a substituted heterocycle (e.g. a purine, substituted purine, pyrimidine, or a substituted pyrimidine).
  • a specific value for R9 is a heterocycle or a substituted heterocycle (e.g. a purine, substituted purine, pyrimidine, or a substituted pyrimidine).
  • nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in United States Patent Application Publication Number 2004/0147464. Accordingly, a specific value for R 9 has the following formula:
  • R 2 o is OH, methoxy, or propoxy; and for R 2 i is H.
  • Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in United States Patent Application Publication Number 2005/0215511. Accordingly, a specific value for R 9 has the following formula:
  • R 3 i is tri(C 1 -C 6 )alkylsilyl, (C 1 -C 6 )alkoxy(C 1 -C 6 )alkyl, heteroaryl, aryl,
  • R 32 is hydrogen or czs-(C 1 -C 6 )alkoxy
  • R 33 is H or (C 1 -C 6 )alkyl
  • R 35 and R 36 are each independently H, (C 1 -C 6 )alkyl or (C 1 -C 6 )alkanoyl;
  • R 37 is (C 1 -C 6 )alkyl, aryl, hetrocycle, or NR 3 gR 39 ;
  • nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 03/061385. Accordingly, a specific value for R9 has one of the following formulae:
  • R 4c , and R 4d are each independently hydrogen, (C 1 -C 6 )alkyl, (C 2 - C 6 )alkenyl, (C 2 -C 6 )alkynyl, heterocycle, or aryl;
  • X, Y, and W are each independently N, C, CR 4c , S or P;
  • R 46 , and R 47 together with the atoms to which they are attached form a heterocyclic ring
  • U is S or O; wherein each aryl or heterocycle is optionally substituted with one or more (C 1 -C 6 )alkyl, (C 1 -C 6 )alkylthio, (C 1 -C 6 )alkoxy, (C 1 -C 6 )alkanoyl, (C 1 - C 6 )alkanoyloxy, (C 1 -C 6 )alkoxycarbonyl, cyano, halo, hydroxy, nitro, carboxy, (C 3 -C 8 )cycloalkyl, (C 3 -C 6 )cycloalkoxy, trifluoromethoxy, mercapto, or trifluoromethyl.
  • each aryl or heterocycle is optionally substituted with one or more (C 1 -C 6 )alkyl, (C 1 -C 6 )alkylthio, (C 1 -C 6 )alkoxy, (C 1 -C 6 )alkanoyl, (C
  • nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 03/062257. Accordingly, a specific value for R 9 has one of the following formulae:
  • R 50 is NR 5a R 5b , ONR 5a R 5b , NR 5a NR 5a R 5b , SR 5b , OR 5b , H, hydroxy, (Cr C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 -C 6 )all ⁇ ynyl, or aryl;
  • R 5 is (C,-C 6 )alkyl, (C 1 -C 6 )alkanoyl, or aryl;
  • R 52 is (C 1 -C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 -C 6 )alkynyl, or aryl;
  • R 53 is H, halo, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 -C 6 )a]kynyl, or aryl;
  • R 54 is H or NH 2 ;
  • R 55 is NR 5a R 5b , ONR 5a R 5b , NR 5a NR 5a R 5b , SR 5b , OR 5b , H, hydroxy, (C 1 - C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 -C 6 )alkynyl, or aryl;
  • R 56 is H, halo, (C 1 -C 6 )alkyl, or (C 1 -C 6 )alkenyl;
  • R 57 and R 5 g are each independently -L-R 5c ; each L is independently a direct bond, -N(R 5J1 )-, O or S; each R 5a and R 5b is independently H, hydroxy, (C 1 -C 6 )alkyl, (C 1 - C 6 )alkenyl, (C !
  • each R 5c is NR 5a R 5b , H, hydroxy, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 - C 6 )alkynyl, or aryl; wherein each (C 1 -C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 -C 6 )alkynyl, or aryl of R 5 o-Rsg and Rs a -Rs c is optionally substituted with one or more (C 1 -C 6 )alkyl, (C 1 - C 6 )alkylthio, (C 1 -C 6 )alkoxy, (C 1 -C 6 )alkanoyl, (C 1 -C 6 )alkanoyloxy, (C 1 - C 6 )alkoxycarbonyl, cyano, hal
  • nucleoside base sti-uctures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 03/072757. Accordingly, a specific value for R 9 has one of the following formulae:
  • X 2, X 3; and X 4 are each independently N, CH, or C-R6 a ;
  • R 6b , and R 6c are each independently H, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 - C 6 )alkynyl, aryl, (C 1 -C 6 )alkanoyl, or aryl(C 1 -C 6 )alkyl.
  • Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 03/073989. Accordingly, a specific value for R 9 has the following formula:
  • X 5 , X 6 , and X 7 are each independently N, CH, or C-R 7a ;
  • R 7b , and R 70 are each independently H, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 - C 6 )alkynyl, aryl, (C 1 -C 6 )alkanoyl, or aryl(C 1 -C 6 )alkyl.
  • Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 2004/080466. Accordingly, a specific value for R 9 has the following formula:
  • nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in
  • R 9 has one of the following formulae:
  • R 9a , and R 9d are each independently H, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkenyl, (C 1 - C 6 )alkynyl, aryl, (C 1 -C 6 )alkanoyl, or aryl(C 1 -C 6 )alkyl.
  • R 9 Another specific value for R 9 has one of the following formulae:
  • nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 2005/123087. Accordingly, a specific value for R 9 has one of the following formulae:
  • X 4 is CRgo & or N
  • X 5 is O, S, orNR 9 i a ;
  • Rp 0 and R9 1 are each independently H, halo, hydroxy, (C 1 -C 6 )alkoxy,
  • R 9 o a is H, halo, methyl, azido, or amino
  • R 91a is H, or (C 1 -C 6 )alkyl
  • R 9Ob and R 91b are each independently H, (C 1 -Ce ⁇ IlCyI, (C 3 -C8)cycloalkyl, or aryl(C 1 -C 6 )alkyl; wherein each (C 1 -C 6 )alkyl, (C 3 -Cs)cycloalkyl, aryl(C 1 -C 6 )alkyl, and heterocycle of R9 0 -R9 1 , R 9U , and R 9 o b -R9ib are optionally substituted with one or more (e.g. 1, 2, 3, or 4) halo, hydroxy, amino, (C 1 -C 6 )alkyl, and (C 1 -C 6 )alkoxy.
  • R 9Ob and R 91b are each independently H, (C 1 -Ce ⁇ IlCyI, (C 3 -C8)cycloalkyl, or aryl(C 1 -C 6 )alkyl; where
  • nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 2006/002231. Accordingly, a specific value for R 9 is a 9-deazapurine derivative, such as, for example, a group of the following formulae:
  • X 6 is O, S, orNR 70a ;
  • R 7O a is H, methyl, ethyl, or acetyl
  • nucleoside base structoes that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 2006/000922. Accordingly, a specific value for R 9 a specific value for R 9 has one of the following formulae:
  • Z 80 is O, S, NR 80 , or CR 80 R 8 I; each V is independently N or CR 80 ; and each R 80 and Rsi is independently H, carbocycle, (C 1 -C 6 )alkyl, (C 1 -
  • R 9 Another specific value for R 9 has one of the following formulae:
  • R9 7 is H, hydroxy, mercapto, (C 1 -C 6 )alkyl, (C 1 -C 6 )aIkoxy, (C 2 - C 6 )alkenyl, (C 3 -C 8 )cycloalkyl, aryl(C 1 -C 6 )alkyl, aryl or (C 2 -C 6 )alkynyl;
  • R 98 is H, hydroxy, mercapto, or (C 1 -C 6 )alkyl;
  • R 99 is H, halo, azido, cyano, nitro, O R 99a , SR 99a , NR 99b R 99c , (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 3 -C 8 )cycloaikyl, aryl(C 1 -C 6 )alkyl, aryl or (C 2 -C 6 )alkyny
  • R 9 Another specific value for R 9 is adenine, guanine, cytosine, uracil, thymine, 7-deazaadenine, 7-deazaguanine, 7-deaza-8-azaguanine, 7-deaza-8- azaadenine, inosine, nebularine, nitropyrrole, nitroindole, 2-aminopurine, 2- amino-6-chloropurine, 2,6-diaminopurine, hypoxanthine, pseudouridine, pseudocytosine, pseudoisocytosine, 5-propynylcytosine, isocytosine, isoguanine, 7-deazaguanine, 2-thiopyrimidine, 6-thioguanine, 4-thiothymine, 4-thiouracil, O 6 -methylguanine, N 6 -methyladenine, O 4 -methylthymine, 5,6-dihydrothymine, 5,6-dihydrouracil, 4-
  • Rg is adenine or cytosine, which adenine or cytosine is optionally substituted with one or more (C 1 -C 6 )all ⁇ yl, (Cr C 6 )alkylthio, (C 1 -C 6 )alkoxy, (C 1 -C 6 )alkanoyl, (C 1 -C 6 )alkanoyloxy, (C 1 - C 6 )alkoxycarbonyl, NH 2 , cyano, halo, hydroxy, nitro, cavboxy, trifluoromethoxy, aryl, or mercapto.
  • the compounds of the invention may have chiral centers, e.g., chiral carbon or phosphorus atoms.
  • the compounds of the invention thus include racemic mixtures of all stereoisomers, including enantiomers, diastereomers, and atropisomers.
  • the compounds of the invention include enriched or resolved optical isomers at any or all asymmetric, chiral atoms.
  • the chiral centers apparent from the depictions are provided as the chiral isomers or racemic mixtures.
  • a single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents ( Stereochemistry of Carbon Compounds, (1962) by E. L. Eliel, McGraw Hill; Lochmuller, C. H., (1975) J.
  • Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions.
  • diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, ⁇ -methyl- ⁇ -phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid.
  • the diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography.
  • addition of chiral carboxylic or sulfonic acids such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts.
  • the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair
  • a diastereomeric pair Eliel, E. and WiI en, S. (1994) Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., p. 322).
  • Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yleld the free, enantiomerically enriched xanthene.
  • a method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, ⁇ -methoxy- ⁇ - (trifluoromethyl)phenyl acetate (Jacob III. (1982) J. Org. Chem. 47:4165), of the racemic mixture, and analyzing the NMR spectrum for the presence of the two atropisomeric diastereomers.
  • chiral esters such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, ⁇ -methoxy- ⁇ - (trifluoromethyl)phenyl acetate (Jacob III. (1982) J. Org. Chem. 47:4165), of the racemic mixture, and analyzing the NMR spectrum for the presence of the two atropisomeric diastereomers.
  • Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (Hoye, T., WO 96/15111).
  • a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase (Chiral Liquid Chromatography (1989) W. J. Lough, Ed. Chapman and Hall, New York; Okamoto, (199O) J. ofChromatogr. 513:375-378).
  • Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
  • the compounds of the invention can also exist as tautomeric isomers in certain cases. Although only one delocalized resonance structure may be depicted, all such forms are contemplated within the scope of the invention.
  • ene-amine tautomers can exist for purine, pyrimidine, imidazole, guanidine, amidine, and tetrazole systems and all their possible tautomeric forms are within the scope of the invention.
  • compositions of this invention optionally comprise salts of the compounds herein, especially pharmaceutically acceptable non-toxic salts containing, for example, Na + , Li + , K + , Ca +2 and Mg +2 .
  • Such salts may include those derived by combination of appropriate cations such as alkali and alkaline earth metal ions or aminonium and quaternary amino ions with an acid anion moiety, typically a carboxylic acid.
  • Monovalent salts maybe preferred if a water soluble salt is desired.
  • Metal salts typically are prepared by reacting the metal hydroxide with a compound of this invention.
  • metal salts which are prepared in this way are salts containing Li + , Na + , and K + .
  • a less soluble metal salt can be precipitated from the solution of a more soluble salt by addition of the suitable metal compound.
  • compositions herein comprise compounds of the invention in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.
  • salts of the parental compounds with one or more amino acids are also included within the scope of this invention.
  • Any amino acid can be used, especially the naturally-occurring amino acids found as protein components, although the amino acid typically is one bearing a side chain with a basic or acidic group, e.g., lysine, arginine or glutamic acid, or a neutral group such as glycine, serine, threonine, alanine, isoleucine, or leucine.
  • salts of active ingredients of the compounds of the invention will typically be physiologically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base.
  • salts of acids or bases which are not physiologically acceptable may also find use, for example, in the preparation or purification of a physiologically acceptable compound. All salts, whether or not derived form a physiologically acceptable acid or base, are within the scope of the present invention.
  • compositions according to the present invention comprise one or more compounds of the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents.
  • Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration.
  • tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
  • excipients which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc.
  • inert diluents such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate
  • granulating and disintegrating agents such as maize starch, or alginic acid
  • binding agents such as cellulose, microcrystalline cellulose, starch, gelatin or acacia
  • lubricating agents such as magnesium
  • Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate).
  • a suspending agent
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.
  • These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium tartrate
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these.
  • Suitable emulsifylng agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • the pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may likewise be used in the preparation of injectables.
  • a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weightweight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ⁇ g of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • Formulations suitable for administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient.
  • the active ingredient is preferably present in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about 1.5% w/w.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs.
  • Suitable formulations include aqueous or oily solutions of the active ingredient.
  • Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of viral infections as described below.
  • Formulations 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.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use.
  • sterile liquid carrier for example water for injection
  • Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
  • Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
  • formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
  • the invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefor.
  • Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered orally, parenterally or by any other desired route.
  • Compounds of the invention can also be formulated to provide controlled release of the active ingredient(s) to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile of the active ingredient(s). Accordingly, the invention also provided compositions comprising one or more compounds of the invention formulated for sustained or controlled release.
  • an effective dose of active ingredient depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses) or against an active viral infection, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. It can be expected to be from about 0.0001 to about 100 mg/kg body weight per day. Typically, from about 0.01 to about 10 mg/kg body weight per day. More typically, from about .01 to about 5 mg/kg body weight per day. More typically, from about .05 to about 0.5 mg/kg body weight per day.
  • the daily candidate dose for an adult human of approximately 70 kg body weight will range from 1 mg to 1000 mg, preferably between 5 mg and 500 mg, and may take the form of single or multiple doses.
  • One or more compounds of the invention are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient.
  • An advantage of the compounds of this invention is that they are orally bioavailable and can be dosed orally.
  • Active ingredients of the invention can also be used in combination with other active ingredients. Such combinations are selected based on the condition to be treated, cross-reactivities of ingredients and pharmaco-properties of the combination. For example, when treating a viral infection the compositions of the invention can be combined with other agents that are effective to treat a viral infection (such as other antiviral agents).
  • any compound of the invention with one or more other active ingredients in a unitary dosage form for simultaneous or sequential administration to a patient.
  • the combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.
  • the combination therapy may provide "synergy” and "synergistic effect", i.e. the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • a synergistic effect may be attained when the active ingredients are: (1) co- formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate fo ⁇ nulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., in separate tablets, pills or capsules, or by different injections in separate syringes.
  • an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
  • the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yleld a metabolic product thereof.
  • Such products typically are identified by preparing a radiolabeled ⁇ e.g., C ⁇ or H ⁇ ) compound of the invention, administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples.
  • a detectable dose e.g., greater than about 0.5 mg/kg
  • an animal such as rat, mouse, guinea pig, monkey, or to man
  • sufficient time for metabolism to occur typically about 30 seconds to 30 hours
  • isolating its conversion products from the urine, blood or other biological samples typically isolating its conversion products from the urine, blood or other biological samples.
  • the metabolite structures are determined in conventional fashion, e.g., by MS or NMR analysis.
  • Another aspect of the invention relates to methods of inhibiting viral infections, comprising the step of treating a sample or subject suspected of needing such inhibition with a composition of the invention.
  • samples suspected of containing a virus include natural or man-made materials such as living organisms; tissue or cell cultures; biological samples such as biological material samples (blood, serum, urine, cerebrospinal fluid, tears, sputum, saliva, tissue samples, and the like); laboratory samples; food, water, or air samples; bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like.
  • biological material samples blood, serum, urine, cerebrospinal fluid, tears, sputum, saliva, tissue samples, and the like
  • laboratory samples food, water, or air samples
  • bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like.
  • samples will be suspected of containing an organism which induces a viral infection, frequently a pathogenic organism such as an tumor virus.
  • Samples can be contained in any medium including water and organic solventVwater mixtures. Samples include living organisms such as humans, and man made materials such as cell cultures.
  • the anti-virus activity of a compound of the invention after application of the composition can be observed by any method including direct and indirect methods of detecting such activity. Quantitative, qualitative, and semiquantitative methods of determining such activity are all contemplated. Typically one of the screening methods described above are applied, however, any other method such as observation of the physiological properties of a living organism are also applicable.
  • the antiviral activity of a compound of the invention can be measured using standard screening protocols that are known.
  • the antiviral activity of a compound can be measured using the following general protocol. HCV IC 5O Determination
  • NS5b polymerase assay (40 ⁇ L) was assembled by adding 28 ⁇ L polymerase mixture (final concentration: 50 mM Tris-HCl at pH 7.5, 10 mM KCL, 5 mM MgCl 2 , 1 mM DTT, 10 mM EDTA, 4 ng/ ⁇ L of RNA template, and 75 nM HCV ⁇ 21 NS5b polymerase) to assay plates followed by 4 ⁇ L of compound dilution.
  • polymerase mixture final concentration: 50 mM Tris-HCl at pH 7.5, 10 mM KCL, 5 mM MgCl 2 , 1 mM DTT, 10 mM EDTA, 4 ng/ ⁇ L of RNA template, and 75 nM HCV ⁇ 21 NS5b polymerase
  • the polymerase and compound were pre-incubated at 35 0 C for 10 minute before the addition of 8 ⁇ L of nucleotide substrate mixture (33P- ⁇ -labeled competing nucleotide at K M and 0.5 mM of the remaining three nucleotides).
  • the assay plates were covered and incubated at 35 0 C for 90 min. Reactions were then filtered through 96-well DEAE-81 filter plates via vacuum. The filter plates were then washed under vacuum with multiple volumes of 0.125 MNaHPO 4 , water, and ethanol to remove unincorporated label. Plates were then counted on TopCount to assess the level of product synthesis over background controls.
  • the IC50 value was determined using Prism fitting program
  • Replicon cells were seeded in 96-well plates at a density of 8 x 10 3 cells per well in 100 ⁇ L of culture medium, excluding Genetic in. Compound was serially diluted in 100% DMSO and then added to the cells at a 1 :200 dilution, achieving a final concentration of 0.5% DMSO and a total volume of 200 ⁇ L. Plates were incubated at 37°C for 3 days, after which culture medium was removed and cells were lysed in lysis buffer provided by Promega's luciferase assay system. Following the manufacturer's instruction, 100 ⁇ L of luciferase substrate was added to the lysed cells and luciferase activity was measured in a TopCount luminometer.
  • cytotoxicity of a compound of the invention can be determined using the following general protocol. Cytotoxicity Cell Culture Assay (Determination of CC50):
  • the assay is based on the evaluation of cytotoxic effect of tested compounds using a metabolic substrate.
  • Assay protocol for determination ofCC50 1. Maintain MT-2 cells in RPMI-1640 medium supplemented with 5% fetal bovine serum and antibiotics.
  • the invention also relates to methods of making the compounds of the invention.
  • the compounds are prepared by any of the applicable techniques of organic synthesis. Many such techniques are well known in the art. However, many of the known techniques are elaborated in Compendium of Organic
  • L-Ribose 1 was converted into the iodides 2, which were reductively cleaved with Zn to afford the aldehyde 3.
  • This aldehyde was unstable and was reacted directly with ethyl diazoacetate to afford keto ester 4.
  • the keto ester was reacted with tosyl azide to afford the diazo derivative 5, which underwent a thermally-induced intramolecular cyclopropanation to give the bicyclo[3.1.0]hexan-2-one derivatives 6 and 7 with a diastereoisomeric ratio of (3:1) favoring isomer 6.
  • the bicyclo derivative 6 was isolated by chromatography and reduced stereospecifically with NaBH 4 to give alcohol 8 as a single product in 69% yleld.
  • This alcohol 8 was subjected to an acid-catalyzed equilibration to produce the isomeric acetonide 9, which was isolated in 35% yleld by crystallization from cyclohexane. The remaining alcohol 8 was recycled.
  • Ethyl (4S,5S)-3-[2,2-dimethyI-5-vinyl(l ⁇ -dioxolan-4-yl)]-3- oxopropanoate 4 Concentrated HCl was added to a solution containing L- Ribose 1 (75g, 0.5 mol) in acetone (200 ml) and methanol (200 ml) at room temperature. The resulting reaction mixture was refluxed for 2 hours, cooled to room temperature, neutralized with pyridine, and concentrated.
  • the crude residue was dissolved in toluene (800 ml) and was added with imidazole (51 g, 0.75 mol), triphenylphosphine (197g, 0.75 mol), and acetonitrile 175 ml).
  • the resulting reaction mixture was heated to 70 0 C, and Iodine (13 Ig, 0.5 mol) was added portion wise and carefully till the reaction mixture remained dark brown.
  • the reaction mixture was maintained at 70 0 C for 1 hour.
  • the top clear organic layer was decanted; and the residue was washed with toluene (200 ml).
  • the combined organic layer was washed with 5% sodium thiosulphate solution, water, and brine and concentrated.
  • Powdered Zn ( 16.2g, 0.25 mol) was added to the solution containing the iodides 2 (15.7 g, 0.05 mol) in methanol (25ml ) and isopropanol (75 ml). The resulting reaction mixture was allowed to reflux for 1 hour and was cooled at room temperature. The solution was decanted and concentrated (under reduced pressure). The residue was treated with water (100 ml) containing 5% acetic acid, extracted with dichloromethane (100 ml) , dried, and concentrated under reduced pressure to give the crude aldehyde 3 (7 g).
  • Example I Ethyl (1'S,2'R,3'S,4'R,5'S)-4'-(2,6-dichloropurin-9-yl]-2',3'-O- (isopropylidene)-bicyclo[3.1.0]hexanecarboxylate 17.
  • Compound 17 was obtained from 2,6-dichloro purine 10.
  • Example 3a (1'S,2'R,3'S,4'R,5'S-4) I -[6-Chloro-2-fluoropu ⁇ n-9-yI]-2 l ,3'- isopropylidenebicyclo[3.1.0]hexane-r-carboxylic Acid Ethyl Ester 19.
  • Example 4a (lW,2 ⁇ 3tf,4W,5 ⁇ SH46-Chloro-2-methylthiopurin-9-yl]-2',3'- isopropylidenebicyclo[3.1.0]hexane-l'-carboxylic Acid Ethyl Ester 20.
  • Compound 20 was obtained from 2-methylthio-6-chloro purine 13.
  • Example 5a (lW,2' ⁇ ,3'5',4W,5 l 5)-4 I - ⁇ 6-ChIoropurin-9-yl]-2',3'- isopropylidenebicyclo[3.1.0]hexane-l'-carboxylic Acid Ethyl Ester 21.
  • Example 6a (l' i S,2' ⁇ ,3W,4W,5' ) S)-4'-[4-chIoro-7H-pyrrolo[2,3-d]pyrimidin- 7-yl] ⁇ ' ⁇ '-isopropylidenebicyclo [3.1.0] hexane- 1 -car boxylic Acid Ethyl Ester 22.
  • Compound 22 was obtained from 4-chloro-7H-pyrrolo[2,3- d]pyrimidine 16.
  • Example Ib (1'S,2'R,3'S,4'R,5'S)-4-(2,6-dichIoro-purin-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 23.
  • HRMS (M + I) + : calculated 371.0678, found 371.0669.
  • Example 2b (1'S,2'R,3'S,4'R,5'S)-4-(6-chloro-2-iodo-purin-9-yI)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 24.
  • Example 3b (1'S,2'R,3'S,4'R,5'S)-4-(6-chIoro-2-fluoro-purin-9-yl)-1- [hydroxymethyl]bicycIo[3.1.0]hexane-2,3-(O-isopropylidine) 25.
  • Example 4b (1'S,2'R,3'S,4'R,5'S)-4-(6-chIoro-2-methythio-purin-9-yl)-1- [hydroxymethyI]bicycIo[3.1.0]hexane-2,3-(O-isopropyIidine) 26.
  • HRMS (M + I) + calculated 383.0945, found 383.0958.
  • Example 6b (1'S,2'R,3'S,4'R,5'S)-4-(4-chloro-7H-pyrrolo[2,3-d]pyrimidi ⁇ - 7-yI)-1-[hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropyIidine) 28.
  • Example 2c (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-iodo-purin ⁇ 9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropyIidine) 30.
  • HRMS (M + I) + calculated 444.0533, found 444.0522
  • Example 3c (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-fluoro-purin-9-yl)-1- [hydroxymethyI]bicycIo[3.1.0]hexane-2,3-(O-isopropylidine) 31.
  • Example 4c (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-methyIthiopurine-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 32.
  • Example 5c (1'S,2'R,3'S,4'R,5'S)-4 ⁇ (6-amino-purine-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 33.
  • Example 7 The following illustrate representative pharmaceutical dosage forms, containing a compound of formula I or II ('Compound X'), for therapeutic or prophylactic use in humans.

