Pharmaceutical combinations comprising a thionucleotide analog

Information

  • Patent Grant
  • 9012427
  • Patent Number
    9,012,427
  • Date Filed
    Tuesday, March 12, 2013
    11 years ago
  • Date Issued
    Tuesday, April 21, 2015
    9 years ago
Abstract
Disclosed herein are pharmaceutical compositions that can include a thionucleotide analog for use in combination therapy with other agents. Also disclosed herein are methods of treating diseases and/or conditions with a pharmaceutical composition that can include a thionucleotide analog in combination therapy with other agents.
Description
BACKGROUND

1. Field


The present application relates to the fields of chemistry, biochemistry and medicine. More particularly, disclosed herein are a thiophosphoroamidate nucleotide analog for use in combination therapy with one or more other agents. Also disclosed herein are methods of treating diseases and/or conditions with a thiophosphoroamidate nucleotide analog in combination with one or more agents.


2. Description


Nucleoside analogs are a class of compounds that have been shown to exert antiviral and anticancer activity both in vitro and in vivo, and thus, have been the subject of widespread research for the treatment of viral infections and cancer. Nucleoside analogs are usually therapeutically inactive compounds that are converted by host or viral enzymes to their respective active metabolites, which, in turn, may inhibit polymerases involved in viral or cell proliferation. The activation occurs by a variety of mechanisms, such as the addition of one or more phosphate groups and, or in combination with, other metabolic processes.


SUMMARY

Some embodiments disclosed herein relate to a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds.


Some embodiments disclosed herein relate to methods of ameliorating and/or treating a viral infection (for example, a hepatitis C viral infection) that can include administering to a subject suffering from the viral infection an effective amount of a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds. Other embodiments described herein relate to using a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds, in the manufacture of a medicament for ameliorating and/or treating a viral infection (for example, a hepatitis C viral infection). Still other embodiments described herein relate to a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds, that can be used for ameliorating and/or treating a viral infection (for example, a hepatitis C viral infection).


Some embodiments disclosed herein relate to methods of ameliorating and/or treating a viral infection (for example, a hepatitis C viral infection) that can include contacting a cell infected with the virus (such as the hepatitis C virus) with an effective amount of a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds. Other embodiments described herein relate to using one or more compounds described herein, or a pharmaceutically acceptable salt of one or more compounds described herein, in the manufacture of a medicament for ameliorating and/or treating a viral infection (for example, a hepatitis C viral infection) that can include contacting a cell infected with the virus (such as the hepatitis C virus) with an effective amount of a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds. Still other embodiments described herein relate to one or more compounds described herein, or a pharmaceutically acceptable salt of one or more compounds described herein, that can be used for ameliorating and/or treating a viral infection (for example, a hepatitis C viral infection) by contacting a cell infected with the virus (such as the hepatitis C virus) with an effective amount of a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds.


Some embodiments disclosed herein relate to methods of inhibiting replication of a virus that can include contacting a cell infected with the virus (for example, a hepatitis C virus) with an effective amount of a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds. Other embodiments described herein relate to using one or more compounds described herein, or a pharmaceutically acceptable salt of one or more compounds described herein, in the manufacture of a medicament for inhibiting replication of a virus (for example, a hepatitis C virus) that can include contacting a cell infected with the virus with an effective amount of a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds. Still other embodiments described herein relate to one or more compounds described herein, or a pharmaceutically acceptable salt of one or more compound described herein, that can be used for inhibiting replication of a virus (for example, a hepatitis C virus) by contacting a cell infected with the virus with an effective amount of an effective amount of a combination of a compound of Formula (A) and one or more compounds of Formula (C), or pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compounds.


Some embodiments described herein relate to a method of inhibiting a polymerase (for example, NS5B polymerase of a hepatitis C virus) that can include contacting a cell (for example, a cell infected with a hepatitis C virus) with an effective amount of a combination of one or more compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for inhibiting a polymerase (for example, NS5B polymerase of a hepatitis C virus) that can include contacting a cell (for example, a cell infected with a hepatitis C virus) with an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for inhibiting a polymerase (for example, NS5B polymerase of a hepatitis C virus) can include contacting a cell (for example, a cell infected with a hepatitis C virus) that with an effective amount of said combination of compounds.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows example compounds of Formula (A), including Compounds 1000 through 1032.



FIG. 2 shows example compounds of Formula (C), including Compounds 3000 through 3067, which include HCV protease inhibitors, nucleoside HCV polymerase inhibitors, non-nucleoside HCV polymerase inhibitors, NS5A inhibitors, and other antivirals.





DETAILED DESCRIPTION

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. All patents, applications, published applications and other publications referenced herein are incorporated by reference in their entirety unless stated otherwise. In the event that there are a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.