Abstract

The invention provides a compound of formula (I) and (II), or a salt or prodrug thereof, as described herein, as well as pharmaceutical compositions comprising the compounds, and therapeutic methods comprising administering the compounds. The compounds have anti-viral properties and are useful for treating viral infections (e.g. HCV) in animals (e.g. humans).

Description

BI CYCLO ( 3 . 1 . 0 ) HEXANE DERIVATIVES AS ANTIVIRAL COMPOUNDS
BACKGROUND OF THE INVENTION
Hepatitis C (HCV) is the most comon blood-born infection in the United States, where about 35,000 new cases are estimeted to occur each year. There is currently a need for compounds, compositions, and methods that are useful for treating viral infections such as HCV.
SUMMARY OF THE INVENTION
The present invention provides compounds that inhibit one or more viral proteases. Accordingly, the compounds may be useful for treating viruses, such as HCV.
In one embodiment the invention provides a compound of the invention, which is a compound of formula I or II:
Figure imgf000002_0001
wherein:
Ri is H, halo, ORa, SRa, or NRbRc;
R2 is H, halo, (C1-C6)alkyl , (C2-C6)alkenyl, or (C2-C6)alkynyl;
R3 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; and R4 is H, halo, azido, cyano, nitro, ORa, SRa, NRbRc, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; or R3 and R4 taken together are =O, =NRd, =CRfRg; or R3 and R4 taken together with the carbon atom to which they are attached form a 3-7 membered carbocyclic ring wherein one carbon atom can optionally be replaced with -O-, -S-, or -NRa-;
R5 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; and R6 is
H, halo, azido, cyano, nitro, ORa, SRa, NRbRc, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; or R5 and R6 taken together are =O, =NRd, =CRfRg; or R5 and R6 taken together with the carbon atom to which they are attached form a 3-7 membered carbocyclic ring wherein one carbon atom can optionally be replaced with -O-, -S-, or -NRa-; R7 and R8 are each independently H or halo;
R9 is a nucleoside base;
R10 is OH, -OP(=O)(OH)2, -OP(=O)(OH)-P(=O)(OH)2, -OP(=OXOH)-P(=O)(OH)-P(=O)(OH)2, or a phosphonate; each R3 is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each Rb and Rc is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each Rd is independently H, (C1-C6)alkyl, or OH; each Re is independently H or (C1-C6)alkyl; and each Rf and Rg is independently H, (C1-C6)alkyl, or halo; wherein each (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl OfR2-R6 and Ra-Rg is optionally substituted with one or more halo, hydroxy, or (C1- C6)alkoxy; or a pharmaceutically acceptable salt or prodrug thereof. The invention also provides a pharmaceutical composition comprising an effective amount of a compound of the invention, or a pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable excipient. The invention also provides metabolites of compounds of the invention, as well as salts thereof.
The invention also provides a method of inhibiting a viral infection in an animal (e.g. a mammal), comprising administering an effective amount a compound of the invention, or a pharmaceutically acceptable salt or prodrug thereof, to the animal.
The invention also provides a compound of the invention for use in medical therapy (preferably for use in treating a viral infection in an animal), as well as the use of a compound of the invention for the manufacture of a medicament useful for the treatment of a viral infection in an animal (e.g. a mammal).
The invention also provides processes and novel intermediates disclosed herein which are useful for preparing compounds of the invention. Some of the compounds of the invention are useful to prepare other compounds of the invention.
In another embodiment the invention provides a method for inhibiting a viral infection in a sample comprising treating a sample suspected of containing a virus, with a compound of the invention, or a pharmaceutically acceptable salt or prodrug thereof.
BRIEF DESCRIPTION OF THE FIGURES
Figures 1-4 illustrate synthetic processes and intermediates that can be used to prepare compounds of the invention.
DETAILED DESCRIPTION OF THE INVENTION
Unless stated otherwise, the following terms and phrases as used herein are intended to have the following meanings:
When tradenames are used herein, applicants intend to independently include the tradename product and the active pharmaceutical ingredient(s) of the tradename product.
Alkyl, alkoxy, alkenyl, alkynyl, etc. denote both straight and branched groups; but reference to an individual radical such as propyl embraces only the straight chain radical, a branched chain isomer such as isopropyl being specifically referred to. "Aryl" means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Typical aiyl groups include, but are not limited to, radicals derived from benzene, substituted benzene, naphthalene, anthracene, biphenyl, and the like. "Carbocycle" refers to a saturated, unsaturated or aromatic ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a polycycle. Monocyclic carbocycles have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms. Bicyclic carbocycles have 7 to 12 ring atoms, e.g., arranged as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as a bicyclo [5,6] or [6,6] system. Examples of monocyclic carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1- cyclopent-1-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1- cyclohex-1-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, phenyl, spiryl and naphthyl.
"Heterocycle" as used herein includes, by way of example and not limitation, those heterocycles described in Paquette, Leo A.; Principles of Modem Heterocyclic Chemistry (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; The Chemistry of Heterocyclic Compounds, A Series of Monographs" (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem, Soc. (1960) 82:5566. In one specific embodiment of the invention "heterocycle" includes a "carbocycle" as defined herein, wherein one or more {e.g., 1, 2, 3, or 4) carbon atoms haVe been replaced with a heteroatom (e.g., O, N, or S).
Examples of heterocycles include, by way of example and not limitation, pyridyl, dihydroypyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofuranyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, octahydroisoquinolinyl, azocinyl, triazinyl, 6H-1,2,5- thiadiazinyl, 2H,6H-l,5,2-dithiazinyl, thienyl, thianthrenyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathinyl, 2H-pyrrolyl, isothiazolyl, isoxazolyl, pyrazinyl, pyridazinyl, indolizinyl, isoindolyl, 3H- indolyl, lH-indazoly, purinyl, 4H-quinolizinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, quinazolinyl, cinnolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, β- carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, furazanyl, phenoxazinyl, isochromanyl, chromanyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperazinyl, indolinyl, isoindolinyl, quinuclidinyl, morpholinyl, oxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, isatinoyl, and bis-tetrahydrofuranyl:
Figure imgf000005_0001
By way of example and not limitation, carbon bonded heterocycles are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline. Still more typically, carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5- pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyI, 6-pyridazinyl, 2- pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3- pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl.
By way of example and not limitation, nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3 -imidazoline, pyrazole, pyrazolone, 2-pyrazoline, 3 -pyrazolone, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or β-carboline. Still more typically, nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, 1-pyrrolyl, 1-imidazolyl, 1- pyrazolyl, and 1-piperidinyl.
Heterocycle encompasses "heteroaryl," which includes a radical attached via a ring carbon of a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(X) wherein X is absent or is H, O, (C1-C6)alkyl, phenyl or benzyl, as well as a radical of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms derived therefrom, particularly a benz-derivative or one derived by fusing a propylene, trimethylene, or tetramethylene diradical thereto.
The term "substituted heterocycle" includes heterocycles as described herein that are substituted with one or more (e.g. 1, 2, 3, or 4) substituents selected from (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, oxo, NRxxRyy -C(=O)NRxxRyy, cyano, halo, hydroxy, nitro, carboxy, (C3-Cs)cycloalkyl, (C3-C8)cycloalkoxy, guanidino, trifluoromethoxy, mercapto, or trifluoromethyl; wherein Rxx and Ryy are each independently H, (C1-C6)alkyl, (C1-C6)alkoxy, (C1-C6)alkanoyls or aryl(C1-C6)alkyl.
The terms "phosphonate" and "phosphonate group" include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double-bonded to a heteroatom, 3) single-bonded to a heteroatom, and 4) single-bonded to another heteroatom, wherein each heteroatom can be the same or different. The terms "phosphonate" and "phosphonate group" also include functional groups or moieties that comprise a phosphorous in the same oxidation state as the phosphorous described above, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having the characteristics described above. For example, the terms "phosphonate" and "phosphonate group" include phosphonic acid, phosphonic monoester, phosphonic diester, phosphonamidate, and phosphonthioate functional groups. In one specific embodiment of the invention, the terms "phosphonate" and "phosphonate group" include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double- bonded to an oxygen, 3) single-bonded to an oxygen, and 4) single-bonded to another oxygen, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having such characteristics. In another specific embodiment of the invention, the terms "phosphonate" and "phosphonate group" include functional groups or moieties within a molecule that comprises a phosphorous that is 1) single-bonded to a carbon, 2) double-bonded to an oxygen, 3) single-bonded to an oxygen or nitrogen, and 4) single-bonded to another oxygen or nitrogen, as well as functional groups or moieties that comprise a prodrug moiety that can separate from a compound so that the compound retains a phosphorous having such characteriatics. The tenn "treatment" or "treating," to the extent it relates to a disease or condition includes preventing the disease or condition from occurring, inhibiting the disease or condition, eliminating the disease or condition, and/or relieving one or more symptoms of the disease or condition. Prodrugs
The term "prodrug" as used herein refers to any compound that when administered to a biological system generates the drug substance, i.e. active ingredient, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), photolysis, and/or metabolic chemical reaction(s). A prodrug is thus a covalently modified analog or latent form of a therapeutically- active compound.
"Prodrug moiety" refers to a labile functional group which separates from the active inhibitory compound during metabolism, systemically, inside a cell, by hydrolysis, enzymatic cleavage, or by some other process (Bundgaard, Hans, "Design and Application of Prodrugs" in A Textbook of Drug Design and Development (1991), P. Krogsgaard-Larsen and H. Bundgaard, Eds. Harwood Academic Publishers, pp. 113-191). Enzymes which are capable of an enzymatic activation mechanism with a phosphate prodrug of the invention include, but are not limited to, amidases, esterases, microbial enzymes, phospholipases, cholinesterases, and phosphases. Prodrug moieties can serve to modulate (e.g. enhanse) solubility, absorption, lipophilicity, drug delivery, bioavailability, and efficacy.
Exemplary prodrug moieties include the hydrolytically sensitive or labile acyloxymethyl esters -CH2OC(=O)Rx and acyloxymethyl carbonates
-CH2OC(=O)ORX where Rx is (C1-C6)alkyl, (C1-C6)substituted alkyl, C6-C20 aryl or C6-C2O substituted aryl. The acyloxyalkyl ester was first used as a prodrug strategy for carboxylic acids and then applied to phosphates and phosphonates by Farquhar et al. (1983) J. Pharm. ScI 72: 324; also US Patent Nos. 4816570, 4968788, 5663159 and 5792756. Subsequently, the acyloxyalkyl ester was used to deliver phosphonic acids across cell membranes and to enhance oral bioavailability. A close variant of the acyloxyalkyl ester, the alkoxycarbonyloxyalkyl ester (carbonate), may also enhance oral bioavailability of a compound of the invention. An exemplary acyloxymethyl ester is pivaloyloxymethoxy, (POM) -CH2OC(=O)C(CH3)3. An exemplary acyloxymethyl carbonate prodrug moiety is pivaloyloxymethylcarbonate (POC) -CH2OC(=O)OC(CH3)3.
Aryl esters of phosphorus groups, especially phenyl esters, are reported to enhance oral bioavailability (De Lombaert et al. (1994) J Med. Chein. 37: 498). Phenyl esters containing a carboxylic ester ortho to the phosphate have also been described (Khamnei and Torrence, (1996) J Med. Chem. 39:4109- 4115). Benzyl esters are reported to generate the parent phosphonic acid. In some cases, substituents at the ortho-ox jtrørø-position may accelerate the hydrolysis. Benzyl analogs with an acylated phenol or an alkylated phenol may generate the phenolic compound through the action of enzymes, e.g., esterases, oxidases, etc., which in turn undergoes cleavage at the benzylic C-O bond to generate the phosphoric acid and a quinone methide compound. Examples of this class of prodrugs are described by Mitchell et al. (1992) J. Chem. Soc.
Perkin Trans. //2345; and Glazier WO 91/19721. Still other benzylic prodrugs have been described containing a carboxylic ester-containing group attached to the benzylic methylene (Glazier WO 91/19721).
Thio-containing prodrugs are reported to be useful for the intracellular delivery of phosphorous containing drugs. Certain of these proesters contain an ethylthio group in which the thiol group is either esterified with an acyl group or combined with another thiol group to form a disulfide. Deesterification or reduction of the disulfide generates the free thio intermediate, which subsequently breaks down to the phosphoric acid and an episulfide (Puech et al. (1993) Antiviral Res., 22: 155-174; Benzaria et al. (1996) J. Med. Chem. 39: 4958).
Prodrug moities that can be incorporated into compounds of the invention include those described by Mackman, R.L. and Cihlar, Tomas, Annual Reports in Medicinal Chemistry, 2004, 39, 305-321. Other prodrug moities that can be incorporated into compounds of the invention include those described by Peyrottes et. al., Minireviews in Medicinal Chemistry, 2004, 4(4), 395-408; and Meier, Chris, Advances in Antiviral Drug Design, 2004, 4, 147-213.
Protecting Groups
In the context of the present invention, protecting groups include prodrug moieties and chemical protecting groups.
"Protecting group" refers to a moiety of a compound that masks or alters the properties of a functional group or the properties of the compound as a whole. Chemical protecting groups and strategies for protection/deprotection are well known in the art. See e.g., Protective Groups in Organic Chemistry, Theodora W. Greene, John Wiley & Sons, Inc., New York, 1991. Protecting groups are often utilized to mask the reactivity of certain functional groups and to assist in the efficiency of desired chemical reactions, e.g., making and breaking chemical bonds in an ordered and planned fashion. Protection of functional groups of a compound alters other physical properties besides the reactivity of the protected functional group, such as the polarity, lipophilicity (hydrophobicity), and other properties that can be measured by common analytical tools. Chemically protected intermediates may themselves be biologically active or inactive. Protected compounds may also exhibit altered, and in some cases, optimized properties in vitro and in vivo, such as passage through cellular membranes and resistance to enzymatic degradation or sequestration. In this role, protected compounds with intended therapeutic effects are also prodrugs.
Various functional groups of the compounds of the invention may be protected. For example, protecting groups for -OH groups (whether hydroxyl, carboxylic acid, phosphonic acid, or other functions) include "ether- or ester- forming groups". Ether- or ester-forming groups are capable of functioning as chemical protecting groups in the synthetic schemes set forth herein.
A very large number of hydroxyl protecting groups and amide-forming groups and corresponding chemical cleavage reactions are described in Protective Groups in Organic Synthesis, Theodora W. Greene (John Wiley & Sons, Inc., New York, 1991, ISBN 0-471-62301-6) ("Greene"). See also Kocienski, Philip J.; Protecting Groups (Georg Thieme Verlag Stuttgart, New York, 1994). In particular Chapter 1, Protecting Groups: An Overview, pages 1-20, Chapter 2, Hydroxyl Protecting Groups, pages 21-94, Chapter 3, Diol Protecting Groups, pages 95-117, Chapter 4, Carboxyl Protecting Groups, pages 118-154, Chapter 5, Carbonyl Protecting Groups, pages 155-184. For protecting groups for carboxylic acid, phosphonic acid, phosphonate, sulfonic acid and other protecting groups for acids see Greene as set forth below. Such groups include by way of example and not limitation, esters, amides, hydrazides, and the like. Specific Embodiments
Specific values described for radicals, substituents, and ranges, as well as specific embodiments of the invention described herein, are for illustration only; they do not exclude other defined values or other values within defined ranges. For example, (C1-C6)alkyl can be methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl, pentyl, 3-pentyl, or hexyl; (C3-C6)cycloalkyl can be cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl; (C3-C6)cycloalkyl(C1- C6)alkyl can be cyclopropylmethyl, cyclobutylmethyl, cyclopeiitylmethyl, cyclohexylmethyl, 2-cyclopropylethyl, 2-cyclobutylethyl, 2-cyclopentylethyl, or 2-cyclohexylethyl; (C1-C6)alkoxy can be methoxy, ethoxy, propoxy, isopropoxy, butoxy, iso-butoxy, sec-butoxy, pentoxy, 3-pentoxy, or hexyloxy; (C2-C6)alkenyl can be vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1,- pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1- hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, or 5-hexenyl; (C2-C6)alkynyl can be ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1- hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, or 5-hexynyl; (C1-C6)alkanoyl can be acetyl, propanoyl or butanoyl; hydroxy(C1-C6)alkyl can be hydroxymethyl, 1- hydroxyethyl, 2-hydroxyethyl, 1-hydroxypropyl, 2-hydroxypropyl, 3- hydroxypropyl, 1-hydroxybutyl, 4-hydroxybutyl, 1-hydroxypentyl, 5- hydroxypentyl, 1-hydroxyhexyl, or 6-hydroxyhexyl; (C!-C6)alkoxycarbonyl can be methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl, pentoxycarbonyl, or hexyloxycarbonyl; (C1-C6)alkylthio can be methylthio, ethylthio, propylthio, isopropylthio, butylthio, isobutylthio, pentylthio, or hexylthio; (C2-C6)alkanoyloxy can be acetoxy, propanoyloxy, butanoyloxy, isobutanoyloxy, pentanoyloxy, or hexanoyloxy; aryl can be phenyl, indenyl, or naphthyl; and heteroaryl can be furyl, imidazolyl, triazolyl, triazinyl, oxazoyl, isoxazoyl, thiazolyl, isothiazoyl, pyrazolyl, pyrrolyl, pyrazinyl, tetrazolyl, pyridyl, (or its N-oxide), thienyl, pyrimidinyl (or its N-oxide), indolyl, isoquinolyl (or its N-oxide) or quinolyl (or its N-oxide). A specific value for R1 is H.
A specific value for R2 is H. A specific value for R3 is H. A specific value for R4 is H or OH. A specific value for R5 is H.
A specific value for R6 is OH.
A specific value for each of R7 and R5 is H.
A specific value for R10 is OH.
A specific value for R10 is -OP(=O)(OH)2.
A specific value for R10 is a phosphonate.
A specific value for R10 has the formula:
Figure imgf000012_0001
wherein:
Y2b is O or NH; each R1 is independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl; and each R2, is independently (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1- C6)alkanoyl, (C1-C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, cyano, halo, hydroxy, nitro, carboxy, (C3-C6)cycloalkyl, (C3-C6)cycloalkoxy, trifluoromethoxy, mercapto, or trifluoromethyl.
A specific value for R10 has the formula:
Figure imgf000013_0001
A specific value for R1Q has the formula:
Figure imgf000013_0002
A specific value for Rio has the formula:
Figure imgf000013_0003
wherein R2 is (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl. A specific value for Rio has the formula:
Figure imgf000014_0001
wherein: Y2b is O or N(R4); R2 is (C1-C6)alkyl, (C2-C6)alkenyl, aryl(C1-C6)alkyl, or (C2-C6)alkynyl; and R4 is (C1-C6)alkyl, (C2-C6)alkenyl, aryl(C1-C6)alkyl, or (C2-C6)alkynyl.
A specific value for Rio has the formula:
Figure imgf000014_0002
In one specific embodiment of the invention, the compound of formula I is not (l'5',2li?,31 iSr,4li?,5'5)-4-(6-amino-purin-9-yl)-1-[hydroxymethyl]bi- cyclo[3.1.0]hexane-2,3-diol.
In another specific embodiment of the invention, the compound of formula I is not 4-(6-amino-purin-9-yl)-1-[hydroxymethyl]bi- cyclo[3.1.0]hexane-2,3-diol.
In another specific embodiment of the invention, the compound of formula I is not: (l'Sr,2'i?,3'5,4'i?,5'ιS)-4-(6-amino-9H-purin-9-yl)-1-
[phosphoryloxymethyl]bi-cyclo[3.1.0]hexane-2,3-diol; 5-(6-amino-9H-purin-9- yl)-1-[phosphoryloxymethyl]bicyclo[3.1.0]hexane-2,3-diol; 5-(6-amino-9H- purin-9-yl)-1-[hydoxymethyl]bicyclo[3.1.0]hexane-2,3-diol; (l'5l,2li?,3W,4'i?,5I»S)-4-(6-amino-9H-purin-9-yl)-1-[triphosphoryloxymethyl]bi- cyclo[3.1.0]hexane-2,3-diol; 5-(6-amino-9H-purin-9-yl)-1-
[triphosphoryloxymethyl]bicyclo[3.1.0]hexane-2,3-diol; (V S, 2%$' SA' R,5' S)-A-
(2,4(H,3H)-dioxopyrimidin-1-yl)-1-(phosphoryloxymethyl)bi- cyclo[3.1.0]hexane-2,3-diol; (rlS,2'i?,3'1S,4'i?,5',S)-4-(2,4(H,3H)-dioxopyrimidin- l-yl)-1- (diphosphoryloxymethyl)bicyclo[3.1.0]hexane-2,3-diol; or (l'5',2'i?,3'1S',41i?,5'5)-4-(2J4(H,3H)-dioxopyπmidin-1-yl)-1- (triphosphoryloxymethyl)bicyclo[3.1.0]hexane-2,3-diol.
In another specific embodiment of the invention the compounds of formula I exclude compounds wherein: Ri is H; R2 is H; R3 is H; R4 is OH; R5 is H; Re is OH; R7 and Rg are each H; R9 is 6-amino-9H-purin-9-yl or
2,4(H,3H)-dioxopyrimidin-1-yl; and R10 is OH, -OP(K))(OH)2, -OP(=O)(OH)- P(8O)(OH)2, or -OP(=OXOH)-P(=O)(OH)-P(=O)(OH)2.
In another sprcific embodiment of the invention the compounds of formula II exclude compounds wherein: Rj is H; R2 is H; R3 is H; R4 is OH; R5 is H; R6 is OH; R7 and R8 are each H; R9 is 6-amino-9H-purin-9-yl; and Rio is OH, -OP(=O)(OH)2, -OP(=O)(OH)-P(=O)(OH)2, or -OP(=O)(OH)-P(=O)(OH)-P(=O)(OH)2,
Nucleoside base The invention provides compounds of formula I and II wherein R9 is a nucleoside base. A multitude of nucleoside base analogs have been reported. The compounds of formula I and II may include any nucleoside base, provided the final compound possesses useful therapeutic (e.g. anti-viral) properties. For example, a specific value for R9 is a heterocycle or a substituted heterocycle (e.g. a purine, substituted purine, pyrimidine, or a substituted pyrimidine).
Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in United States Patent Application Publication Number 2004/0147464. Accordingly, a specific value for R9 has the following formula:
Figure imgf000015_0001
wherein: R20 is OH or (C1-C6)alkoxy that is optionally substituted with one or more R22; and R2j is H or (C1-C6)alkyl that is optionally substituted with one or more R22; or R20 and R21 together with the nitrogen to which they are attached form a heterocyclic ring that is optionally subsitituted with one or more R22; each R22 is independently (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NR23R24, -C(=O)NR23R24, aryl, heteroaryl, cyano, halo, hydroxy, nitro, carboxy, or (C3- C8)cycloalkyl;
R23 and R24 are each independently H, (C1-C6)alkyl or (CrQs)alkanoyl; wherein each aryl or heteroaryl of R22 is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1- C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NR23R24, -C(=O)NR23R24, cyano, halo, hydroxy, nitro, carboxy, (C3-Cs)cycloalkyl, trifluoromethoxy, mercapto, or trifluoromethyl; and A, B, D, F, and E are each independently >N or >C-R2s; each R25 is independently H, cyano, nitro, (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, -NHCONH2, CC=O)NR26R27, COOR28, hydroxy, (C1-C6)alkoxy, -NR26R2?, halo, l,3-oxazol-2-yl, l,3-oxazol-5-yl, l,3-thiazol-2-yl, imidazol-2-yl, 2-oxo-[l,3]dithiol-4-yl, furan-2-yl, or 2H-[l,2,3]triazol-4-yl; each R26 and R27 is independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, (C3-C8)cycloalkyl, aryl, heterocycle, hydroxy, (C1-C6)alkoxy; or R26 and R27 together with the nitrogen to which they are attached form a heterocycle; and each R2S is independently H, (C1-C6)alkyl, (C1-C6)allcenyl, (Cr C6)alkynyl, (C3-Cs)cycloalkyl, aryl, or heterocycle; wherein each (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, (C3- C6)cycloalkyl, aryl, heterocycle, and (C1-C6)alkoxy of R26 and R27 is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1- C6)alkanoyl, (C1-C6)alkanoyloxy, (C1-C6)alkoxycarbony3, NH2, cyano, halo, hydroxy, nitro, carboxy, (C3-C6)cycloalkyl, (C3-C8)cycloalkoxy, trifluoromethoxy, or mercapto.
A specific value for R2o is OH, methoxy, or propoxy; and for R2i is H. A specific value for R20 and R2i together with the nitrogen to which they are attached form a pyrrolidin-1-yl, l,3,4,9-tetrahydro-beta-carbolin-2-yl, piperidinyl, azetidinyl, 3,6-dihydro-2H-pyridin-1-yl, or 3,4-dihydro-lH- isoquinolin-2-yl ring, which ring is optionally substituted with -(X=O)NH2. Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in United States Patent Application Publication Number 2005/0215511. Accordingly, a specific value for R9 has the following formula:
Figure imgf000017_0001
wherein:
R30 is -C≡ CR31, -CH=CHR32, formyl, -CH=NHNR33, -CH=N(OR33), -CH(OR34), Or-B(OR33); R3i is tri(C1-C6)alkylsilyl, (C1-C6)alkoxy(C1-C6)alkyl, heteroaryl, aryl,
Carboxy, or (C1-C6)alkoxycarbonyl;
R32 is hydrogen or czs-(C1-C6)alkoxy;
R33 is H or (C1-C6)alkyl; and
R34 is (C1-C6)alkyl; wherein each aryl or heteroaryl of R31 is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1- C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NR35R36, -C(K))NR35R36, cyano, halo, hydroxy, nitro, carboxy, (C3-Cs)cycloalkyl, (C3-C6)cycloalkoxy, guanidino, trifluoromethoxy, mercapto, -S(=O)mR37, or trifluoromethyl; m is 0, 1, or 2;
R35 and R36 are each independently H, (C1-C6)alkyl or (C1-C6)alkanoyl;
R37 is (C1-C6)alkyl, aryl, hetrocycle, or NR3gR39; and
R38 and R39 are each independently H, (C1-C6)alkyl or (C1-C6)alkanoyl; wherein each aryl or heterocycle OfR37 is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1- C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NR35R36, -Ct=O)NR35R36, cyano, halo, hydroxy, nitro, carboxy, (C3-Cs)cycloalkyl, (C3-C8)cycloalkoxy, guanidino, trifluoromethoxy, mercapto, or trifluoromethyl.
A specific value for R30 is 2-trimethylsilylethynyl, 2-(2-pyridyl)ethynyl, 2-(4-pyridyl)ethynyl, 2-(4-methoxy)ethynyl, 2-(aminocarbonyl)ethynyl, 3,3- diethoxypropyn-1-yl, 2-(dimethylaminocarbonyl)ethynyl, 2-(N- amino(aminocarbonyl)ethynyl, 2-carboxyetliynyl, 2-ethoxycarbonylethynyl, 2- methoxycarbonylethynyl, 2-phenylethynyl, 2-(4-fluorophenyl)ethynyl, 2-(4- methylphenyl)ethynyl, vinyl, 2-methoxyvinyl, formyl, -CH=N-NH2, -CH=NOH, 1,1-diisopropoxymethyl, or -B(OH) 2.
Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 03/061385. Accordingly, a specific value for R9 has one of the the following formulae:
Figure imgf000018_0001
Figure imgf000019_0001
wherein:
R40 is H, NR4aR4b, NHC(=O)R4b, (C1-C6)alkylNR4aR4b, NHNH2, cyano, (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, aryl(C1-C6)alkyl, heterocycle(C1- C6)alkyl, halo, (C1-C6)alkylthio, (C1-C6)alkoxy, hydroxy, or mercapto;
R41 is H, (C1-C6)alkyl, hydroxy(C1-C6)alkyl, (C2-C6)alkenyl, (C2- C6)alkynyl, heterocycle, aryl, aryl(C1-C6)alkyl; each R42 is independently H, hydroxy, mercapto, cyano, -SNR4cR4d, -C(NH)NR4cR4d, -C(=NH)NHOH, -C(NH)NHO4c, -C(=NH)NHNR4cR4d,
NHCOR4c, SR4c, OR4c, SOR4c, SO2R4c, -C(=O)NR4cR4d, -C(=S)NR4cR4d, or R4c;
R43 is H, hydroxy, NR4cR4d, NHC(=O)NR4c, NHNHR4c, (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, heterocycle, aryl, aryl(C1-C6)alkyl, halo, COOR4c, CONR4cR4d, or absent when Y is N; R43 and R4b are each independently hydrogen, (C1-C6)alkyl, (C2-
C6)alkenyl, (C2-C6)alkynyl, heterocycle, or aryl;
R4c, and R4d are each independently hydrogen, (C1-C6)alkyl, (C2- C6)alkenyl, (C2-C6)alkynyl, heterocycle, or aryl;
X, Y, and W are each independently N, C, CR4c, S or P; R44 is H, hydroxy, mercapto, cyano, -SNR4cR4d, -C(NH)NR4cR4d, -C(=NH)NHOH, -C(NH)NHOR4c, -C(=NH)NHNR4oR4d, NHCOR4c, SR4c, OR4c, SOR4c, SO2R4c, -C(=O)NR4cR4d, -C(=S)NR4cR4d, Or R4c;
R45 is H, hydroxy, mercapto, cyano, -SNR4cR4d, -C(NH)NR4cR4d, -C(=NH)NHOH, -C(NH)NHO4c, -C(N=H)NHNR4cR4c1, NHCOR4c, SR4c, OR4c, SOR4c, SO2R4c, -C(O=)NR4cR4d, -C(=S)NR4cR4d, or R4c;
R46, and R47 together with the atoms to which they are attached form a heterocyclic ring; and
U is S or O; wherein each aryl or heterocycle is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1- C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, cyano, halo, hydroxy, nitro, carboxy, (C3-C8)cycloalkyl, (C3-C6)cycloalkoxy, trifluoromethoxy, mercapto, or trifluoromethyl.
Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 03/062257. Accordingly, a specific value for R9 has one of the following formulae:
Figure imgf000020_0001
Figure imgf000021_0001
wherein:
R50 is NR5aR5b, ONR5aR5b, NR5aNR5aR5b, SR5b, OR5b, H, hydroxy, (Cr C6)alkyl, (C1-C6)alkenyl, (C1-C6)all<ynyl, or aryl;
R5) is (C,-C6)alkyl, (C1-C6)alkanoyl, or aryl;
R52 is (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, or aryl;
R53 is H, halo, (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)a]kynyl, or aryl;
R54 is H or NH2; R55 is NR5aR5b, ONR5aR5b, NR5aNR5aR5b, SR5b, OR5b, H, hydroxy, (C1- C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, or aryl;
R56 is H, halo, (C1-C6)alkyl, or (C1-C6)alkenyl;
R57 and R5g are each independently -L-R5c; each L is independently a direct bond, -N(R5J1)-, O or S; each R5a and R5b is independently H, hydroxy, (C1-C6)alkyl, (C1- C6)alkenyl, (C!-C6)alkynyl, or aryl; and each R5c is NR5aR5b, H, hydroxy, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, or aryl; wherein each (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, or aryl of R5o-Rsg and Rsa-Rsc is optionally substituted with one or more (C1-C6)alkyl, (C1- C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-C6)alkanoyloxy, (C1- C6)alkoxycarbonyl, cyano, halo, hydroxy, nitro, carboxy, (C3-C8)cycloalkyl, (C3- C8)cycloalkoxy, trifluoromethoxy, mercapto, or trifluoromethyl.
Specific nucleoside base sti-uctures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 03/072757. Accordingly, a specific value for R9 has one of the following formulae:
Figure imgf000022_0001
wherein:
X2, X3; and X4, are each independently N, CH, or C-R6a; R60, R61, and R6a are each independently H, halo, NR6bR6c, hydroxyamino, NR6bNR6bR6c, N3, NO, NO2, formyl, cyano, -C(=O)NR6bR6c, -C(=S)NR6bR6c; -C(=O)OR6b, R6b, OR6b, or SR6b; and
R6b, and R6c are each independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl. Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 03/073989. Accordingly, a specific value for R9 has the following formula:
Figure imgf000022_0002
wherein:
X5, X6, and X7, are each independently N, CH, or C-R7a; R70 and R7a are each independently H, halo, NR7bR7c, hydroxyamino, NR7bNR7bR7c, N3, NO, NO2, formyl, cyano, -C(=O)NR7bR7c, -C(=S)NR7bR7c, -C(=O)OR7b, R7b, OR7b, or SR7b; and
R7b, and R70 are each independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl. Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 2004/080466. Accordingly, a specific value for R9 has the following formula:
Figure imgf000023_0001
wherein:
Y1=Y2 is -N=N-, -CH=N-, -N=CR8a-, or -CH=CR83-; and each R8a is independently H, halo, or (C1-C6)alkyl.
Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in
International Patent Application Publication Number WO 2005/021568.
Accordingly, a specific value for R9 has one of the following formulae:
Figure imgf000023_0002
wherein: each Z1 is independently N, C-R9a, O, S, NR913, >C=O, >C=S, >C=NR9b, >S=O, >S(O)2 or CH-R93; provided that if a Z1 participates in an optional bond represented by a dotted line — in the formula, then that Zi is N or C-R9a; and provided that if a Zi does not participate in an optional bond represented by a dotted line — in the formula, then that Z\ is O, S, NR9b, >C=O, >C=S, >C=NR%, >S=O, >S(O)2 or CH-R93;
X5a is O, S, SO, SO2, Se, SeO, SeO2 or NR9b; each W1 is C, CH, or N; wherein if a W1 participates in an optional bond represented by a dotted line — in the formula, then that W1 is C; and if a W1 does not participate in an optional bond represented by a dotted line — in the formula, then that W1 is CH, or N; each R9a is independently H, halo, NR9cR9d, hydroxyamino, NR9cNR9cR9d, N3, cyano, -C(=O)NR9cR9d, -C(=S)NR9cR9d, -C(=S)NR9cR9d, -C(=NH)0R9c, R9C, OR9C, or SR9c; each R9b is independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl; and
R9a, and R9d are each independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl.
Another specific value for R9 has one of the following formulae:
Figure imgf000024_0001
Figure imgf000025_0001
Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 2005/123087. Accordingly, a specific value for R9 has one of the following formulae:
Figure imgf000026_0001
wherein:
X4 is CRgo& or N;
X5 is O, S, orNR9ia ; Rp0 and R91 are each independently H, halo, hydroxy, (C1-C6)alkoxy,
NRgobR9ib, or heterocycle;
R9oa is H, halo, methyl, azido, or amino;
R91a is H, or (C1-C6)alkyl; and
R9Ob and R91b are each independently H, (C1-Ce^IlCyI, (C3-C8)cycloalkyl, or aryl(C1-C6)alkyl; wherein each (C1-C6)alkyl, (C3-Cs)cycloalkyl, aryl(C1-C6)alkyl, and heterocycle of R90-R91, R9U, and R9ob-R9ib are optionally substituted with one or more (e.g. 1, 2, 3, or 4) halo, hydroxy, amino, (C1-C6)alkyl, and (C1-C6)alkoxy.
Specific nucleoside base structures that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 2006/002231. Accordingly, a specific value for R9 is a 9-deazapurine derivative, such as, for example, a group of the following formulae:
Figure imgf000027_0001
wherein:
X6 is O, S, orNR70a;
R.70 and R7J are each independently H, halo, hydroxy, niercapto, aryl, heterocycle, (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, (C1-C6)alkylthio, arylthio, -S(=O)(C,-C6)alkyl, -S(=O)2(C1-C6)alkyl, -S(=O)2NR7obR7ib, NR7obR7ib) (C1-C6)alkoxy, aryloxy, (heterocycle)oxy;
R7Oa is H, methyl, ethyl, or acetyl; and
R7Ob and R7] b are each independently H, (C1-C6)alkyl, aryl, aryl(Cj- C6)alkyl, (C1-C6)alkanoyl, or aryl-C(=O)-;
Specific nucleoside base structoes that can be incorporated into the compounds of the invention include the nucleoside bases reported in International Patent Application Publication Number WO 2006/000922. Accordingly, a specific value for R9 a specific value for R9 has one of the following formulae:
Figure imgf000027_0002
Figure imgf000028_0001
wherein:
Ago, Bso, and Yso, are each independently H, halo, OR8O, S(O)nRsO, NRsoRst, cyano, trifluoromethyl, C(=W)OR80, C(=W)SR80, C(=W)NR80 Rsi, nitro, azido, carbocyclic, (C1-C6^IlCyI, (C1-C6)alkenyl, (C1-C6)alkynyl, aryl, aryl(C1-C6)alkyl, or heterocycle; or A80 and Bso taken together with the carbon atoms to which they are attached from a 4-7 membered carbocyclic or heterocyclic ring; n is O, I, or 2
Z80 is O, S, NR80, or CR80R8I; each V is independently N or CR80; and each R80 and Rsi is independently H, carbocycle, (C1-C6)alkyl, (C1-
C6)alkenyl, (C1-C6)alkynyl, halo, (C!-C6)alkoxy, amino, methylamino, dimethylamino, cyano, (C1-C6)alkanoyl, aryl, a1yl(C1-C6)alkyl, an amino acid residue (e.g. a natural amino acid residue) or heterocycle; or R80 and Rsi taken together with the atom(s) to which they are attached form a 3-7 membered carbocyclic or heterocyclic ring.
Another specific value for R9 has one of the following formulae:
Figure imgf000029_0001
wherein:
R97 is H, hydroxy, mercapto, (C1-C6)alkyl, (C1-C6)aIkoxy, (C2- C6)alkenyl, (C3-C8)cycloalkyl, aryl(C1-C6)alkyl, aryl or (C2-C6)alkynyl; R98 is H, hydroxy, mercapto, or (C1-C6)alkyl; R99 is H, halo, azido, cyano, nitro, O R99a, SR99a, NR99bR99c, (C1-C6)alkyl, (C2-C6)alkenyl, (C3-C8)cycloaikyl, aryl(C1-C6)alkyl, aryl or (C2-C6)alkynyl; each R99a is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each R99b and R99c is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; wherein each (C2-C6)alkyl, (C1-C6)alkoxy (C1-C6)alkenyl, (C1- C6)alkynyl, (C3-C8)cycloalkyl, aryl, and aryl(C1-C6)alkyl of R97, R98, R99 , R99a, R99b, and R99c, is optionally substituted with one or more (C1-C6)alkyl, (C1- C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-C6)alkanoyloxy, (C1- C6)alkoxycarbonyl, NH2, cyano, azido, halo, hydroxy, nitro, carboxy, trifluoromethoxy, aryl, or mercapto.