As used herein, any “R” group(s) such as, without limitation, R, R1, R2, R3a, R3b, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, R16, R17, R1A, R2A, R3A, R3B, R4A, R5A, R6A, R7A, R8A and R″ represent substituents that can be attached to the indicated atom. An R group may be substituted or unsubstituted. If two “R” groups are described as being “taken together” the R groups and the atoms they are attached to can form a cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl or heterocycle. For example, without limitation, if Ra and Rb of an NRaRb group are indicated to be “taken together,” it means that they are covalently bonded to one another to form a ring:




embedded image



In addition, if two “R” groups are described as being “taken together” with the atom(s) to which they are attached to form a ring as an alternative, the R groups are not limited to the variables or substituents defined previously.


Whenever a group is described as being “optionally substituted” that group may be unsubstituted or substituted with one or more of the indicated substituents. Likewise, when a group is described as being “unsubstituted or substituted” if substituted, the substituent(s) may be selected from one or more the indicated substituents. If no substituents are indicated, it is meant that the indicated “optionally substituted” or “substituted” group may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, heteroaralkyl, (heteroalicyclyl)alkyl, hydroxy, protected hydroxyl, alkoxy, aryloxy, acyl, mercapto, alkylthio, arylthio, cyano, halogen, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, protected C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, an amino, a mono-substituted amino group and a di-substituted amino group, and protected derivatives thereof.


As used herein, “Ca to Cb” in which “a” and “b” are integers refer to the number of carbon atoms in an alkyl, alkenyl or alkynyl group, or the number of carbon atoms in the ring of a cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl or heteroalicyclyl group. That is, the alkyl, alkenyl, alkynyl, ring of the cycloalkyl, ring of the cycloalkenyl, ring of the cycloalkynyl, ring of the aryl, ring of the heteroaryl or ring of the heteroalicyclyl can contain from “a” to “b”, inclusive, carbon atoms. Thus, for example, a “C1 to C4 alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH3—, CH3CH2—, CH3CH2CH2—, (CH3)2CH—, CH3CH2CH2CH2—, CH3CH2CH(CH3)— and (CH3)3C—. If no “a” and “b” are designated with regard to an alkyl, alkenyl, alkynyl, cycloalkyl cycloalkenyl, cycloalkynyl, aryl, heteroaryl or heteroalicyclyl group, the broadest range described in these definitions is to be assumed.


As used herein, “alkyl” refers to a straight or branched hydrocarbon chain that comprises a fully saturated (no double or triple bonds) hydrocarbon group. The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 6 carbon atoms. The alkyl group of the compounds may be designated as “C1-C4 alkyl” or similar designations. By way of example only, “C1-C4 alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl and hexyl. The alkyl group may be substituted or unsubstituted.


As used herein, “alkenyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. An alkenyl group may be unsubstituted or substituted.


As used herein, “alkynyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. An alkynyl group may be unsubstituted or substituted.


As used herein, “cycloalkyl” refers to a completely saturated (no double or triple bonds) mono- or multi-cyclic hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused fashion. Cycloalkyl groups can contain 3 to 10 atoms in the ring(s) or 3 to 8 atoms in the ring(s). A cycloalkyl group may be unsubstituted or substituted. Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.


As used herein, “cycloalkenyl” refers to a mono- or multi-cyclic hydrocarbon ring system that contains one or more double bonds in at least one ring; although, if there is more than one, the double bonds cannot form a fully delocalized pi-electron system throughout all the rings (otherwise the group would be “aryl,” as defined herein). When composed of two or more rings, the rings may be connected together in a fused fashion. A cycloalkenyl group may be unsubstituted or substituted.


As used herein, “cycloalkynyl” refers to a mono- or multi-cyclic hydrocarbon ring system that contains one or more triple bonds in at least one ring. If there is more than one triple bond, the triple bonds cannot form a fully delocalized pi-electron system throughout all the rings. When composed of two or more rings, the rings may be joined together in a fused fashion. A cycloalkynyl group may be unsubstituted or substituted.


As used herein, “aryl” refers to a carbocyclic (all carbon) monocyclic or multicyclic aromatic ring system (including fused ring systems where two carbocyclic rings share a chemical bond) that has a fully delocalized pi-electron system throughout all the rings. The number of carbon atoms in an aryl group can vary. For example, the aryl group can be a C6-C14 aryl group, a C6-C10 aryl group, or a C6 aryl group. Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene. An aryl group may be substituted or unsubstituted.


As used herein, “heteroaryl” refers to a monocyclic or multicyclic aromatic ring system (a ring system with fully delocalized pi-electron system) that contain(s) one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur. The number of atoms in the ring(s) of a heteroaryl group can vary. For example, the heteroaryl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s). Furthermore, the term “heteroaryl” includes fused ring systems where two rings, such as at least one aryl ring and at least one heteroaryl ring, or at least two heteroaryl rings, share at least one chemical bond. Examples of heteroaryl rings include, but are not limited to, furan, furazan, thiophene, benzothiophene, phthalazine, pyrrole, oxazole, benzoxazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, thiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, benzothiazole, imidazole, benzimidazole, indole, indazole, pyrazole, benzopyrazole, isoxazole, benzoisoxazole, isothiazole, triazole, benzotriazole, thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, purine, pteridine, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline, and triazine. A heteroaryl group may be substituted or unsubstituted.