Another specific value for R9 is adenine, guanine, cytosine, uracil, thymine, 7-deazaadenine, 7-deazaguanine, 7-deaza-8-azaguanine, 7-deaza-8- azaadenine, inosine, nebularine, nitropyrrole, nitroindole, 2-aminopurine, 2- amino-6-chloropurine, 2,6-diaminopurine, hypoxanthine, pseudouridine, pseudocytosine, pseudoisocytosine, 5-propynylcytosine, isocytosine, isoguanine, 7-deazaguanine, 2-thiopyrimidine, 6-thioguanine, 4-thiothymine, 4-thiouracil, O6-methylguanine, N6-methyladenine, O4-methylthymine, 5,6-dihydrothymine, 5,6-dihydrouracil, 4-methylindole, substituted triazole, or pyrazolo[3,4- D]pyrimidine; Another specific value for R9 is 6-amino-2-chloro-purin-9-yl; 6-amino-2- iodo-purin-9-yl; 6-amino-2-fluoro-purin-9-yl; 6-amino-2-methylthio-purin-9-yl; 6-amino-purin-9-yl; or 4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl. Another specific value for Rg is adenine or cytosine, which adenine or cytosine is optionally substituted with one or more (C1-C6)all<yl, (Cr C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-C6)alkanoyloxy, (C1- C6)alkoxycarbonyl, NH2, cyano, halo, hydroxy, nitro, cavboxy, trifluoromethoxy, aryl, or mercapto.
Stereoisomers
The compounds of the invention may have chiral centers, e.g., chiral carbon or phosphorus atoms. The compounds of the invention thus include racemic mixtures of all stereoisomers, including enantiomers, diastereomers, and atropisomers. In addition, the compounds of the invention include enriched or resolved optical isomers at any or all asymmetric, chiral atoms. In other words, the chiral centers apparent from the depictions are provided as the chiral isomers or racemic mixtures. Both racemic and diastereomeric mixtures, as well as the individual optical isomers isolated or synthesized, substantially free of their enantiomeric or diastereomeric partners, are all within the scope of the invention. Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and ElIeI, E. and Wilen, S., Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York. A single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents ( Stereochemistry of Carbon Compounds, (1962) by E. L. Eliel, McGraw Hill; Lochmuller, C. H., (1975) J. Chromatogr., 113:(3) 283-302). Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. Under method (1), diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, α-methyl-β-phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts. Alternatively, by method (2), the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and WiI en, S. (1994) Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yleld the free, enantiomerically enriched xanthene. A method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, α-methoxy-α- (trifluoromethyl)phenyl acetate (Jacob III. (1982) J. Org. Chem. 47:4165), of the racemic mixture, and analyzing the NMR spectrum for the presence of the two atropisomeric diastereomers. Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (Hoye, T., WO 96/15111). By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase (Chiral Liquid Chromatography (1989) W. J. Lough, Ed. Chapman and Hall, New York; Okamoto, (199O) J. ofChromatogr. 513:375-378). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
The compounds of the invention can also exist as tautomeric isomers in certain cases. Although only one delocalized resonance structure may be depicted, all such forms are contemplated within the scope of the invention. For example, ene-amine tautomers can exist for purine, pyrimidine, imidazole, guanidine, amidine, and tetrazole systems and all their possible tautomeric forms are within the scope of the invention.
Salts and Hydrates
The compositions of this invention optionally comprise salts of the compounds herein, especially pharmaceutically acceptable non-toxic salts containing, for example, Na+, Li+, K+, Ca+2 and Mg+2. Such salts may include those derived by combination of appropriate cations such as alkali and alkaline earth metal ions or aminonium and quaternary amino ions with an acid anion moiety, typically a carboxylic acid. Monovalent salts maybe preferred if a water soluble salt is desired.
Metal salts typically are prepared by reacting the metal hydroxide with a compound of this invention. Examples of metal salts which are prepared in this way are salts containing Li+, Na+, and K+. A less soluble metal salt can be precipitated from the solution of a more soluble salt by addition of the suitable metal compound.
In addition, salts may be formed from acid addition of certain organic and inorganic acids, e.g., HCl, HBr, ^SO^ H3PO4 or organic sulfonic acids, to basic centers, typically amines, or to acidic groups. Finally, it is to be understood that the compositions herein comprise compounds of the invention in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.
Also included within the scope of this invention are the salts of the parental compounds with one or more amino acids. Any amino acid can be used, especially the naturally-occurring amino acids found as protein components, although the amino acid typically is one bearing a side chain with a basic or acidic group, e.g., lysine, arginine or glutamic acid, or a neutral group such as glycine, serine, threonine, alanine, isoleucine, or leucine. For therapeutic use, salts of active ingredients of the compounds of the invention will typically be physiologically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base. However, salts of acids or bases which are not physiologically acceptable may also find use, for example, in the preparation or purification of a physiologically acceptable compound. All salts, whether or not derived form a physiologically acceptable acid or base, are within the scope of the present invention.
Pharmaceutical Formulations
Pharmaceutical formulations according to the present invention comprise one or more compounds of the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents. Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed. Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifylng agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent. The pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.
The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weightweight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 μg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur. Formulations suitable for administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient. The active ingredient is preferably present in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about 1.5% w/w.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of viral infections as described below.
Formulations 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.
Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
The formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents. The invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefor.
Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered orally, parenterally or by any other desired route.
Compounds of the invention can also be formulated to provide controlled release of the active ingredient(s) to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile of the active ingredient(s). Accordingly, the invention also provided compositions comprising one or more compounds of the invention formulated for sustained or controlled release.
An effective dose of active ingredient depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses) or against an active viral infection, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. It can be expected to be from about 0.0001 to about 100 mg/kg body weight per day. Typically, from about 0.01 to about 10 mg/kg body weight per day. More typically, from about .01 to about 5 mg/kg body weight per day. More typically, from about .05 to about 0.5 mg/kg body weight per day. For example, the daily candidate dose for an adult human of approximately 70 kg body weight will range from 1 mg to 1000 mg, preferably between 5 mg and 500 mg, and may take the form of single or multiple doses.
Routes of Administration
One or more compounds of the invention (herein referred to as the active ingredients) are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient. An advantage of the compounds of this invention is that they are orally bioavailable and can be dosed orally.
Combination Therapy
Active ingredients of the invention can also be used in combination with other active ingredients. Such combinations are selected based on the condition to be treated, cross-reactivities of ingredients and pharmaco-properties of the combination. For example, when treating a viral infection the compositions of the invention can be combined with other agents that are effective to treat a viral infection (such as other antiviral agents).
It is also possible to combine any compound of the invention with one or more other active ingredients in a unitary dosage form for simultaneous or sequential administration to a patient. The combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations. The combination therapy may provide "synergy" and "synergistic effect", i.e. the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect may be attained when the active ingredients are: (1) co- formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate foπnulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., in separate tablets, pills or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
Metabolites of the Compounds of the Invention
Also falling within the scope of this invention are the in vivo metabolic products of the compounds described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yleld a metabolic product thereof. Such products typically are identified by preparing a radiolabeled {e.g., C^ or H^) compound of the invention, administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g., by MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention even if they possess no antiviral activity of their own.
Methods for determining stability of compounds in surrogate gastrointestinal secretions are known. Compounds are defined herein as stable in the gastrointestinal tract where less than about 50 mole percent of the protected groups are deprotected in surrogate intestinal or gastric juice upon incubation for 1 hour at 37 °C. Simply because the compounds are stable to the gastrointestinal tract does not mean that they cannot be hydrolyzed in vivo. The phosphonate prodrugs of the invention typically will be stable in the digestive system but are substantially hydrolyzed to the parental drug in the digestive lumen, liver or other metabolic organ, or within cells in general.
Antiviral Activity
Another aspect of the invention relates to methods of inhibiting viral infections, comprising the step of treating a sample or subject suspected of needing such inhibition with a composition of the invention.
Within the context of the invention samples suspected of containing a virus include natural or man-made materials such as living organisms; tissue or cell cultures; biological samples such as biological material samples (blood, serum, urine, cerebrospinal fluid, tears, sputum, saliva, tissue samples, and the like); laboratory samples; food, water, or air samples; bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like. Typically the sample will be suspected of containing an organism which induces a viral infection, frequently a pathogenic organism such as an tumor virus. Samples can be contained in any medium including water and organic solventVwater mixtures. Samples include living organisms such as humans, and man made materials such as cell cultures. If desired, the anti-virus activity of a compound of the invention after application of the composition can be observed by any method including direct and indirect methods of detecting such activity. Quantitative, qualitative, and semiquantitative methods of determining such activity are all contemplated. Typically one of the screening methods described above are applied, however, any other method such as observation of the physiological properties of a living organism are also applicable.
The antiviral activity of a compound of the invention can be measured using standard screening protocols that are known. For example, the antiviral activity of a compound can be measured using the following general protocol. HCV IC5O Determination
Assay Protocol: NS5b polymerase assay (40 μL) was assembled by adding 28 μL polymerase mixture (final concentration: 50 mM Tris-HCl at pH 7.5, 10 mM KCL, 5 mM MgCl2, 1 mM DTT, 10 mM EDTA, 4 ng/μL of RNA template, and 75 nM HCV Δ21 NS5b polymerase) to assay plates followed by 4 μL of compound dilution. The polymerase and compound were pre-incubated at 35 0C for 10 minute before the addition of 8 μL of nucleotide substrate mixture (33P-α-labeled competing nucleotide at KM and 0.5 mM of the remaining three nucleotides). The assay plates were covered and incubated at 35 0C for 90 min. Reactions were then filtered through 96-well DEAE-81 filter plates via vacuum. The filter plates were then washed under vacuum with multiple volumes of 0.125 MNaHPO4, water, and ethanol to remove unincorporated label. Plates were then counted on TopCount to assess the level of product synthesis over background controls. The IC50 value was determined using Prism fitting program
HCV EC50 Determination
Replicon cells were seeded in 96-well plates at a density of 8 x 103 cells per well in 100 μL of culture medium, excluding Genetic in. Compound was serially diluted in 100% DMSO and then added to the cells at a 1 :200 dilution, achieving a final concentration of 0.5% DMSO and a total volume of 200 μL. Plates were incubated at 37°C for 3 days, after which culture medium was removed and cells were lysed in lysis buffer provided by Promega's luciferase assay system. Following the manufacturer's instruction, 100 μL of luciferase substrate was added to the lysed cells and luciferase activity was measured in a TopCount luminometer.
Typically, compounds of the invention that were tested were found to have an EC50 of less than about 1000 μM (Huh7). Some compounds demonstrated an EC50 of less than about 250 μM (Huh 7). The cytotoxicity of a compound of the invention can be determined using the following general protocol. Cytotoxicity Cell Culture Assay (Determination of CC50):
The assay is based on the evaluation of cytotoxic effect of tested compounds using a metabolic substrate. Assay protocol for determination ofCC50: 1. Maintain MT-2 cells in RPMI-1640 medium supplemented with 5% fetal bovine serum and antibiotics.
2. Distribute the cells into a 96-well plate (20,000 cell in 100 μl media per well) and add various concentrations of the tested compound in triplicate (100 μl/well). Include untreated control. 3. Incubate the cells for 5 days at 37 0C.
4. Prepare XTT solution (6 ml per assay plate) in dark at a concentration of 2mg/ml in a phosphate-buffered saline pH 7.4. Heat the solution in a water- bath at 550C for 5 min. Add 50 μl of N-methylphenazonium methasulfate (5 μg/ml) per 6 ml of XTT solution. 5. Remove 100 μl media from each well on the assay plate and add 100 μl of the XTT substrate solution per well. Incubate at 37 0C for 45 to 60 min in a CO2 incubator. 6. Add 20 μl of 2% Triton X-100 per well to stop the metabolic conversion of
XTT. 7. Read the absorbance at 450 nm with subtracting off the background at 650 nm.
8. Plot the percentage absorbance relative to untreated control and estimate the CC50 value as drug concentration resulting in a 50% inhibition of the cell growth. Consider the absorbance being directly proportional to the cell growth.
Exemplary Methods of Making the Compounds of the Invention.
The invention also relates to methods of making the compounds of the invention. The compounds are prepared by any of the applicable techniques of organic synthesis. Many such techniques are well known in the art. However, many of the known techniques are elaborated in Compendium of Organic
Synthetic Methods (John Wiley & Sons, New York), Vol. I, Ian T. Harrison and Shuyen Harrison, 1971; Vol. 2, Ian T. Harrison and Shuyen Harrison, 1974; Vol.
3, Louis S. Hegedus and Leroy Wade, 1977; Vol. 4, Leroy G. Wade, jr., 1980;
Vol. 5, Leroy G. Wade, Jr., 1984; and Vol. 6, Michael B. Smith; as well as
March, J., Advanced Organic Chemistry, Third Edition, (John Wiley & Sons, New York, 1985), Comprehensive Organic Synthesis. Selectivity, Strategy &
Efficiency in Modern Organic Chemistry. In 9 Volumes, Barry M. Trost,
Editor-in-Chief (Pergamon Press, New York, 1993 printing).
Exemplary methods for the preparation of the compounds of the invention are provided below. These methods are intended to illustrate the nature of such preparations are not intended to limit the scope of applicable methods. Exemplary methods for the preparation of the compounds of the invention are also illustrated in Figures 1-6.
Synthetic intermediates and methods that are useful for preparing compounds of the invention are described by Joshi et al., J.Org. Chem., 2005, 70, 439-447; Gallos et al., Tetrahedron, 2002, 58, 8043-8053; Paquette et al., J.
Org. Chem., 1995, 60, 7849-7856; and Kim et al., J. Med. Chem., 2002, 45, 208-
218.
Representative compounds of the invention were prepared as described below. An intermediate compound 9 was prepared as illustrated below.
Figure imgf000043_0001
Figure imgf000044_0001
L-Ribose 1 was converted into the iodides 2, which were reductively cleaved with Zn to afford the aldehyde 3. This aldehyde was unstable and was reacted directly with ethyl diazoacetate to afford keto ester 4. The keto ester was reacted with tosyl azide to afford the diazo derivative 5, which underwent a thermally-induced intramolecular cyclopropanation to give the bicyclo[3.1.0]hexan-2-one derivatives 6 and 7 with a diastereoisomeric ratio of (3:1) favoring isomer 6. The bicyclo derivative 6 was isolated by chromatography and reduced stereospecifically with NaBH4 to give alcohol 8 as a single product in 69% yleld. This alcohol 8 was subjected to an acid-catalyzed equilibration to produce the isomeric acetonide 9, which was isolated in 35% yleld by crystallization from cyclohexane. The remaining alcohol 8 was recycled.
Preparation of compounds 2-9
Ethyl (4S,5S)-3-[2,2-dimethyI-5-vinyl(lβ-dioxolan-4-yl)]-3- oxopropanoate 4: Concentrated HCl was added to a solution containing L- Ribose 1 (75g, 0.5 mol) in acetone (200 ml) and methanol (200 ml) at room temperature. The resulting reaction mixture was refluxed for 2 hours, cooled to room temperature, neutralized with pyridine, and concentrated. The residue was partitioned between water (300 ml ) and ethyl acetate (300 ml three times); the combined organic phase was washed with saturated solution of copper sulphate, followed by brine, and the resulting solution was dried over sodium sulphate and concentrated. The crude residue was used directly in the next step ( 82 g).
The crude residue was dissolved in toluene (800 ml) and was added with imidazole (51 g, 0.75 mol), triphenylphosphine (197g, 0.75 mol), and acetonitrile 175 ml). The resulting reaction mixture was heated to 70 0C, and Iodine (13 Ig, 0.5 mol) was added portion wise and carefully till the reaction mixture remained dark brown. The reaction mixture was maintained at 70 0C for 1 hour. The top clear organic layer was decanted; and the residue was washed with toluene (200 ml). The combined organic layer was washed with 5% sodium thiosulphate solution, water, and brine and concentrated. The crude residue was treated with mixture containing hexane and ethyl acetate (3:1). Solid triphenylphosphine oxide separated out, was filtered, and the filtrate was concentrated. Further purification was effected by flash column chromatography, with ethyl acetate and hexane (5: 95 ) as the eluant, to give the iodo derivative 2 as a mixture of anomers. ( 95g, 64% in two steps).
Powdered Zn ( 16.2g, 0.25 mol) was added to the solution containing the iodides 2 (15.7 g, 0.05 mol) in methanol (25ml ) and isopropanol (75 ml). The resulting reaction mixture was allowed to reflux for 1 hour and was cooled at room temperature. The solution was decanted and concentrated (under reduced pressure). The residue was treated with water (100 ml) containing 5% acetic acid, extracted with dichloromethane (100 ml) , dried, and concentrated under reduced pressure to give the crude aldehyde 3 (7 g).
Aldehyde 3 (7 g) in dichloromethane (40 ml) was treated with SnCl4 (3.8g, 0.02 mol) and ethyldiazoacetate (7.6 g, 0.04 mol) at 0 0C, and the mixture was allowed to stir at room temperature for 8 hours. The mixture was filtered through a pad of celite, concentrated, and purified by column chromatography (silica gel, hexane: ethylacetate, 95:5) to afford 4, ( 5g, 42% based on the iodide derivative) as an oil. 1H NMR (CDCl3) δ 12.08 (s, 0.1 H, D2O exchangeable, enolic OH), 5.92-6.06 (m, 1 Η), 5.37-5.56 (m, 2 Η), 4.46-4.62 (m, 1 Η), 4.18- 438 (m, 3 H), 3.75 (AB q, 2 H, J= 16.4 Hz), 1.54-1.57 (m, 6 H, 2), 1.34-1.41 (m, 3 H); FAB MS m/z (relative intensity) 185 (7.2), 243 (MH+, 2).
Ethyl (45',55)-3-[2,2-dimethyI-5-vinyl(l,3-dioxolan-4-yl)]-2-diazo-3- oxopropanoate 5. To a stirred solution of keto ester 4 (9,68 g, 0.04 mol) in acetonitrile (40 mL) was successively added tosyl azide (8.26 g, 0.042mol) and Et3N (8.8 g, 80 mmol). The resulting reaction mixture was concentrated after stirring for 1 hour at room temperature The diazo derivative was directly purified by column chromatography (silica gel; hexanes:EtOAc, 90:10) to furnish 5 (7.5 g, 70%) as an oil 1H NMR (CDCl3) δ 5.79(ddd, 1 H, J= 17.3, 10.1, 7.3 Hz), 5.70 (d, 1 H, J= 7.6 Hz), 5.45 (d, 1 H, J= 17.1 Hz), 5.33 (d, 1 H, J= 10.2 Hz), 5.07 (t, 1 H, J= 7.5 Hz), 4.34 (q, 2 H, J= 7.1 Hz), 1.74 (s, 3 H), 1.51 (s, 3 H), 1.41 (t, 3 H, J= 7.1 Hz); FAB MS m/z (relative intensity) 269 (MH+, 86).
Ethyl (l£β5;4£;5£)-3,4-0-isopropyIidene-2-oxobicyclo[3.1.0]- hexanecarboxylate 6. To a stirred solution of diazo compound 5 (5.36 g, 20 mmol) in dry toluene (30 mL) was added CuI (0.190 g, 1 mmol) at room temperature. The reaction mixture was refluxed for 8 hours, cooled to room temperature, concentrated, and purified by column chromatography (silica gel; hexanes.ΕtOAc, 75:25) to provide bicylic compound 6 (1.72 g, 36%) and compound 7 (0.57 g, 12%). Compound 6: 1HNMR (CDCl3) δ 5.13 (ddd, 1 H, J = 8.3, 5.4, 1.0 Hz), 4.39 (d, 1 H, J= 8.3 Hz), 4.29 (q, 2 H, J= 7.1 Hz), 2.84 (dt, 1 H, J= 8.3, 5.3 Hz), 2.18 (dd, 1 H, J= 8.1, 5.1 Hz), 1.89 (t, 1 H, J- 5.2 Hz), 1.59 (s, 3 H), 1.39 (s, 3 H), 1.36 (t, 3 H, J= 7.1 Hz); FAB MS m/z (relative intensity) 241 (MH+, 100). 7: 1HNMR (CDCl3) δ 4.81 (d, 1 H, J= 5.1 Hz), 4.38 (dd, 1 H, J= 4.8, 1.7 Hz), 4.32 (dq, 2 H, J= 7.1, 1.5 Hz), 2.96 (dd, I H, J= 8.7, 5.7 Hz), 2.21 (ddd, 1 H, J- 8.8, 5.7, 1.7 Hz), 1.53 (s, 3 H), 1.45 (s, 3 H), 1.37 (t, 3 H, J= 4.1 Hz), 1.35 (irregular t, 1 H); FAB MS m/z (relative intensity) 241 (MH+, 100).
Ethyl (l^^iϊβ^^S^-S^-O-isopropylidene-1-hydroxybicycloIS.l.O]- hexane-carboxylate (8). To a stirred solution of compound 6 (2.4 g, 10 mmol) in ethanol (20 mL) at room temperature was added NaBH4 (0.38 g, 5 mmol) and stirring was continued for an additional 1 hour. The reaction mixture was treated with acetone (2 mL) and concentrated to dryness. The residue was purified by column chromatography (silica gel; hexanes:£tOAc, 70:30) to give compound 8 (1.68g, 69%) as a white solid; m.p.109 °C (cyclohexane); 1H NMR (CDCl3) δ 4.95 (t, 1 H, J= 7.5 Hz), 4.88 (t, 1 H, J= 6 Hz), 4.60 (t, 1 H, J= 7 Hz), 4.12- 4.24, (m, 2 H), 2.48 (d, 1 H, J= 12 Hz, OH), 2.16-2.26 (m, 1 H), 1.48-1.61 (m, 5 H), 1.32 (s, 3 H), 1.26 (t, 3 H, J= 6.5 Hz). FAB MS m/z (relative intensity) 243 (MH+, 100).
Ethyl (15',2i?,35',45',55)-2,3-O-(isopropyHdene)-4-hydroxybicyclo[3.1.0]- hexane-carboxylate 9. A solution of compound 8 (0.96 g, 4.0 mmol) andp- TsOH*H2O (0.38 g, 2 mmol) in acetone (20 mL) was refluxed for 8 hours. Following the addition OfNEt3 (2 mL), the solution was concentrated under reduced pressure. Flash column chromatography (silica gel; CHCl3MeOH, 9:1) of the residue furnished a mixture of isomerized alcohols 8 and 9 in a 6:4 ratio based on NMR. This crude mixture was further purified by careful crystallization from cyclohexane to obtain pure 9 (0.340 g, 35%) as colorless crystals. The remaining alcohol 8 was recycled. Compound 9: 1H NMR (CDCI3) δ 5.38 (d, 1 H, J= 5.5 Hz), 4.42-4.64 (m, 2 H), 4.08-4.21 (m, 2 H), 2.39-2.45 (m,
2 H), 1.42-1.62 (m, 5 H), 1.35 (s, 3 H), 1.12 (t, 2 H, J= 3.2 Hz); FAB MS m/z (relative intensity) 243.1 (MH+, 100).
As described in Examples 1-6 representative compounds of the invention were prepared from Compound 9 as illustrated in Figure 1. Alcohol 9 was subjected to a Mitsunobu coupling conditions using variety of purine bases 10- 16 to afford the condensed products 17-22. Reduction with DIBAL-H afforded compounds 23-28. The chloro functionality was substituted with an amino group to afford compounds 29-34. The final hydrolysis of the isopropylidene group using 10% TFA in MeOH at 70 °C afforded compounds 35-40, which are compounds of formula I.
Examples 1-6
The following general procedure was used to prepare the compounds of Examples 1-6. A mixture containing an isopropylidene analog 29-34 (0.04 mmol), 10% trifluoroacetic acid /MeOH (5 mL), and H2O (0.5 mL) was heated at 70 0C for
3 hours. The solvent was removed and the residue was dried by coevaporation with toluene. The residue was purified using preparative TLC (CHCl3:MeOH, 9:1) to afford the title compound 35-40 as a colorless solid in 62-67% ylelds.
Example 1 (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-chloro-purin-9-yl)-1- [hydroxymethyl]bicycIo[3.1.0]hexane-2,3-dioI 35. 1H NMR (CD3OD, 300 MHz) 50.71-0.78 (m, 1H), 1.50-1.64 (m, 2H), 3.85 (d, J= 6.9, Hz, 1H), 3.25 (d, J= 12, Hz, 1H), 4.75 (dd, J= 1.5, 1.5 Hz 1H), 4.76-4.79 (m, 2H), 8.49 (s, 1H). HRMS (M + I)+: calculated 312.0863, found 312.0871.
Example 2 (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-iodo-purin-9-yl)-1-
[hydroxymethyI]bicyclo[3.1.0]hexane-2,3-diol 36. 1HNMR (CD3OD, 300 MHz) S 0.72-0.78 (m, 1H), 1.51-1.62 (ra, 2H), 3.33-3.37 (m, 1H), 3.86 (d, J= 6.6, Hz, 1H), 4.25 (d, J= 13.5, Hz 1H), 4.76-4.79 (m, 2H), 8.42 (s, 1H). HRMS (M + I)+: calculated 404.0220, found 404.0204.
Example 3 (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-fluoro-purin-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-diol 37. 1HNMR (CD3OD, 300 MHz) 50.72-0.76 (m, 1H), 1.51-1.63 (m, 2H), 3.33-3.37 (m, 1H), 3.85 (d, J= 6.9, Hz, 1H), 4.24 (d, J= 12, Hz 1H), 4.76-4.79 (m, 2H), 8.45 (s, 1H). HRMS (M + I)+: calculated 296.1159, found 296.1162.
Example 4 (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-methylthio-purin-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-diol 38. 1H NMR (CD3OD, 300 MHz) 30.72-0.78 (m, 1H), 1.52-1.63 (m, 2H), 2.57 (s, 3H), 3.43-3.56 (m, 1H), 3.85-3.92 (m, 1H), 4.25 (d, J= 11.4, Hz, 1H), 4.76-4.79 (m, 2H), 8.41 (s, 1H). HRMS (M + I)+: calculated 324.1130, found 324.1120.
Example 5 (1'S,2'R,3'S,4'R,5'S)-4-(6-amino -purin-9-yI)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-diol 39. 1H NMR (CD3OD, 300 MHz) 50.73-0.79 (m, 1H), 1.52-1.66 (m, 2H), 3.28-3.33 (m, 1H), 3.88 (d, J= 3.6, Hz, 1H), 4.26 (d, J= 12, Hz, 1H), 4.76-4.79 (m, 2H), 8.20 (s, 1H), 8.51 (s, 1H). HRMS (M + I)+: calculated 278.1253, found 278.1252. Example 6 (1'S,2'R,3'S,4'R,5'S)-4-(4-amino-7H-pyrrolo[2,3-d]pyrimidin- 7-yI)4-[hydroxymethyl]bicycIo[3.1.0]hexane-2,3-diol 40. 1H NMR (CD3OD, 300 MHz) S 0.66-0.74 (m, 1H), 1.48-1.58 (m, 2H)3 3.28-3.33 (m, 1H), 3.76 (d, J = 6.9, Hz, 1H), 4.26 (d, J= 10.8, Hz, 1H), 4.75 (d, J= 6.6, Hz, 1H), 4.97 (s, 1H), 6.57-6.58 (m, 1H), 7.45 (s, 1H), 8.12 (s, 1H). HRMS (M + I)+: calculated 277.1301, found 277.1283.
The starting isopropylidene analogs 29-34 used in Examples 1 -6 were prepared as follows.
Examples la-6a
The following general procedure was used for the Mitsunobu coupling reaction of nucleoside purine bases 10-16 and the sugar moiety 9.
A mixture of triphenyl phosphine (0.104 g, 0.4 mmol), and the substituted purine 10-16 (0.4 mmol) in dry THF (2 niL) was treated with diisopropylazodicarboxylate (0.080 g, 0.4 mmol) at room temperature. After 20 minutes stirring, a solution of sugar 9 (0.048 g, 0.2 mmol) in THF (1 mL) was added and the mixture was stirred further for 8 hours. Concentration and purification of the residue by column chromatography (silica gel; Ethylacetate:- Hexane, 3:1) provided nucleoside ester analogues 17-22 in 35% to 42 % ylelds.
Example Ia Ethyl (1'S,2'R,3'S,4'R,5'S)-4'-(2,6-dichloropurin-9-yl]-2',3'-O- (isopropylidene)-bicyclo[3.1.0]hexanecarboxylate 17. Compound 17 was obtained from 2,6-dichloro purine 10. 1HNMR (CDCl3) δ 8.09 (s, 1 H), 5.85 (d, 1 H, 6.5 Hz), 4.91 (s, 1 H), 4.72 (d, 1 H, J= 5.5 Hz), 4.05-4.38 (m, 2 H), 2.14- 2.2 (m, 1 H), 1.75-1.82 (m, 1 H), 1.52-1.62 (m, 4 H), 1.15-1.38 (m, 6 H); FAB MS m/z (relative intensity) 413.1 (MH+, 100).
Example 2a (1'S,2'R,3'S,4'S,5'S)-4'-[6-ChIoro-2-iodopurin-9-yI]-2',3'- isopropylidenebicyclo[3.1.0]hexane-1'-carboxylic Acid Ethyl Ester 18.
Compound 18 was obtained from 2-iodo-6-chloro-purine 11. 1HNMR (CDCI3, 300 MHz) 61.30-1.36 (m, 4H), 1.57 (s, 6H), 1.75-1.80 (m, 1H), 2.12-2.22 (m, 1H), 4.1-4.2 (m, 2H)3 4.75 (d, J= 14 Hz, 1H), 4.92 (s, 1H), 5.85 (d, J= 15 Hz, 1H), 7.98 (s, 1H). MS (m/e) (positive-FAB) 505.0 (M + I)+.
Example 3a (1'S,2'R,3'S,4'R,5'S-4)I-[6-Chloro-2-fluoropuπn-9-yI]-2l,3'- isopropylidenebicyclo[3.1.0]hexane-r-carboxylic Acid Ethyl Ester 19.
Compound 19 was obtained from 2-fluoro-6-chloiO-purine 12. 1HNMR (CDCI3, 300 MHz) 51.23-1.36 (m, 4H), 1.55 (s, 6H), 1.76-1.82 (m, 1H), 2.42-2.46 (m, 1H), 4.14-4.28 (m, 2H), 4.73 (d, J= 10 Hz, 1H), 4.93 (s, 1H), 5.88 (d, J= 7 Hz, 1H), 8.07 (s, 1H). HRMS (M + I)+: calculated 397.1079, found 397.1083.
Example 4a (lW,2^3tf,4W,5^SH46-Chloro-2-methylthiopurin-9-yl]-2',3'- isopropylidenebicyclo[3.1.0]hexane-l'-carboxylic Acid Ethyl Ester 20. Compound 20 was obtained from 2-methylthio-6-chloro purine 13. 1H NMR (CDCl3, 300 MHz) 51.25-1.55 (m, 4H), 1.56 (s, 6H), 1.75-1.82 (m, 1H), 2.25- 2.32 (m, 1H), 2.64 (s, 3H), 4.18-4.29 (m, 2H), 4.82 (d, J= 12 Hz, 1H), 4.92 (s, 1H), 5.85 (d, J= 11 Hz, 1H), 7.90 (s, 1H). MS (m/e) (positive-FAB) 425.10 (M
+ D+.
Example 5a (lW,2'Λ,3'5',4W,5l5)-4I-{6-ChIoropurin-9-yl]-2',3'- isopropylidenebicyclo[3.1.0]hexane-l'-carboxylic Acid Ethyl Ester 21.
Compound 21 was obtained from 6-chloropurine 15. 1HNMR (CDCl3, 300 MHz) 51.26-1.57 (m, 4H), 1.59 (s, 6H), 1.75-1.8 (m, 1H), 2.24-2.32 (m, 1H), 4.12-4.38 (m, 2H), 4.76 (d, J= 8 Hz, 1H), 5.08 (s, 1H), 5.89 (d, J= 7 Hz, 1H), 8.10 (s, 1H), 8.71 (s, 1H). HRMS (M+ I)+: calculated 379.1173, found 379.1188.
Example 6a (l'iS,2'Λ,3W,4W,5')S)-4'-[4-chIoro-7H-pyrrolo[2,3-d]pyrimidin- 7-yl] ^'^'-isopropylidenebicyclo [3.1.0] hexane- 1 -car boxylic Acid Ethyl Ester 22. Compound 22 was obtained from 4-chloro-7H-pyrrolo[2,3- d]pyrimidine 16. 1HNMR (CDCl3, 300 MHz) 51.22-1.38 (m, 4H), 1.56 (s, 3H), 1.59 (s, 3H) 1.76-1.80 (m, 1H), 2.18-2.26 (m, 1H), 4.14-4.23 (m, 2H), 4.69 (d, J = 8 Hz, 1H), 5.05 (s, 1H), 5.83 (d, J= 8Hz, 1H), 6.64 (s, 1H), 7.15 (d, J= 3.6Hz, 1H), 8.60 (s, 1H) . HRMS (M + I)+: calculated 378.1221, found 378.1245. Examples 1b-6b
The following general procedure was used for the DIBAL-H reduction of esters 17-22. A stirred solution of ester 17-22 (0.5 mmol) in toluene (10 mL) was cooled to -78 0C and treated dropwise with diisobutylaluminum hydride (DIBAL-H, 1 mL, 1.5 M solution in THF). After 1 hour, methanol (1.5 mL) and 1 M cold sulfuric acid (10 ml) (pH = 4), were added carefully. The reaction mixture was filtered through a pad of Celite and the filtrate was collected, separated, and the aqueous phase was extracted with dichloromethane. The combined organic layer was evaporated and concentrated under reduced pressure. The residue was purified by silica gel flash column chromatography (CHCl3 :MeOH, 10:1) to give the corresponding alcohols 23-28 in 35 to 56 % ylelds.
Example Ib (1'S,2'R,3'S,4'R,5'S)-4-(2,6-dichIoro-purin-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 23. 1H NMR (CDCl3, 300 MHz) 51.05-1.08 (m, 1H), 1.21-138 (m, 7H), 1.70-1.74 (m, 1H), 3.52(d, J= 12Hz, 1H), 4.21-4.27 (m, 1H), 4.63 (d, J= 7.5Hz, 1H), 4.96 (s, 1H), 5.51 (d, J= 12Hz, 1H), 8.32 (s, 1H). HRMS (M + I)+: calculated 371.0678, found 371.0669.
Example 2b (1'S,2'R,3'S,4'R,5'S)-4-(6-chloro-2-iodo-purin-9-yI)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 24. 1H NMR (CDCl3, 300 MHz) 51.02-1.05 (m, 1H), 1.21-42 (m, 7H), 1.66-1.71 (m, 1H), 3.60 (d, J= 12Hz, 1H), 4.16(d, J= 12Hz, 1H), 4.65(d, J= 7.2Hz, 1H), 4.97 (s, 1H), 5.47 (d, J= 7.5Hz, 1H), 8.28 (s, 1H). HRMS (M + I)+: calculated 463.0034, found 463.0021.
Example 3b (1'S,2'R,3'S,4'R,5'S)-4-(6-chIoro-2-fluoro-purin-9-yl)-1- [hydroxymethyl]bicycIo[3.1.0]hexane-2,3-(O-isopropylidine) 25. 1H NMR (CDCl3, 300 MHz) 51.01-1.09 (m, 2H), 1.25 (s, 6H), 1.66-1.74 (m, 1H), 3.50 (d, J= 11Hz, 1H), 4.18-4.23(m, 1H), 4.62(d, J= 4.8Hz, 1H), 4.95 (s, 1H), 5.48 (dd, J= 1.5, 1.6Hz, 1H), 8.35 (s, 1H). HRMS (M + I)+: calculated 355.0973, found 355.0971.
Example 4b (1'S,2'R,3'S,4'R,5'S)-4-(6-chIoro-2-methythio-purin-9-yl)-1- [hydroxymethyI]bicycIo[3.1.0]hexane-2,3-(O-isopropyIidine) 26. HRMS (M + I)+: calculated 383.0945, found 383.0958.
Example 5b (1'S,2'R,3'S,4'R,5'S)-4-(6-chloropurin-9-yl)-l- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropyIidine) 27. HRMS (M + I)+: calculated 337.1067, found 337.1069.
Example 6b (1'S,2'R,3'S,4'R,5'S)-4-(4-chloro-7H-pyrrolo[2,3-d]pyrimidiπ- 7-yI)-1-[hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropyIidine) 28.
HRMS (M + I)+: calculated 336.1115, found 336.1115.
Examples 1c-6c
The following general procedures were used to convert the chloro group in analogs 23-28 to the corresponding amine. The chloro analog was added to a 20 fold excess of 2M NH3 in THF and the resulting mixture was stirred at room temperature for 16 hours. Alternatively, the chloro analog was added to a 20 fold excess of saturated aminonia in ethanol and the resulting mixture was stirred at 95 °C under pressure in a sealed vessel for 16 hours. Using the above procedures compounds 29-34 were obtained in 60 to 65 % ylelds.
Example 1c (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-chloro-purin-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropyIidine) 29. HRMS
(M + I)+: calculated 352.1176, found 353.1163.
Example 2c (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-iodo-purin~9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropyIidine) 30. HRMS (M + I)+: calculated 444.0533, found 444.0522 Example 3c (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-fluoro-purin-9-yl)-1- [hydroxymethyI]bicycIo[3.1.0]hexane-2,3-(O-isopropylidine) 31. HRMS
(M + I)+: calculated 336.1472, found 336.1474.
Example 4c (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-2-methyIthiopurine-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 32. 1H NMR (CDCI3, 300 MHz) 50.94-0.99 (m, 1H), 1.13-1.17 (m, 1H), 1.28 (s, 3H), 1.55 (s, 3H), 1.67-1.71 (m, 1H), 2.59 (s, 3H), 3.33 (dd, J= 2.1, 2.1 Hz, 1H), 3.86 (dd, J= 2.4, 2.4 Hz, 1H), 4.22-4.26 (m, 1H), 4.71-4.75 (m, 2H), 5.55-5.61 (m, 2H), 7.72 (s, 1H). HRMS (M + I)+: calculated 364.1443, found 364.1447.
Example 5c (1'S,2'R,3'S,4'R,5'S)-4~(6-amino-purine-9-yl)-1- [hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 33. 1H NMR (CD3OD, 300 MHz) 50.95-1.10 (m, 1H), 1.14-1.19 (m, 1H), 1.24 (s, 3H), 1.51 (s, 3H), 1.67-1.78 (m, 1H), 3.44 (d, J= 5.7 Hz, 1H), 4.06 (d, J= 11 Hz, 1H), 4.66 (d, J= 4.2 Hz, 1H), 5.02 (s, 1H), 5.40 (d, J= 7.2 Hz, 1H), 8.19 (s, 1H), 8.31 (s, 1H). HRMS (M + I)+: calculated 318.1566, found 318.1575.
Example 6c (1'S,2'R,3'S,4'R,5'S)-4-(4-amino-7H-pyrrolo[2,3-d]pyrJmidin- 7-yl)-1-[hydroxymethyl]bicyclo[3.1.0]hexane-2,3-(O-isopropylidine) 34. HRMS (M + I)+: calculated 317.1614, found 317.1615.
Example 7 The following illustrate representative pharmaceutical dosage forms, containing a compound of formula I or II ('Compound X'), for therapeutic or prophylactic use in humans.
(i) Tablet 1 mg/tablet
Compound X= 100.0
Lactose 77.5
Povidone 15.0
Croscarmellose sodium 12.0
Microcrystalline cellulose 92.5
Magnesium stearate 3.0
300.0 (if) Tablet 2 ma/tablet
Compound X= 20.0
Microcrystalline cellulose 410.0
Starch 50.0
Sodium starch glycolate 15.0
Magnesium stearate
500.0
(WY) Capsule mg/capsule
Compound X= 10.0
Colloidal silicon dioxide 1.5
Lactose 465.5
Pregelatinized starch 120.0
Magnesium stearate 3.0
600.0
Civ) Injection 1 (I mg/ml) mg/ml
Compound X= (free acid form) 1.0
Dibasic sodium phosphate 12.0
Monobasic sodium phosphate 0.7
Sodium chloride 4.5
1.0 N Sodium hydroxide solution
(pH adjustment to 7.0-7.5) q.s.
Water for injection q.s. ad 1 mL
Cv) Iniection 2 ("10 mg/ml) mg/ml
Compound X= (free acid form) 10.0
Monobasic sodium phosphate 0.3
Dibasic sodium phosphate 1.1
Polyethylene glycol 400 200.0
01 N Sodium hydroxide solution
(pH adjustment to 7.0-7.5) q.s.
Water for injection q.s. ad 1 mL
(vi) Aerosol mg/can
Compound X= 20.0
Oleic acid 10.0
Trichloromonofluoromethane 5,000.0
Dichlorodifluoromethane 10,000.0
Dichlorotetrafluoroethane 5,000.0
The above formulations may be obtained by conventional procedures well known in the pharmaceutical art.
All publications, patents, and patent documents cited hereinabove are incorporated by reference herein, as though individually incorporated by reference. The invention has been described with reference to various specific and preferred embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention.