As used herein, “heterocyclyl” or “heteroalicyclyl” refers to a three-, four-, five-, six-, seven-, eight-, nine-, ten-, up to 18-membered monocyclic, bicyclic, and tricyclic ring system wherein carbon atoms together with from 1 to 5 heteroatoms constitute said ring system. A heterocycle may optionally contain one or more unsaturated bonds situated in such a way, however, that a fully delocalized pi-electron system does not occur throughout all the rings. The heteroatom(s) is an element other than carbon including, but not limited to, oxygen, sulfur, and nitrogen. A heterocycle may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio-systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic carbamates. When composed of two or more rings, the rings may be joined together in a fused fashion. Additionally, any nitrogens in a heteroalicyclic may be quaternized. Heterocyclyl or heteroalicyclic groups may be unsubstituted or substituted. Examples of such “heterocyclyl” or “heteroalicyclyl” groups include but are not limited to, 1,3-dioxin, 1,3-dioxane, 1,4-dioxane, 1,2-dioxolane, 1,3-dioxolane, 1,4-dioxolane, 1,3-oxathiane, 1,4-oxathiin, 1,3-oxathiolane, 1,3-dithiole, 1,3-dithiolane, 1,4-oxathiane, tetrahydro-1,4-thiazine, 2H-1,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, trioxane, hexahydro-1,3,5-triazine, imidazoline, imidazolidine, isoxazoline, isoxazolidine, oxazoline, oxazolidine, oxazolidinone, thiazoline, thiazolidine, morpholine, oxirane, piperidine N-Oxide, piperidine, piperazine, pyrrolidine, pyrrolidone, pyrrolidione, 4-piperidone, pyrazoline, pyrazolidine, 2-oxopyrrolidine, tetrahydropyran, 4H-pyran, tetrahydrothiopyran, thiamorpholine, thiamorpholine sulfoxide, thiamorpholine sulfone, and their benzo-fused analogs (e.g., benzimidazolidinone, tetrahydroquinoline, and 3,4-methylenedioxyphenyl).


As used herein, “aralkyl” and “aryl(alkyl)” refer to an aryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, 2-phenylalkyl, 3-phenylalkyl, and naphthylalkyl.


As used herein, “heteroaralkyl” and “heteroaryl(alkyl)” refer to a heteroaryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2-thienylalkyl, 3-thienylalkyl, furylalkyl, thienylalkyl, pyrrolylalkyl, pyridylalkyl, isoxazolylalkyl, and imidazolylalkyl, and their benzo-fused analogs.


A “(heteroalicyclyl)alkyl” and “(heterocyclyl)alkyl” refer to a heterocyclic or a heteroalicyclylic group connected, as a substituent, via a lower alkylene group. The lower alkylene and heterocyclyl of a (heteroalicyclyl)alkyl may be substituted or unsubstituted. Examples include but are not limited tetrahydro-2H-pyran-4-yl)methyl, (piperidin-4-yl)ethyl, (piperidin-4-yl)propyl, (tetrahydro-2H-thiopyran-4-yl)methyl, and (1,3-thiazinan-4-yl)methyl.


“Lower alkylene groups” are straight-chained —CH2— tethering groups, forming bonds to connect molecular fragments via their terminal carbon atoms. Examples include but are not limited to methylene (—CH2—), ethylene (—CH2CH2—), propylene (—CH2CH2CH2—), and butylene (—CH2CH2CH2CH2—). A lower alkylene group can be substituted by replacing one or more hydrogen of the lower alkylene group with a substituent(s) listed under the definition of “substituted.”


As used herein, “alkoxy” refers to the formula —OR wherein R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, a cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl as defined herein. A non-limiting list of alkoxys are methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, phenoxy and benzoxy. An alkoxy may be substituted or unsubstituted.


As used herein, “acyl” refers to a hydrogen, alkyl, alkenyl, alkynyl, or aryl connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl, and acryl. An acyl may be substituted or unsubstituted.


As used herein, “hydroxyalkyl” refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a hydroxy group. Exemplary hydroxyalkyl groups include but are not limited to, 2-hydroxyethyl, 3-hydroxypropyl, 2-hydroxypropyl, and 2,2-dihydroxyethyl. A hydroxyalkyl may be substituted or unsubstituted.


As used herein, “haloalkyl” refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkyl, di-haloalkyl and tri-haloalkyl). Such groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1-chloro-2-fluoromethyl, and 2-fluoroisobutyl. A haloalkyl may be substituted or unsubstituted.


As used herein, “haloalkoxy” refers to an alkoxy group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkoxy, di-haloalkoxy and tri-haloalkoxy). Such groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, 1-chloro-2-fluoromethoxy, and 2-fluoroisobutoxy. A haloalkoxy may be substituted or unsubstituted.


As used herein, “arylthio” refers to RS—, in which R is an aryl, such as but not limited to phenyl. An arylthio may be substituted or unsubstituted.


A “sulfenyl” group refers to an “—SR” group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. A sulfenyl may be substituted or unsubstituted.


A “sulfinyl” group refers to an “—S(═O)—R” group in which R can be the same as defined with respect to sulfenyl. A sulfinyl may be substituted or unsubstituted.