Claims

ClaimsWe claim:
1. A compound of formula I or II:
wherein:
Figure imgf000056_0001
R1 is H, halo, ORa, SRa, or NRbRc;
R2 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; R3 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; and R4 is H, halo, azido, cyano, nitro, ORa, SRa, NRbRc, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; or R3 and R4 taken together are =O, =NRd, =CRfRg; or R3 and R4 taken together with the carbon atom to which they are attached form a 3-7 membered carbocyclic ring wherein one carbon atom can optionally be replaced with -O-, -S-, or -NRa-;
R5 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; and R6 is H, halo, azido, cyano, nitro, ORa, SR3, NRbRc, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; or R5 and R6 taken together are =O, =NRd, =CRfRg; or R5 and R6 taken together with the carbon atom to which they are attached form a 3-7 membered carbocyclic ring wherein one carbon atom can optionally be replaced with -O-, -S-, or -NRa-; R7 and R8 are each independently H or halo;
R9 is a nucleoside base;
R10 is OH, -OP(=O)(OH)2, -OP(=O)(OH)-P(=O)(OH)2, -OP(=O)(OH)-P(-O)(OH)-P(=O)(OH)2, or a phosphonate; each Ra is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each Rb and Rc is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each Rd is independently H, (C1-C6)alkyl, or OH; each Re is independently H or (C1-C6)alkyl; and each Rf and Rg is independently H, (C1-C6)alkyl, or halo; wherein each (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl of R2-R6 and Ra-Rg is optionally substituted with one or more halo, hydroxy, or (C1- C6)alkoxy; or a pharmaceutically acceptable salt or prodrug thereof; provided the compound is not (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-purin-9- yl)-1-[hydroxymethyljbicyclo[3.1.0]hexane-2,3-diol; and provided the compound is not (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-9H-purin- 9-yl)-1-[phosphoryloxymethyl]bi-cyclo[3.1.0]hexane-2,3-diol; 5-(6-amino-9H- purin-9-yl)-1-[phosphoryloxymethyl]bicyclo[3.1.0]hexane-2,3-diol; 5-(6-amino- 9H-purin-9-yl)-1-[hydoxymethyl]bicyclo[3.1.0]hexane-2,3-diol; (1'S,2'R,3'S,4'R,5'S)-4-(6-amino-9H-purin-9-yl)-1-[triphosphoryloxymethyl]bi- cyclo[3.1.0]hexane-2,3-diol; 5-(6-amino-9H-purin-9-yl)-1- [triphosphoryloxymethyl]bicyclo[3.1.0]hexane-2,3-diol; (1'S,2'R,3'S,4'R,5'S)-4- (2,4(H,3H)-dioxopyrimidin-1-yl)-1-(phosphoryloxymethyl)bi- cyclo[3.1.0]hexane-2,3-diol; (1'S,2'R,3'S,4'R,5'S)-4-(2,4(H,3H)-dioxopyrimidin- 1-yl)-1- (diphosphoryloxymethyl)bicyclo[3.1.0]hexane-2,3-diol; or (1'S,2'R,3'S,4'R,5'S)-4-(2,4(Η,3Η)-dioxopyrimidin-1-yl)-1- (triphosphoryloxymethyl)bicyclo[3.1.0]hexane-2,3-diol.
2. The compound of claim 1 wherein Ri is H; R2 is H; R3 is H; R4 is H or OH; R5 is H; and R6 is OH.
3. The compound of claim 1 wherein R7 and R8 are each H.
4. The compound of claim 1 wherein R9 is a heterocycle or a substituted heterocycle.
5. The compound of claim 1 wherein R9 has the following formula:
Figure imgf000058_0001
wherein:
R20 is OH or (C1-C6)alkoxy that is optionally substituted with one or more R22; and R2] is H or (C1-C6)alkyl that is optionally substituted with one or more R22; or R20 and R2i together with the nitrogen to which they are attached form a heterocyclic ring that is optionally substituted with one or more R22; each R22 is independently (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NR23R24, -C(=O)NR23R24, aryl, heteroaryl, cyano, halo, hydroxy, nitro, carboxy, or (C3- Cs)cycloalkyl; R23 and R24 are each independently H, (C1-C6)alkyl or (C1-C6)alkanoyl; wherein each aryl or heteroaryl OfR22 is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1- C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NR23R24, -C(=O)NR23R24, cyano, halo, hydroxy, nitro, carboxy, (C3-Cs)cycloalkyl, trifluoromethoxy, mercapto, or trifluoromethyl; and
A, B, D, F, and E are each independently >N or >C-R2s; each R25 is independently H, cyano, nitro, (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, -NHCONH2, C(=O)NR26R27, COOR28, hydroxy, (C1-C6)alkoxy, -NR26R27, halo, l,3-oxazol-2-yl, l,3-oxazol-5-yl, l,3-thiazol-2-yl, imidazol-2-yl, 2-oxo-[l, 3]dithiol-4-yl, furan-2-yl, or 2H-[1,2,3]triazol-4-yl; each R26 and R27 is independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, (C3-Cs)cycloalkyl, aryl, heterocycle, hydroxy, or (C1-C6)alkoxy; or R26 and R27 together with the nitrogen to which they are attached form a heterocycle; and each R28 is independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, (C3-C g)cycloalkyl, aryl, or heterocycle; wherein each (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, (C3- C8)cycloall<yl, aryl, heterocycle, and (C1-C6)alkoxy of R26 and R27 is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1- C6)alkanoyl, (C1-C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NH2, cyano, halo, hydroxy, nitro, carboxy, (C3-C6)cycloalkyl, (C3-Cs)cycloalkoxy, trifluoromethoxy, or mercapto.
6. The compound of claim 5 wherein R20 is OH, methoxy, or propoxy; and R21 is H.
7. The compound of claim 5 wherein R20 and R2; together with the nitrogen to which they are attached form a pyrrolidin-1-yl, 1,3,4,9-tetrahydro-beta- carbolin-2-yl, piperidinyl, azetidinyl, 3,6-dihydro-2H-pyridin-1-yl, or 3,4- dihydro-lH-isoquinolin-2-yl ring, which ring is optionally substituted with -C(=O)NH2.
8. The compound of claim 1 wherein R9 has the following formula:
Figure imgf000059_0001
wherein: R30 is -C≡ CR31, -CH=CHR32, formyl, -CH=NHNR33, -CH=N(OR33),
-CH(OR34X Or -B(OR33);
R31 is tri(C1-C6)alkylsilyl, (C1-C6)alkoxy(C1-C6)alkyl, heteroaryl, aryl, Carboxy, or (C1-C6)alkoxycarbonyl;
R32 is hydrogen or cis-(Cι-C(s)alkoxy; R33 is H or (C1-C6)alkyl; and
R34 is (C1-C6)alkyl; wherein each aryl or heteroaryl of R31 is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-
C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NR35R36, -C(=O)NR35R36, cyano, halo, hydroxy, nitro, carboxy, (C3-C8)cycloalkyl, (C3-C6)cycloalkoxy, guanidino, trifluoromethoxy, mercapto, -S(=O)mR37, or trifluoromethyl; m is 0, 1, or 2;
R.35 and R36 are each independently H, (C1-C6)alkyl or (C1-C6)alkanoyl; R37 is (C1-C6)alkyl, aryl, hetrocycle, or NR38R39; and
R38 and R39 are each independently H, (C1-C6)alkyl or (C1-C6)alkanoyl; wherein each aryl or heterocycle OfR37 is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1- C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, NR35R36, -C(=O)NR35R36, cyano, halo, hydroxy, nitro, carboxy, (C3-C6)cycloalkyl, (C3-C8)cycloalkoxy, guanidino, trifluoromethoxy, mercapto, or trifluoromethyl.
9. The compound of claim 8 wherein R30 is 2-trimethylsilylethynyl, 2-(2- pyridyl)ethynyl, 2-(4-pyridyl)ethynyl, 2-(4-methoxy)ethynyl, 2-(aminocarbonyl)ethynyl, 3,3-diethoxypropyn-1-yl,
2-(dimethylaminocarbonyl)ethynyl, 2-(N-amino(aminocarbonyl)ethynyl, 2-carboxyethynyl, 2-ethoxycarbonylethyiiyl, 2-methoxycarbonylethynyl, 2-phenylethynyl, 2-(4-fluorophenyl)ethynyl, 2-(4-methylphenyl)ethynyl, vinyl, 2-methoxyvinyl, formyl, -CH=N-NH2, -CH=NOH, 1,1-diisopropoxymethyl, or -B(OH)2.
10. The compound of claim 1 wherein R9 has one of the the following formulae:
Figure imgf000061_0001
wherein:
R40 is H, NR4aR4b, NHC(=O)R4b, (C1-C6)alkylNR4aR4b, NHNH2, cyano, (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, aryl(C1-C6)alkyl, heterocycle(C1- C6)alkyl, halo, (C1-C6)alkylthio, (C1-C6)alkoxy, hydroxy, or mercapto; R41 is H, (C1-C6)alkyl, hydroxy (C1-C6)alkyl, (C2-C6)alkenyl, (C2- C6)alkynyl, heterocycle, aryl, or aryl(C1-C6)alkyl; each R42 is independently H, hydroxy, mercapto, cyano, -SNR4cR4d, -C(NH)NR4cR4d, -C(=NH)NHOH, -C(NH)NHO4c, -C(=NH)NHNR4cR4d, NHCOR4c, SR4c, OR4c, SOR4c, SO2R4c, -Ct=O)NR4cR4d, -C(=S)NR4cR4d, or R4c;
R43 is H, hydroxy, NR4cR4d, NHC(=O)NR4c, NHNHR4c, (C1-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, heterocycle, aryl, aryl(C1-C6)alkyl, halo, COOR4c, CONR4cR4c, or absent when Y is N;
R43 and R4b are each independently hydrogen, (C1-C6)alkyl, (C2- C6)alkenyl, (C2-C6)alkynyl, heterocycle, or aryl; R4c, and R4d are each independently hydrogen, (C1-C6)alkyl, (C1- C6)alkenyl, (C2-C6)alkynyl, heterocycle, or aryl;
X, Y, and W are each independently N, C, CR4c, S or P;
R44 is H, hydroxy, mercapto, cyano, -SNR4cR4d, -C(NH)NR4cR4d, -C(=NH)NHOH, -C(NH)NHOR4c, -C(=NH)NHNR4cR4C NHCOR4c, SR4c, OR4c, SOR4c, SO2R4c, -C(=O)NR4cR4d, -C(=S)NR4cR4d, or R4c;
R45 is H, hydroxy, mercapto, cyano, -SNR4cR4d, -C(NH)NR4cR4d, -C(=NH)NHOH, -C(NH)NHO4c, -C(=NH)NHNR4cR4d, NHCOR4c, SR4c, OR4c, SOR4c, SO2R4c, -C(=O)NR4cR4d, -C(=S)NR4cR4d, or R4c;
R46, and R47 together with the atoms to which they are attached form a heterocyclic ring; and
U is S or O; wherein each aryl or heterocycle is optionally substituted with one or more (C1-C6)alkyl, (C1-C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1- C6)alkanoyloxy, (C1-C6)alkoxycarbonyl, cyano, halo, hydroxy, nitro, carboxy, (C3-C6)cycloalkyl, (C3-C8)cycloalkoxy, trifluoromethoxy, mercapto, or trifluoromethyl.
11. The compound of claim 1 wherein R9 has one of the following formulae:
Figure imgf000063_0001
wherein:
R50 is NR5aR5b, ONR5aR5b, NR5aNR5aR5b, SR5b, OR5b, H, hydroxy, (C1- C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, or aryl; Rsi is (C1-C6)alkyl, (C1-C6)aIkanoyl, or aryl; R53 is (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, or aryl; R53 is H, halo, (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, or aryl;
R54 is H or NH2;
R55 is NR5aR5b, ONR5aR5b, NR5aNR5aR5b, SR5b, OR5b, H, hydroxy, (C1- C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, or aryl;
R56 is H, halo, (C1-C6)alkyl, or (C1-C6)alkenyl; R57 and R58 are each independently -L-R5c; each L is independently a direct bond, -N(R5a)-, O or S; each R5a and R5b is independently H, hydroxy, (C1-C6)alkyl, (C1- C6)alkenyl, (C1-C6)alkynyl, or aryl; and each R5c is NR5aR5b, H, hydroxy, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, or aryl; wherein each (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, or aryl of R50-R58 and R5a-R5c is optionally substituted with one or more (C1-C6)alkyl, (C1- C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-C6)alkanoyloxy, (C1- C6)alkoxycarbonyl, cyano, halo, hydroxy, nitro, carboxy, (C3-C8)cycloalkyl, (C3- C8)cycloalkoxy, trifluoromethoxy, mercapto, or trifluoromethyl.
12. The compound of claim 1 wherein R9 has one of the the following formulae:
Figure imgf000064_0001
wherein:
X2, X3, and X4 are each independently N, CH, or C-R6a;
R60, R61, and R6a are each independently H, halo, NR6b R6c, hydroxyamino, NR6bNR6bR6c, N3, NO, NO2, formyl, cyano, -C(=O)NR6b R6c, -C(=S)NR6b R6c, -C(=O)OR6b, R6b, OR6b, or SR6b; and R6b, and R6c are each independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl.
13. The compound of claim 1 wherein R9 has the following formula:
Figure imgf000065_0001
wherein:
Xs1 X6, and X7, are each independently N, CH, or C-R7a;
R7O and R7a are each independently H, halo, NR7bR7c, hydroxyamino, NR7bNR7bR7c, N3, NO, NO2, formyl, cyano, -C(=O)NR7bR7c, -C(=S)NR7bR7c, -C(=O)OR7b, R?b, OR7b, or SR7b; and
R7b, and R70 are each independently H, (C1-C6)all<yl, (C1-C6)alkenyl, (C1- C6)alkynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl.
14. The compound of claim 1 wherein R9 has the following formula:
Figure imgf000065_0002
wherein:
Y1=Y2 is -N=N-, -CH=N-, -N=CR8a-, or -CH=CR83-; and each R8a is independently H, halo, or (C1-C6)alkyl.
15. The compound of claim 1 wherein R9 has one of the following formulae:
Figure imgf000066_0001
wherein: each Zi is independently N, C-R9a, O, S, NR9b, >C=O, >C=S, >C=NR9b, >S=O, >S(O)2 or CH-R9a; provided that if a Zi participates in an optional bond represented by a dotted line — in the formula, then that Zi is N or C-R9a; and provided that if a Zi does not participate in an optional bond represented by a dotted line — in the formula, then that Zi is O, S, NR%, >C=O, >C=S, >C=NR9b, >S=O, >S(O)2 or CH-R9a; X5 is O, S, SO, SO2; Se, SeO, SeO2 or NR%; each Wi is C, CH, or N; wherein if a Wi participates in an optional bond represented by a dotted line — in the formula, then that Wi is C; and if a Wi does not participate in an optional bond represented by a dotted line — in the formula, then that Wi is CH, or TSI; each R9a is independently H, halo, NR90R91J, hydroxyamino,
NR9cNR9cR9d, N3, cyano, -C(=O)NR9cR9d, -C(=S)NR9cR9d, -C(=S)NR9cR9d, -C(=NH)0R9c, R90, OR90, or SR9c; each R9b is independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (Cr C6)a]kynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl; and R9c, and R9d are each independently H, (C1-C6)alkyl, (C1-C6)alkenyl, (C1- C6)alkynyl, aryl, (C1-C6)alkanoyl, or aryl(C1-C6)alkyl.