A “sulfonyl” group refers to an “SO2R” group in which R can be the same as defined with respect to sulfenyl. A sulfonyl may be substituted or unsubstituted.


An “O-carboxy” group refers to a “RC(═O)O—” group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl, as defined herein. An O-carboxy may be substituted or unsubstituted.


The terms “ester” and “C-carboxy” refer to a “—C(═O)OR” group in which R can be the same as defined with respect to O-carboxy. An ester and C-carboxy may be substituted or unsubstituted.


A “thiocarbonyl” group refers to a “—C(═S)R” group in which R can be the same as defined with respect to O-carboxy. A thiocarbonyl may be substituted or unsubstituted.


A “trihalomethanesulfonyl” group refers to an “X3CSO2-” group wherein X is a halogen.


A “trihalomethanesulfonamido” group refers to an “X3CS(O)2N(RA)—” group wherein each X is a halogen and RA hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl.


The term “amino” as used herein refers to a —NH2 group.


As used herein, the term “hydroxy” refers to a —OH group.


A “cyano” group refers to a “—CN” group.


The term “azido” as used herein refers to a —N3 group.


An “isocyanato” group refers to a “—NCO” group.


A “thiocyanato” group refers to a “—CNS” group.


An “isothiocyanato” group refers to an “—NCS” group.


A “mercapto” group refers to an “—SH” group.


A “carbonyl” group refers to a C═O group.


An “S-sulfonamido” group refers to a “—SO2N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An S-sulfonamido may be substituted or unsubstituted.


An “N-sulfonamido” group refers to a “RSO2N(RA)—” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An N-sulfonamido may be substituted or unsubstituted.


An “O-carbamyl” group refers to a “—OC(═O)N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An O-carbamyl may be substituted or unsubstituted.


An “N-carbamyl” group refers to an “ROC(═O)N(RA)—” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An N-carbamyl may be substituted or unsubstituted.


An “O-thiocarbamyl” group refers to a “—OC(═S)—N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An O-thiocarbamyl may be substituted or unsubstituted.


An “N-thiocarbamyl” group refers to an “ROC(═S)N(RA)—” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. An N-thiocarbamyl may be substituted or unsubstituted.


A “C-amido” group refers to a “—C(═O)N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, (heteroaryl)alkyl or (heteroalicyclyl)alkyl. A C-amido may be substituted or unsubstituted.


An “N-amido” group refers to a “RC(═O)N(RA)—” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, heteroalicyclyl, aralkyl, or (heteroalicyclyl)alkyl. An N-amido may be substituted or unsubstituted.


The term “halogen atom” or “halogen” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, such as, fluorine, chlorine, bromine and iodine.


Where the numbers of substituents is not specified (e.g. haloalkyl), there may be one or more substituents present. For example “haloalkyl” may include one or more of the same or different halogens. As another example, “C1-C3 alkoxyphenyl” may include one or more of the same or different alkoxy groups containing one, two or three atoms.


As used herein, the abbreviations for any protective groups, amino acids and other compounds, are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (See, Biochem. 11:942-944 (1972)).


The term “nucleoside” is used herein in its ordinary sense as understood by those skilled in the art, and refers to a compound composed of an optionally substituted pentose moiety or modified pentose moiety attached to a heterocyclic base or tautomer thereof via a N-glycosidic bond, such as attached via the 9-position of a purine-base or the 1-position of a pyrimidine-base. Examples include, but are not limited to, a ribonucleoside comprising a ribose moiety and a deoxyribonucleoside comprising a deoxyribose moiety. A modified pentose moiety is a pentose moiety in which an oxygen atom has been replaced with a carbon and/or a carbon has been replaced with a sulfur or an oxygen atom. A “nucleoside” is a monomer that can have a substituted base and/or sugar moiety. Additionally, a nucleoside can be incorporated into larger DNA and/or RNA polymers and oligomers. In some instances, the nucleoside can be a nucleoside analog drug.


The term “nucleotide” is used herein in its ordinary sense as understood by those skilled in the art, and refers to a nucleoside having a phosphate ester bound to the pentose moiety, for example, at the 5′-position.


As used herein, the term “heterocyclic base” refers to an optionally substituted nitrogen-containing heterocyclyl that can be attached to an optionally substituted pentose moiety or modified pentose moiety. In some embodiments, the heterocyclic base can be selected from an optionally substituted purine-base, an optionally substituted pyrimidine-base and an optionally substituted triazole-base (for example, a 1,2,4-triazole). The term “purine-base” is used herein in its ordinary sense as understood by those skilled in the art, and includes its tautomers. Similarly, the term “pyrimidine-base” is used herein in its ordinary sense as understood by those skilled in the art, and includes its tautomers. A non-limiting list of optionally substituted purine-bases includes purine, adenine, guanine, hypoxanthine, xanthine, alloxanthine, 7-alkylguanine (e.g., 7-methylguanine), theobromine, caffeine, uric acid and isoguanine Examples of pyrimidine-bases include, but are not limited to, cytosine, thymine, uracil, 5,6-dihydrouracil and 5-alkylcytosine (e.g., 5-methylcytosine). An example of an optionally substituted triazole-base is 1,2,4-triazole-3-carboxamide. Other non-limiting examples of heterocyclic bases include diaminopurine, 8-oxo-N6-alkyladenine (e.g., 8-oxo-N6-methyladenine), 7-deazaxanthine, 7-deazaguanine, 7-deazaadenine, N4,N4-ethanocytosin, N6,N6-ethano-2,6-diaminopurine, 5-halouracil (e.g., 5-fluorouracil and 5-bromouracil), pseudoisocytosine, isocytosine, isoguanine, and other heterocyclic bases described in U.S. Pat. Nos. 5,432,272 and 7,125,855, which are incorporated herein by reference for the limited purpose of disclosing additional heterocyclic bases. In some embodiments, a heterocyclic base can be optionally substituted with an amine or an enol protecting group(s).


The term “—N-linked amino acid” refers to an amino acid that is attached to the indicated moiety via a main-chain amino or mono-substituted amino group. When the amino acid is attached in an —N-linked amino acid, one of the hydrogens that is part of the main-chain amino or mono-substituted amino group is not present and the amino acid is attached via the nitrogen. As used herein, the term “amino acid” refers to any amino acid (both standard and non-standard amino acids), including, but not limited to, α-amino acids, β-amino acids, γ-amino acids and δ-amino acids. Examples of suitable amino acids include, but are not limited to, alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan and valine. Additional examples of suitable amino acids include, but are not limited to, ornithine, hypusine, 2-aminoisobutyric acid, dehydroalanine, gamma-aminobutyric acid, citrulline, beta-alanine, alpha-ethyl-glycine, alpha-propyl-glycine and norleucine. N-linked amino acids can be substituted or unsubstituted.


The term “—N-linked amino acid ester derivative” refers to an amino acid in which a main-chain carboxylic acid group has been converted to an ester group. In some embodiments, the ester group has a formula selected from alkyl-O—C(═O)—, cycloalkyl-O—C(═O)—, aryl-O—C(═O)— and aryl(alkyl)-O—C(═O)—. A non-limiting list of ester groups include substituted and unsubstituted versions of the following: methyl-O—C(═O)—, ethyl-O—C(═O)—, n-propyl-O—C(═O)—, isopropyl-O—C(═O)—, n-butyl-O—C(═O)—, isobutyl-O—C(═O)—, tert-butyl-O—C(═O)—, neopentyl-O—C(═O)—, cyclopropyl-O—C(═O)—, cyclobutyl-O—C(═O)—, cyclopentyl-O—C(═O)—, cyclohexyl-O—C(═O)—, phenyl-O—C(═O)—, benzyl-O—C(═O)— and naphthyl-O—C(═O)—. N-linked amino acid ester derivatives can be substituted or unsubstituted.


The terms “phosphorothioate” and “phosphothioate” refer to a compound of the general formula




embedded image



its protonated forms (for example,




embedded image



and its tautomers (such as




embedded image


As used herein, the term “phosphate” is used in its ordinary sense as understood by those skilled in the art, and includes its protonated forms (for example,




embedded image



As used herein, the terms “monophosphate,” “diphosphate,” and “triphosphate” are used in their ordinary sense as understood by those skilled in the art, and include protonated forms.


The terms “protecting group” and “protecting groups” as used herein refer to any atom or group of atoms that is added to a molecule in order to prevent existing groups in the molecule from undergoing unwanted chemical reactions. Examples of protecting group moieties are described in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3. Ed. John Wiley & Sons, 1999, and in J. F. W. McOmie, Protective Groups in Organic Chemistry Plenum Press, 1973, both of which are hereby incorporated by reference for the limited purpose of disclosing suitable protecting groups. The protecting group moiety may be chosen in such a way, that they are stable to certain reaction conditions and readily removed at a convenient stage using methodology known from the art. A non-limiting list of protecting groups include benzyl; substituted benzyl; alkylcarbonyls and alkoxycarbonyls (e.g., t-butoxycarbonyl (BOC), acetyl, or isobutyryl); arylalkylcarbonyls and arylalkoxycarbonyls (e.g., benzyloxycarbonyl); substituted methyl ether (e.g., methoxymethyl ether); substituted ethyl ether; a substituted benzyl ether; tetrahydropyranyl ether; silyls (e.g., trimethylsilyl, triethylsilyl, triisopropylsilyl, t-butyldimethylsilyl, tri-iso-propylsilyloxymethyl, [2-(trimethylsilyl)ethoxy]methyl or t-butyldiphenylsilyl); esters (e.g., benzoate ester); carbonates (e.g., methoxymethylcarbonate); sulfonates (e.g. tosylate or mesylate); acyclic ketal (e.g., dimethyl acetal); cyclic ketals (e.g., 1,3-dioxane, 1,3-dioxolanes, and those described herein); acyclic acetal; cyclic acetal (e.g., those described herein); acyclic hemiacetal; cyclic hemiacetal; cyclic dithioketals (e.g., 1,3-dithiane or 1,3-dithiolane); orthoesters (e.g., those described herein) and triarylmethyl groups (e.g., trityl; monomethoxytrityl (MMTr); 4,4′-dimethoxytrityl (DMTr); 4,4′,4″-trimethoxytrityl (TMTr); and those described herein).