16. The compound of claim 1 wherein R9 has one of the following formulae:
Figure imgf000067_0001
wherein:
Rg7 is H, hydroxy, mercapto., (C1-C6)alkyl, (C1-C6)alkoxy, (C2- C6)alkenyl, (C3-C8)cycloalkyl, aryl(C1-C6)alkyl, aryl or (C2-C6)alkynyl
R98 is H, hydroxy, mercapto, or (C1-C6)alkyl; R99 is H, halo, azido, cyano, nitro, OR99a, SR99a, NR99bR99C, (C1-C6)alkyl, (C2-C6)alkenyl, (C3-C8)cycloalkyl, aryl(C1-C6)alkyl, aryl or (C2-C6)alkynyl; each R99a is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each R99b and R990 is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; wherein each (C1-C6)alkyl, (C1-C6)alkoxy (C1-C6)alkenyl, (C1- C6)alkynyl, (C3-C6)cycloalkyl, aryl, and aryl(C1-C6)alkyl of R97, R98, R99 , R99a, R99b, and R99C, is optionally substituted with one or more (C1-C6)alkyl, (C1- C6)alkylthio, (C1-C6)alkoxy, (C1-C6)alkanoyl, (C1-C6)alkanoyloxy, (d- C6)alkoxycarbonyl, NH2, cyano, halo, azido, hydroxy, nitro, carboxy, trifluoromethoxy, aryl, or mercapto.
17. The compound of claim 1 wherein R9 has the following formula:
Figure imgf000067_0002
wherein: X4 is CR9Oa or N;
X5 is O, S, or NR9ia ; Rgo and R91 are each independently H, halo, hydroxy, (C1-C6)alkoxy, NRgobRgibj or heterocycle;
R9oa is H, halo, methyl, azido, or amino;
Rgia is H, or (C1-C6)alkyl; and Rgob and Rgib are each independently H, (C1-C6)alkyl, (C3-C8)cycloalkyl, or aryl(C1-C6)alkyl; wherein each (C1-C6)alkyl, (C3-C8)cycloalkyl, aryl(C1-C6)alkyl, and heterocycle of R90-R91, R9ia, and Rgob-R9ib are optionally substituted with one or more (e.g. 1, 2, 3, or 4) halo, hydroxy, amino, (C1-C6)alkyl, and (C1-C6)alkoxy.
18 The compound of claim 1 wherein R9 has the following formula:
Figure imgf000068_0001
wherein:
X6 is O, S, or NR70a; R7o and R71 are each independently H, halo, hydroxy, mercapto, aryl, heterocycle, (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, (C1-C6)alkylthio, arylthio, -S(=O)(C1-C6)alkyl, -SC=O)2(C1 -C6)alkyl, -S(=O)2NR70bR7ib, NR7ObR71b, (C1-C6)alkoxy, aryloxy, (heterocycle)oxy;
R7Oa is H, methyl, ethyl, or acetyl; and R7Ob and R7Ib are each independently H, (C1-C6)alkyl, aryl, aryl(Cr C6)alkyl, (C1-C6)alkanoyl, or aryl-C(=O)-;
19. The compound of claim 1 wherein R9 has one of the following formulae:
Figure imgf000069_0001
wherein:
A8O, B80, and Y80, are each independently H, halo, OR80, S(O)nR8O, NR80R81, cyano, trifluoromethyl, C(=W)OR80, C(=W)SR80, C(=W)NR80 R81, nitro, azido, carbocyclic, (C1-C6)alkyl, (C1-C6)alkenyl, (C1-C6)alkynyl, aryl, aryl(C1-C6)alkyl, or heterocycle; or A80 and B80 taken together with the carbon atoms to which they are attached from a 4-7 membered carbocyclic or heterocyclic ring; n is 0, 1, or 2
Z80 is O, S, NRs0, or CRS0R8!; each V is independently N or CR80; and each R80 and R81 is independently H, carbocycle, (C1-C6^IlCyI, (C1- C6)alkenyl, (C1-C6)alkynyl, halo, (CVC6)alkoxy, amino, methylamino, dimethylamino, cyano, (C1-C6)alkanoyl, aryl, aryl(C1-C6)alkyl, an amino acid residue (e.g. a natural amino acid residue) or heterocycle; or Rso and Rsi taken together with the atom(s) to which they are attached form a 3-7 membered carbocyclic or heterocyclic ring.
20. The compound of claim 1 wherein R9 is adenine, guanine, cytosine, uracil, thymine, 7-deazaadenine, 7-deazaguanine, 7-deaza-8-azaguanine, 7-deaza-8- azaadenine, inosine, nebularine, nitropyrrole, nitroindole, 2-aminopurine, 2- amino-6-chloropurine, 2,6-diaminopurine, hypoxanthine, pseudouridine, pseudocytosine, pseudoisocytosine, 5-propynylcytosine, isocytosine, isoguanine, 7-deazaguanine, 2-thiopyrimidine, 6-thioguanine, 4-thiothymine, 4-thiouraciL O^-methylguanine, N^-methyladeniiie, (94-methylthymine, 5,6-dihydrothymine, 5,6-dihydrouracil, 4-methylindole, substituted triazole, or pyrazolo[3,4- Djpyrimidine.
21. A prodrug of a compound of claim 1 werein Ri 0 is OH, or -OP(=O)(OH)2.
22. A pharmaceutical composition comprising a pharmaceutical excipient and a compound of formula I or II:
Figure imgf000070_0001
wherein:
Ri is H, halo, ORa, SRa, or ΝRbRc;
R2 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; R3 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; and R4 is
H, halo, azido, cyano, nitro, OR3, SRa, NRbRc, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; or R3 and R4 taken together are =O, =NRdRe, =CRfRg; or R3 and R4 taken together with the carbon atom to which they are attached form a 3-7 membered carbocyclic ring wherein one carbon atom can optionally be replaced with -0-, -S-, or -NRa-; R5 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; and R6 is
H, halo, azido, cyano, nitro, ORa, SRa, NRbRc, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; or R5 and R6 taken together are =O, =NRdRe, =CRfRg; or R5 and Re taken together with the carbon atom to which they are attached form a 3-7 membered carbocyclic ring wherein one carbon atom can optionally be replaced with -O-, -S-, or -NRa-;
R7 and Rg are each independently H or halo; R9 is a nucleoside base;
Rio is OH, -OP(=O)(OH)2, -OP(=O)(OH)-P(=O)(OH)2, -OP(=O)(OH)-P(=O)(OH)-P(=O)(OH)2, or a phosphonate; each R3 is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each Rb and Rc is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each Rd is independently H, (C1-C6)alkyl, or OH; each Re is independently H or (C1-C6)alkyl; and each Rf and Rg is independently H, (C1-C6)alkyl, or halo; wherein each (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl of R2-R5 and Ra-Rg is optionally substituted with one or more halo, hydroxy, or (C1- C6)alkoxy; or a pharmaceutically acceptable salt or prodrug thereof; provided the compound is not (l'S,2'i?,3'S,4'i?,5'S)-4-(6-amino-9H-purin- 9-yl)-1-[phosphoryloxymethyl]bi-cyclo[3.1.0]hexane-2,3-diol; 5-(6-amino-9H- purin-9-yl)-1-[phosphoryloxymethyl]bicyclo[3.1.0]hexane-2,3-diol; 5-(6-amino- 9H-purin-9-yl)-1-[hydoxymethyl]bicyclo[3.1.0]hexane-2,3-diol; (lW,2'i?,3'<S',4'i?,5I6}-4-(6-amino-9H-purin-9-yl)-1-[triphosphoryloxymethyl]bi- cyclo[3.1.0]hexane-2,3-diol; 5-(6-amino-9H-purin-9-yl)-l - [triphosphoryloxymethyηbicyclo^.l.OJhexane^^-diol^lW^^S'.S'^'^S'.S)^- (2,4(H,3H)-dioxopyrimidin-1-yl)-1-(phosphoryloxymethyl)bi- cyclo[3.1.0]hexane-2,3-diol; (lW,2'i?,3'5l,4ti?,5'»S)-4-(2,4(H,3H)-dioxopyrimidin- l-yl)-1- (diphosphoryloxymethyl)bicyclo[3.1.0]hexane-2,3-diol; or (lW,2y?,3W,47?,5\S)-4-(2,4(H,3H)-dioxopyrimidin~l-yl)-1- (triphosphoryloxymethyl)bicyclo[3.1.0]hexane-2,3-diol.
23. A pharmaceutical composition comprising a pharmaceutical excipient and a compound, salt, or prodrug as descrobed in any one of claims 1-22.
24. A method for treating a viral infection in an animal comprising administering to the animal an effective anti-viral amount of a compound of formula I or II:
Figure imgf000072_0001
wherein:
Ri is H, halo, ORa, SRa, or NRbRc;
R2 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C1-C6)alkynyl;
R3 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; and R4 is H, halo, azido, cyano, nitro, ORa, SRa, NRbRc, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; or R3 and R4 taken together are =O, ^NRdRe, -CRfRg; or R3 and R4 taken together with the carbon atom to which they are attached form a 3-7 membered carbocyclic ring wherein one carbon atom can optionally be replaced with -0-, -S-, or -NRa-;
R5 is H, halo, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; and R6 is H, halo, azido, cyano, nitro, OR3, SR3, NRbRc, (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl; or R5 and R6 taken together are =O, =NRdRe, =€RfRg; or R5 and Rδ taken together with the carbon atom to which they are attached form a 3-7 membered carbocyclic ring wherein one carbon atom can optionally be replaced with -Q-, -S-, or -NRaS
R7 and Re are each independently H or halo;
R9 is a nucleoside base; R10 is OH, -OP(=O)(OH)2, -OPC=O)(OH)-PC=O)(OH)2, -0P(=O)(OH)-P(=O)(OH)-P(=O)(OH)2, or a phosphonate; each Ra is independently H, (Cι-C6)alkyl, or (C1-C6)alkanoyl; each Rb and Rc is independently H, (C1-C6)alkyl, or (C1-C6)alkanoyl; each Rd is independently H, (C1-C6)alkyl, or OH; each Re is independently H or (C1-C6)alkyl; and each Rf and Rg is independently H, (C1-C6)alkyl, or halo; wherein each (C1-C6)alkyl, (C2-C6)alkenyl, or (C2-C6)alkynyl OfR2-Rs and Ra-Rg is optionally substituted with one or more halo, hydroxy, or (Cj- C6)alkoxy; or a pharmaceutically acceptable salt or prodrug thereof.
25. The method of claim 24 wherein the viral infection is Hepatitis C.
26. The use of a compound as described in any one of claims 1-22 in medical therapy.
27. The use of a compound as described in claim 26 to prepare a medicament useful for treating a viral infection in an animal.
28. The use of claim 27 wherein the viral infection is Hepatitis C.
PCT/US2006/006763 2005-02-25 2006-02-24 Bicyclo (3.1.0) hexane derivatives as antiviral compounds WO2006091905A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US65668905P 2005-02-25 2005-02-25
US60/656,689 2005-02-25

Publications (1)

Publication Number Publication Date
WO2006091905A1 true WO2006091905A1 (en) 2006-08-31

Family

ID=36602474

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/006763 WO2006091905A1 (en) 2005-02-25 2006-02-24 Bicyclo (3.1.0) hexane derivatives as antiviral compounds

Country Status (1)

Country Link
WO (1) WO2006091905A1 (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110046166A1 (en) * 2008-03-31 2011-02-24 Office of Technology Transfer, NIH Purine derivatives as a3 adenosine receptor-selective agonists
US8343983B2 (en) 2009-03-31 2013-01-01 Arqule, Inc. Substituted pyrazolo-pyrimidine compounds
US8623879B2 (en) 2008-04-02 2014-01-07 Boehringer Ingelheim International Gmbh 1-heterocyclyl-1,5-dihydro-pyrazolo[3,4-D] pyrimidin-4-one derivates and their use as PDE9A modulators
US8623901B2 (en) 2009-03-31 2014-01-07 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
US8648085B2 (en) 2007-11-30 2014-02-11 Boehringer Ingelheim International Gmbh 1, 5-dihydro-pyrazolo (3, 4-D) pyrimidin-4-one derivatives and their use as PDE9A mudulators for the treatment of CNS disorders
US8796291B2 (en) 2008-08-01 2014-08-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A3 adenosine receptor antagonists and partial agonists
US8809345B2 (en) 2011-02-15 2014-08-19 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
US8822434B2 (en) 2010-02-22 2014-09-02 The University Of Connecticut Compositions and methods to treat cardiac diseases
US8912201B2 (en) 2010-08-12 2014-12-16 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
US8916570B2 (en) 2008-03-31 2014-12-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A3 adenosine receptor agonists and antagonists
US9067945B2 (en) 2002-08-23 2015-06-30 Boehringer Ingehleim International GmbH Selective phosphodiesterase 9A inhibitors as medicaments for improving cognitive processes
US9079905B2 (en) 2008-09-08 2015-07-14 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
WO2015142910A1 (en) * 2014-03-17 2015-09-24 Isis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
US9181253B2 (en) 2008-08-01 2015-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Adenosine receptor agonists, partial agonists, and antagonists
US9789131B1 (en) 2016-04-21 2017-10-17 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating neurological and cardiovascular conditions
WO2017205848A1 (en) * 2016-05-27 2017-11-30 Legacy Emanuel Hospital & Health Center Methanocarba derivatives of pseudoribose that inhibit adenosine kinase
WO2019116302A1 (en) 2017-12-13 2019-06-20 Lupin Limited Substituted bicyclic heterocyclic compounds as prmt5 inhibitors
WO2020018670A1 (en) * 2018-07-17 2020-01-23 Board Of Regents, The University Of Texas System Compounds useful as inhibitors of indoleamine 2,3-dioxygenase and/or tryptophan dioxygenase
WO2020069068A1 (en) * 2018-09-26 2020-04-02 Astrocyte Pharmaceuticals, Inc. Polymorphic compounds and uses thereof
WO2020250123A1 (en) 2019-06-10 2020-12-17 Lupin Limited Prmt5 inhibitors
US11173145B2 (en) 2017-01-17 2021-11-16 Board Of Regents, The University Of Texas System Compounds useful as inhibitors of indoleamine 2,3-dioxygenase and/or tryptophan dioxygenase
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11787833B2 (en) 2019-05-09 2023-10-17 Aligos Therapeutics, Inc. Modified cyclic dinucleoside compounds as sting modulators
US11839615B2 (en) 2018-02-09 2023-12-12 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating addiction and related disorders