“Leaving group” as used herein refers to any atom or moiety that is capable of being displaced by another atom or moiety in a chemical reaction. More specifically, in some embodiments, “leaving group” refers to the atom or moiety that is displaced in a nucleophilic substitution reaction. In some embodiments, “leaving groups” are any atoms or moieties that are conjugate bases of strong acids. Examples of suitable leaving groups include, but are not limited to, tosylates and halogens. Non-limiting characteristics and examples of leaving groups can be found, for example in Organic Chemistry, 2d ed., Francis Carey (1992), pages 328-331; Introduction to Organic Chemistry, 2d ed., Andrew Streitwieser and Clayton Heathcock (1981), pages 169-171; and Organic Chemistry, 5th ed., John McMurry (2000), pages 398 and 408; all of which are incorporated herein by reference for the limited purpose of disclosing characteristics and examples of leaving groups.


The term “pharmaceutically acceptable salt” refers to a salt of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In some embodiments, the salt is an acid addition salt of the compound. Pharmaceutical salts can be obtained by reacting a compound with inorganic acids such as hydrohalic acid (e.g., hydrochloric acid or hydrobromic acid), sulfuric acid, nitric acid and phosphoric acid. Pharmaceutical salts can also be obtained by reacting a compound with an organic acid such as aliphatic or aromatic carboxylic or sulfonic acids, for example formic, acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-toluensulfonic, salicylic or naphthalenesulfonic acid. Pharmaceutical salts can also be obtained by reacting a compound with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C1-C7 alkylamine, cyclohexylamine, triethanolamine, ethylenediamine, and salts with amino acids such as arginine and lysine.


Terms and phrases used in this application, and variations thereof, especially in the appended claims, unless otherwise expressly stated, should be construed as open ended as opposed to limiting. As examples of the foregoing, the term ‘including’ should be read to mean ‘including, without limitation,’ ‘including but not limited to,’ or the like; the term ‘comprising’ as used herein is synonymous with ‘including,’ ‘containing,’ or ‘characterized by,’ and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps; the term ‘having’ should be interpreted as ‘having at least;’ the term ‘includes’ should be interpreted as ‘includes but is not limited to;’ the term ‘example’ is used to provide exemplary instances of the item in discussion, not an exhaustive or limiting list thereof; and use of terms like ‘preferably,’ ‘preferred,’ ‘desired,’ or ‘desirable,’ and words of similar meaning should not be understood as implying that certain features are critical, essential, or even important to the structure or function, but instead as merely intended to highlight alternative or additional features that may or may not be utilized in a particular embodiment. In addition, the term “comprising” is to be interpreted synonymously with the phrases “having at least” or “including at least”. When used in the context of a process, the term “comprising” means that the process includes at least the recited steps, but may include additional steps. When used in the context of a compound, composition or device, the term “comprising” means that the compound, composition or device includes at least the recited features or components, but may also include additional features or components. Likewise, a group of items linked with the conjunction ‘and’ should not be read as requiring that each and every one of those items be present in the grouping, but rather should be read as ‘and/or’ unless expressly stated otherwise. Similarly, a group of items linked with the conjunction ‘or’ should not be read as requiring mutual exclusivity among that group, but rather should be read as ‘and/or’ unless expressly stated otherwise.


With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity. The indefinite article “a” or “an” does not exclude a plurality. A single processor or other unit may fulfill the functions of several items recited in the claims. The mere fact that certain measures are recited in mutually different dependent claims does not indicate that a combination of these measures cannot be used to advantage. Any reference signs in the claims should not be construed as limiting the scope.


It is understood that, in any compound described herein having one or more chiral centers, if an absolute stereochemistry is not expressly indicated, then each center may independently be of R-configuration or S-configuration or a mixture thereof. Thus, the compounds provided herein may be enantiomerically pure, enantiomerically enriched, racemic mixture, diastereomerically pure, diastereomerically enriched, or a stereoisomeric mixture. In addition it is understood that, in any compound described herein having one or more double bond(s) generating geometrical isomers that can be defined as E or Z, each double bond may independently be E or Z a mixture thereof.


Likewise, it is understood that, in any compound described, all tautomeric forms are also intended to be included. For example all tautomers of a phosphate and a thiophosphate are intended to be included. Examples of tautomers of a phosphorothioate include the following:




embedded image



Furthermore, all tautomers of heterocyclic bases known in the art are intended to be included, including tautomers of natural and non-natural purine-bases and pyrimidine-bases.


It is to be understood that where compounds disclosed herein have unfilled valencies, then the valencies are to be filled with hydrogens or isotopes thereof, e.g., hydrogen-1 (protium) and hydrogen-2 (deuterium).


It is understood that the compounds described herein can be labeled isotopically. Substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. Each chemical element as represented in a compound structure may include any isotope of said element. For example, in a compound structure a hydrogen atom may be explicitly disclosed or understood to be present in the compound. At any position of the compound that a hydrogen atom may be present, the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen-1 (protium) and hydrogen-2 (deuterium). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise.