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0577558A2 (en) * 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
WO1995008541A1 (en) * 1993-09-24 1995-03-30 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Conformationally locked nucleoside analogues
WO1998005662A1 (en) * 1996-08-07 1998-02-12 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Conformationally locked nucleoside analogs as antiherpetic agents
WO2001051490A1 (en) * 2000-01-14 2001-07-19 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Methanocarba cycloalkyl nucleoside analogues
WO2002008204A2 (en) * 2000-07-26 2002-01-31 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 5-substituted pyrimidine derivatives of conformationally locked nucleoside analogues

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0577558A2 (en) * 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
WO1995008541A1 (en) * 1993-09-24 1995-03-30 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Conformationally locked nucleoside analogues
US5869666A (en) * 1993-09-24 1999-02-09 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Conformationally locked nucleoside analogues
WO1998005662A1 (en) * 1996-08-07 1998-02-12 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Conformationally locked nucleoside analogs as antiherpetic agents
WO2001051490A1 (en) * 2000-01-14 2001-07-19 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Methanocarba cycloalkyl nucleoside analogues
WO2002008204A2 (en) * 2000-07-26 2002-01-31 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 5-substituted pyrimidine derivatives of conformationally locked nucleoside analogues

Non-Patent Citations (27)

* Cited by examiner, † Cited by third party
Title
ALTMANN K-H ET AL: "4',6'-METHANO CARBOCYCLIC THYMIDINE: A CONFORMATIONALLY CONSTRAINEDBUILDING BLOCK FOR OILGONUCLEOTIDES", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 35, no. 15, 1994, pages 2331 - 2334, XP000996351, ISSN: 0040-4039 *
BIN XU ET AL: "Acyclic Analogues of Adenosine Biphosphates as P2Y Receptor Antagonists: Phosphate Substitution Leads to Multiple Pathways of Inhibition of Platelet Aggregation", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 45, 2002, pages 5694 - 5709, XP002285317, ISSN: 0022-2623 *
CHHATRIWALA MARIYA ET AL: "Induction of novel agonist selectivity for the ADP-activated P2Y1 receptor versus the ADP-activated P2Y12 and P2Y13 receptors by conformational constraint of an ADP analog", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 311, no. 3, December 2004 (2004-12-01), pages 1038 - 1043, XP002388424, ISSN: 0022-3565 *
CHOI YONGSEOK ET AL: "Synthesis of a conformationally locked version of puromycin amino nucleoside", ORGANIC LETTERS, vol. 4, no. 4, 21 February 2002 (2002-02-21), pages 589 - 592, XP002388442, ISSN: 1523-7060 *
COSTANZI STEFANO ET AL: "Human P2Y(6) receptor: Molecular modeling leads to the rational design of a novel agonist based on a unique conformational preference", JOURNAL OF MEDICINAL CHEMISTRY, vol. 48, no. 26, December 2005 (2005-12-01), pages 8108 - 8111, XP002388449, ISSN: 0022-2623 *
EZZITOUNI A ET AL: "CONFORMATIONALLY LOCKED CARBOCYCLIC NUCLEOSIDES BUILT ON A BICYCLO3.1.0HEXANE TEMPLATE WITH A FIXED SOUTHERN CONFORMATION SYNTHESIS AND ANTIVIRAL ACTIVITY", JOURNAL OF THE CHEMICAL SOCIETY, PERKIN TRANSACTIONS 1, CHEMICAL SOCIETY. LETCHWORTH, GB, vol. 1, 1997, pages 1073 - 1078, XP000996608, ISSN: 0300-922X *
HERNANDEZ SUSANA ET AL: "Is the anomeric effect an important factor in the rate of adenosine deaminase catalyzed hydrolysis of purine nucleosides? A direct comparison of nucleoside analogues constructed on ribose and carbocyclic templates with equivalent heterocyclic bases selected to promote hydration", BIOORGANIC AND MEDICINAL CHEMISTRY, vol. 10, no. 8, August 2002 (2002-08-01), pages 2723 - 2730, XP002388428, ISSN: 0968-0896 *
JACOBSON KENNETH A ET AL: "Methanocarba analogues of purine nucleosides as potent and selective adenosine receptor agonists", JOURNAL OF MEDICINAL CHEMISTRY, vol. 43, no. 11, 1 June 2000 (2000-06-01), pages 2196 - 2203, XP002388444, ISSN: 0022-2623 *
JEONG LAK SHIN ET AL: "Carbocyclic analogues of the potent cytidine deaminase inhibitor 1-(beta-D-ribofuranosyl)-1,2-dihydropyrimidine-2-one (zebularine)", JOURNAL OF MEDICINAL CHEMISTRY, vol. 41, no. 14, 2 July 1998 (1998-07-02), pages 2572 - 2578, XP002388446, ISSN: 0022-2623 *
KIM HAK SUNG ET AL: "2-substitution of adenine nucleotide analogues containing a bicyclo(3.1.0)hexane ring system locked in a northern conformation: Enhanced potency as P2Y1 receptor antagonists.", JOURNAL OF MEDICINAL CHEMISTRY, vol. 46, no. 23, 6 November 2003 (2003-11-06), pages 4974 - 4987, XP002388426, ISSN: 0022-2623 *
KIM HAK SUNG ET AL: "Methanocarba modification of uracil and adenine nucleotides: High potency of Northern ring conformation at P2Y1, P2Y2, P2Y4, and P2Y11 but not P2Y6 receptors", JOURNAL OF MEDICINAL CHEMISTRY, vol. 45, no. 1, 3 January 2002 (2002-01-03), pages 208 - 218, XP002388443, ISSN: 0022-2623 *
LEE K ET AL: "Ring-constrained (N)-methanocarba nucleosides as adenosine receptor agonists: independent 5'-uronamide and 2'-deoxy modifications", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, OXFORD, GB, vol. 11, 2001, pages 1333 - 1337, XP002362940, ISSN: 0960-894X *
LEE K ET AL: "Synthesis using ring closure metathesis and effect on nucleoside transport of a (N)-methanocarba S-(4-nitrobenzyl)thioinosine derivative.", ORGANIC LETTERS. 22 FEB 2001, vol. 3, no. 4, 22 February 2001 (2001-02-22), pages 597 - 599, XP002388447, ISSN: 1523-7060 *
MAIER MARTIN A ET AL: "Synthesis and characterization of oligonucleotides containing conformationally constrained bicyclo(3.1.0)hexane pseudosugar analogs", NUCLEIC ACIDS RESEARCH, vol. 32, no. 12, July 2004 (2004-07-01), pages 3642 - 3650, XP002388425, ISSN: 0305-1048 *
MARQUEZ V E ET AL: "CONFORMATIONALLY RESTRICTED NUCLEOSIDES. THE REACTION OF ADENOSINE DEAMINASE WITH SUBSTRATES BUILT ON A BICYCLO3.1.0HEXANE TEMPLATE", NUCLEOSIDES & NUCLEOTIDES, MARCEL DEKKER, INC, US, vol. 18, no. 4/5, 1999, pages 521 - 530, XP000996270, ISSN: 0732-8311 *
MARQUEZ V E ET AL: "HIV-1 REVERSE TRANSCRIPTASE CAN DISCRIMINATE BETWEEN TWO CONFORMATIONALLY LOCKED CARBOCYCLIC AZT TRIPHOSPHATE ANALOGUES", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, DC, US, vol. 120, no. 12, 1998, pages 2780 - 2789, XP000996607, ISSN: 0002-7863 *
MARQUEZ V E ET AL: "Nucleosides with a Twist. Can Fixed Forms of Sugar Ring Pucker Influence Biological Activity in Nucleosides and Oligonucleotides ?", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 39, 13 September 1996 (1996-09-13), pages 3739 - 3747, XP002094300, ISSN: 0022-2623 *
MARQUEZ VICTOR E ET AL: "Experimental and structural evidence that herpes 1 kinase and cellular DNA polymerase(s) discriminate on the basis of sugar pucker.", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 126, no. 2, 21 January 2004 (2004-01-21), pages 543 - 549, XP002388423, ISSN: 0002-7863 *
MOON H R ET AL: "A remarkably simple chemicoenzymatic approach to structurally complex bicyclo[3.1.0]hexane carbocyclic nucleosides.", ORGANIC LETTERS. 30 NOV 2000, vol. 2, no. 24, 30 November 2000 (2000-11-30), pages 3793 - 3796, XP002388448, ISSN: 1523-7060 *
NANDANAN ERATHODIYIL ET AL: "Synthesis, biological activity, and molecular modeling of ribose-modified deoxyadenosine bisphosphate analogues as P2Y1 receptor ligands", JOURNAL OF MEDICINAL CHEMISTRY, vol. 43, no. 5, 9 March 2000 (2000-03-09), pages 829 - 842, XP002388445, ISSN: 0022-2623 *
RAVI G ET AL: "Synthesis and purine receptor affinity of 6-oxopurine nucleosides and nucleotides containing (N)-methanocarba-pseudoribose rings", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, OXFORD, GB, vol. 11, 2001, pages 2295 - 2300, XP002362941, ISSN: 0960-894X *
RAVI R GNANA ET AL: "Adenine nucleotide analogues locked in a Northern methanocarba conformation: Enhanced stability and potency as P2Y1 receptor agonists", JOURNAL OF MEDICINAL CHEMISTRY, vol. 45, no. 10, 9 May 2002 (2002-05-09), pages 2090 - 2100, XP002388429, ISSN: 0022-2623 *
RODRIGUEZ J B ET AL: "CONFORMATIONALLY LOCKED NUCLEOSIDE ANALOGUES. SYNTHESIS OF DIDEOXYCARBOCYCLIC NUCLEOSIDE ANALOGUES STRUCTURALLY RELATED TO NEPLANOCIN C", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 37, 1994, pages 3389 - 3399, XP000996507, ISSN: 0022-2623 *
RUSS PAMELA ET AL: "Synthesis and biological evaluation of 5-substituted derivatives of the potent antiherpes agent (north)-methanocarbathymine.", JOURNAL OF MEDICINAL CHEMISTRY, vol. 46, no. 23, 6 November 2003 (2003-11-06), pages 5045 - 5054, XP002388422, ISSN: 0022-2623 *
SIDDIQUI M A ET AL: "SYNTHESIS, CONFORMATIONAL ANALYSIS, AND BIOLOGICAL ACTIVITY OF A RIGID CARBOCYCLIC ANALOGUE OF 2'-DEOXYARISTEROMYCIN BUILT ON A BICYCLOÚ3.1.0HEXANE TEMPLATE", NUCLEOSIDES & NUCLEOTIDES, MARCEL DEKKER, INC, US, vol. 15, no. 1/3, 1996, pages 235 - 250, XP000615548, ISSN: 0732-8311 *
TCHILIBON S ET AL: "(N)-Methanocarba 2,N6-disubstituted adenine nucleoside as highly potent and selective A3 adenosine receptor agonists", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 48, 23 September 2004 (2004-09-23), pages 1745 - 1758, XP002362942, ISSN: 0022-2623 *
WALDO GARY L ET AL: "Quantitation of the P2Y1 receptor with a high affinity radiolabeled antagonist.", MOLECULAR PHARMACOLOGY, vol. 62, no. 5, November 2002 (2002-11-01), pages 1249 - 1257, XP002388427, ISSN: 0026-895X *

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9067945B2 (en) 2002-08-23 2015-06-30 Boehringer Ingehleim International GmbH Selective phosphodiesterase 9A inhibitors as medicaments for improving cognitive processes
US8648085B2 (en) 2007-11-30 2014-02-11 Boehringer Ingelheim International Gmbh 1, 5-dihydro-pyrazolo (3, 4-D) pyrimidin-4-one derivatives and their use as PDE9A mudulators for the treatment of CNS disorders
US8916570B2 (en) 2008-03-31 2014-12-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A3 adenosine receptor agonists and antagonists
US20110046166A1 (en) * 2008-03-31 2011-02-24 Office of Technology Transfer, NIH Purine derivatives as a3 adenosine receptor-selective agonists
US8735407B2 (en) * 2008-03-31 2014-05-27 The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Purine derivatives as A3 adenosine receptor-selective agonists
US8623879B2 (en) 2008-04-02 2014-01-07 Boehringer Ingelheim International Gmbh 1-heterocyclyl-1,5-dihydro-pyrazolo[3,4-D] pyrimidin-4-one derivates and their use as PDE9A modulators
US9096603B2 (en) 2008-04-02 2015-08-04 Boehringer Ingelheim International Gmbh 1-heterocyclyl-1,5-dihydro-pyrazolo[3,4-D] pyrimidin-4-one derivatives and their use as PDE9A modulators
US8796291B2 (en) 2008-08-01 2014-08-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A3 adenosine receptor antagonists and partial agonists
US9181253B2 (en) 2008-08-01 2015-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Adenosine receptor agonists, partial agonists, and antagonists
US9079905B2 (en) 2008-09-08 2015-07-14 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
US8343983B2 (en) 2009-03-31 2013-01-01 Arqule, Inc. Substituted pyrazolo-pyrimidine compounds
US9102679B2 (en) 2009-03-31 2015-08-11 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
US8623901B2 (en) 2009-03-31 2014-01-07 Boehringer Ingelheim International Gmbh Compounds for the treatment of CNS disorders
US8822434B2 (en) 2010-02-22 2014-09-02 The University Of Connecticut Compositions and methods to treat cardiac diseases
US9526739B2 (en) 2010-02-22 2016-12-27 University Of Connecticut Compositions and methods to treat cardiac diseases
US9303053B2 (en) 2010-02-22 2016-04-05 University Of Connecticut Compositions and methods to treat cardiac diseases
US8912201B2 (en) 2010-08-12 2014-12-16 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
US9328120B2 (en) 2010-08-12 2016-05-03 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
US8809345B2 (en) 2011-02-15 2014-08-19 Boehringer Ingelheim International Gmbh 6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders
WO2015142910A1 (en) * 2014-03-17 2015-09-24 Isis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
US10036019B2 (en) 2014-03-17 2018-07-31 Ionis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
IL262487A (en) * 2016-04-21 2018-12-31 Univ Texas Compounds and methods for treating neurological and cardiovascular conditions
US11484545B2 (en) * 2016-04-21 2022-11-01 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating neurological and cardiovascular conditions
CN109310695B (en) * 2016-04-21 2022-04-01 阿斯特罗赛特制药公司 Compounds and methods for treating neurological and cardiovascular conditions
WO2017185061A1 (en) 2016-04-21 2017-10-26 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating neurological and cardiovascular conditions
CN109310695A (en) * 2016-04-21 2019-02-05 阿斯特罗赛特制药公司 The Compounds and methods for for treating nerve and the cardiovascular patient's condition
US10265338B2 (en) 2016-04-21 2019-04-23 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating neurological and cardiovascular conditions
JP2019515920A (en) * 2016-04-21 2019-06-13 アストロサイト ファーマシューティカルズ, インコーポレイテッド Compounds and methods for treating neurological and cardiovascular conditions
US10953031B2 (en) 2016-04-21 2021-03-23 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating neurological and cardiovascular conditions
JP7109005B2 (en) 2016-04-21 2022-07-29 アストロサイト ファーマシューティカルズ, インコーポレイテッド Compounds and methods for treating neurological and cardiovascular conditions
US9789131B1 (en) 2016-04-21 2017-10-17 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating neurological and cardiovascular conditions
US10016432B2 (en) 2016-05-27 2018-07-10 Legacy Emanuel Hospital & Health Center Methanocarba derivatives of pseudoribose that inhibit adenosine kinase
WO2017205848A1 (en) * 2016-05-27 2017-11-30 Legacy Emanuel Hospital & Health Center Methanocarba derivatives of pseudoribose that inhibit adenosine kinase
US11173145B2 (en) 2017-01-17 2021-11-16 Board Of Regents, The University Of Texas System Compounds useful as inhibitors of indoleamine 2,3-dioxygenase and/or tryptophan dioxygenase
US11903959B2 (en) 2017-12-07 2024-02-20 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2019116302A1 (en) 2017-12-13 2019-06-20 Lupin Limited Substituted bicyclic heterocyclic compounds as prmt5 inhibitors
US11459330B2 (en) 2017-12-13 2022-10-04 Lupin Limited Substituted bicyclic heterocyclic compounds as PRMT5 inhibitors
US11952380B2 (en) 2017-12-13 2024-04-09 Lupin Limited Substituted bicyclic heterocyclic compounds as PRMT5 inhibitors
US11839615B2 (en) 2018-02-09 2023-12-12 Astrocyte Pharmaceuticals, Inc. Compounds and methods for treating addiction and related disorders
US11046649B2 (en) 2018-07-17 2021-06-29 Board Of Regents, The University Of Texas System Compounds useful as inhibitors of indoleamine 2,3-dioxygenase and/or tryptophan dioxygenase
WO2020018670A1 (en) * 2018-07-17 2020-01-23 Board Of Regents, The University Of Texas System Compounds useful as inhibitors of indoleamine 2,3-dioxygenase and/or tryptophan dioxygenase
JP2022502405A (en) * 2018-09-26 2022-01-11 アストロサイト ファーマシューティカルズ, インコーポレイテッド Polymorphic compounds and their use
US11324769B2 (en) 2018-09-26 2022-05-10 Astrocyte Pharmaceuticals, Inc. Polymorphic compounds and uses thereof
CN112930349A (en) * 2018-09-26 2021-06-08 阿斯特罗赛特制药公司 Polymorphic compounds and uses thereof
US10765693B2 (en) 2018-09-26 2020-09-08 Astrocyte Pharmaceuticals, Inc. Polymorphic compounds and uses thereof
WO2020069068A1 (en) * 2018-09-26 2020-04-02 Astrocyte Pharmaceuticals, Inc. Polymorphic compounds and uses thereof
US11787833B2 (en) 2019-05-09 2023-10-17 Aligos Therapeutics, Inc. Modified cyclic dinucleoside compounds as sting modulators
WO2020250123A1 (en) 2019-06-10 2020-12-17 Lupin Limited Prmt5 inhibitors

Similar Documents

Publication Publication Date Title
WO2006091905A1 (en) Bicyclo (3.1.0) hexane derivatives as antiviral compounds
US7645747B2 (en) Therapeutic phosphonate compounds
EP1628685B1 (en) Antiviral phosphonate analogs
EP1778249B1 (en) Phosphonate prodrugs of a 2&#39;-fluoro-2&#39;,3&#39;-didehydro-2&#39;,3&#39;-dideoxyadenosine analogue as anti-hiv agents
US8324179B2 (en) Nucleoside analogs for antiviral treatment
CA2804840C (en) Methods and compounds for treating paramyxoviridae virus infections
US20060046981A1 (en) Anti-nonmelanoma carcinoma compounds, compositions, and methods of use thereof
CA2807584A1 (en) 2&#39;-fluoro substituted carba-nucleoside analogs for antiviral treatment
AU2004260789B2 (en) Nucleobase phosphonate analogs for antiviral treatment
AU2004309418B2 (en) 4&#39;-substituted carbovir-and abacavir-derivatives as well as related compounds with HIV and HCV antiviral activity
KR20200108330A (en) Phosphor (phosphorone) amidatacetal and phosph (one) atalcetal compounds
US10266558B2 (en) Macroheterocyclic nucleoside derivatives and their analogues, production and use thereof
ES2357770T3 (en) ANTIVIRAL PHOSPHONATE ANALOGS.
JPH04505920A (en) therapeutic nucleosides
WO2002098397A2 (en) Therapeutic compositions for modulating the immune response in a mammal and use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 06736144

Country of ref document: EP

Kind code of ref document: A1