It is understood that the methods and combinations described herein include crystalline forms (also known as polymorphs, which include the different crystal packing arrangements of the same elemental composition of a compound), amorphous phases, salts, solvates, and hydrates. In some embodiments, the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, ethanol, or the like. In other embodiments, the compounds described herein exist in unsolvated form. Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, or the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. In addition, the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.


Where a range of values is provided, it is understood that the upper and lower limit, and each intervening value between the upper and lower limit of the range is encompassed within the embodiments.


Compound of Formula (A)


Some embodiments disclosed herein relate to a compound of Formula (A) or a pharmaceutically acceptable salt thereof:




embedded image



wherein: B1 can be an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with a protected amino group; R1 can be selected from O, OH, an optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester derivative; R2 can be selected from an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted heterocyclyl and




embedded image



wherein R19, R20 and R21 can be independently absent or hydrogen, and n can be 0 or 1; provided that when R1 is O or OH, then R2 is




embedded image



R3a and R3b can be independently selected from hydrogen, deuterium, an optionally substituted C1-6 alkyl, an optionally substituted C2-6 alkenyl, an optionally substituted C2-6 alkynyl, an optionally substituted C1-6 haloalkyl and aryl(C1-6 alkyl); or R3a and R3b can be taken together to form an optionally substituted C3-6 cycloalkyl; R4 can be selected from hydrogen, azido, an optionally substituted C1-6 alkyl, an optionally substituted C2-6 alkenyl and an optionally substituted C2-6 alkynyl; R5 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR10 and —OC(═O)R11; R6 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR12 and —OC(═O)R13; R7 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR14 and —OC(═O)R15; or R6 and R7 can be both oxygen atoms and linked together by a carbonyl group; R8 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR16 and —OC(═O)R17; R9 can be selected from hydrogen, azido, cyano, an optionally substituted C1-6 alkyl and —OR18; R10, R12, R14, R16 and R18 can be independently selected from hydrogen and an optionally substituted C1-6 alkyl; and R11, R13, R15 and R17 can be independently selected from an optionally substituted C1-6 alkyl and an optionally substituted C3-6 cycloalkyl; with the proviso that when R3a, R3b, R4, R5, R7, R8 and R9 are all hydrogen, then R6 cannot be azido.


With respect to R2, in some embodiments, R2 can be an optionally substituted heteroaryl. In other embodiments, R2 can be an optionally substituted heterocyclyl. In still other embodiments, R2 can be an optionally substituted aryl. For example, R2 can be an optionally substituted phenyl or an optionally substituted naphthyl. If R2 is a substituted phenyl or a substituted naphthyl, the phenyl ring and the naphthyl ring(s) can be substituted one or more times. Suitable substituents that can be present on optionally substituted phenyl and an optionally substituted naphthyl include electron-donating groups and electron-withdrawing groups. In some embodiments, R2 can be a para-substituted phenyl. In other embodiment, R2 can be an unsubstituted phenyl or an unsubstituted naphthyl. In yet still other embodiments, R2 can be




embedded image



wherein R19, R20 and R21 can be independently absent or hydrogen, and n can be 0 or 1. In some embodiments, n can be 0. In other embodiments, n can be 1. Those skilled in the art understand when n is 0, R2 can be an α-thiodiphosphate. Similarly, those skilled in the art understand when n is 1, R2 can be an α-thiotriphosphate. In some embodiments, at least one of R19, R20 and R21 can be absent. In other embodiments, at least one of R19, R20 and R21 can be hydrogen. In some embodiments, R20 and R21 can be absent. In other embodiments, R20 and R21 can be hydrogen. In some embodiments, R19, R20 and R21 can be absent. In some embodiments, R19, R20 and R21 can be hydrogen. Those skilled in the art understand that when any of R19, R20 and R21 are absent the oxygen atom to which R19, R20 and R21 are associated with can have a negative charge. For example, when R20 is absent, the oxygen atom to which R20 is associated with can be O. Depending upon the substituents attached to each phosphorus atoms, one or more the phosphorus atoms can be a chiral center. For example, when n is 1, the alpha-phosphorus (the phosphorus nearest to the pentose ring) can be a chiral center. In some embodiments, the alpha-phosphorus can be a (R)-stereocenter. In other embodiments, the alpha-phosphorus can be a (S)-stereocenter.


In some embodiments, R1 can be absent. In other embodiments, R1 can be hydrogen. In still other embodiments, R1 can be an optionally substituted N-linked α-amino acid. In yet still other embodiments, R1 can be an optionally substituted N-linked α-amino acid ester derivative. Various amino acids and amino acid ester derivatives can be used, including those described herein. Suitable amino acids include, but are not limited to, alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan and valine. Additional suitable amino acids include, but are not limited to, alpha-ethyl-glycine, alpha-propyl-glycine and beta-alanine Examples of an N-linked amino acid ester derivatives include, but are not limited to, an ester derivatives of any of the following amino acids: alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan and valine. Additional examples of N-linked amino acid ester derivatives include, but are not limited to, an ester derivative of any of the following amino acids: alpha-ethyl-glycine, alpha-propyl-glycine and beta-alanine.


In an embodiment, R1 can be an ester derivative of alanine. In an embodiment, R1 can be selected from alanine methyl ester, alanine ethyl ester, alanine isopropyl ester, alanine cyclohexyl ester, alanine neopentyl ester, valine isopropyl ester and leucine isopropyl ester. In some embodiments, the optionally substituted N-linked amino acid or the optionally substituted N-linked amino acid ester derivative can be in the L-configuration. In other embodiments, the optionally substituted N-linked amino acid or the optionally substituted N-linked amino acid ester derivative can be in the D-configuration.


In some embodiments, when R1 is an optionally substituted N-linked α-amino acid or an optionally substituted N-linked α-amino acid ester derivative, then R2 can be selected from optionally substituted aryl, an optionally substituted heteroaryl and an optionally substituted heterocyclyl. In some embodiments, when R1 is an optionally substituted N-linked α-amino acid ester derivative, then R2 can be an optionally substituted aryl. In other embodiments, when R1 is an optionally substituted N-linked α-amino acid ester derivative, then R2 can be an optionally substituted heteroaryl. In still other embodiments, when R1 is an optionally substituted N-linked α-amino acid ester derivative, then R2 can be an optionally substituted heterocyclyl.


In some embodiments, R1 can have the structure




embedded image



wherein R22 can be selected from hydrogen, an optionally substituted C1-6-alkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted aryl, an optionally substituted aryl(C1-6 alkyl) and an optionally substituted C1-6 haloalkyl; and R23 can be selected from hydrogen, an optionally substituted C1-6 alkyl, an optionally substituted C1-6 haloalkyl, an optionally substituted C3-6 cycloalkyl, an optionally substituted C6 aryl, an optionally substituted C10 aryl and an optionally substituted aryl(C1-6 alkyl); and R24 can be hydrogen or an optionally substituted C1-4-alkyl; or R23 and R24 can be taken together to form an optionally substituted C3-6 cycloalkyl.


When R1 has the structure shown above, R23 can be an optionally substituted C1-6-alkyl. Examples of suitable optionally substituted C1-6-alkyls include optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained). When R23 is substituted, R23 can be substituted with one or more substituents selected from N-amido, mercapto, alkylthio, an optionally substituted aryl, hydroxy, an optionally substituted heteroaryl, O-carboxy, and amino. In some embodiment, R23 can be an unsubstituted C1-6-alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained). In an embodiment, R23 can be methyl.


As to R22, in some embodiments, R22 can be an optionally substituted C1-6 alkyl. Examples of optionally substituted C1-6-alkyls include optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained). In some embodiments, R22 can be methyl or isopropyl. In some embodiments, R22 can be ethyl or neopentyl. In other embodiments, R22 can be an optionally substituted C3-6 cycloalkyl. Examples of optionally substituted C3-6 cycloalkyl include optionally substituted variants of the following: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. In an embodiment, R22 can be an optionally substituted cyclohexyl. In still other embodiments, R22 can be an optionally substituted aryl, such as phenyl and naphthyl. In yet still other embodiments, R22 can be an optionally substituted aryl(C1-6 alkyl). In some embodiments, R22 can be an optionally substituted benzyl. In some embodiments, R22 can be an optionally substituted C1-6 haloalkyl, for example, CF3.


In some embodiments, R24 can be hydrogen. In other embodiments, R24 can be an optionally substituted C1-4-alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl. In an embodiment, R24 can be methyl. In some embodiments, R23 and R24 can be taken together to form an optionally substituted C3-6 cycloalkyl. Examples of optionally substituted C3-6 cycloalkyl include optionally substituted variants of the following: cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Depending on the groups that are selected for R23 and R24, the carbon to which R23 and R24 are attached may be a chiral center. In some embodiment, the carbon to which R23 and R24 are attached may be a (R)-chiral center. In other embodiments, the carbon to which R23 and R24 are attached may be a (S)-chiral center.


Examples of a suitable




embedded image



groups include the following:




embedded image


The substituents attached to the 5′-position of a compound of Formula (A) can vary. In some embodiments, R3a and R3b can be the same. In other embodiments, R3a and R3b can be different. In some embodiments, R3a and R3b can be both hydrogen. In some embodiments, at least one of R3a and R3b can be an optionally substituted C1-6-alkyl; and the other of R3a and R3b can be hydrogen. Examples of suitable optionally substituted C1-6 alkyls include optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained). In an embodiment, at least one of R3a and R3b can be methyl, and the other of R3a and R3b can be hydrogen. In other embodiments, at least one of R3a and R3b can be an optionally substituted C1-6-haloalkyl, and the other of R3a and R3b can be hydrogen. One example of a suitable optionally substituted C1-6-haloalkyl is CF3. In other still embodiments, R3a and R3b can be taken together to form an optionally substituted C3-6 cycloalkyl. When the substituents attached to the 5′-carbon make the 5′-carbon chiral, in some embodiments, the 5′-carbon can be a (R)-stereocenter. In other embodiments, the 5′-carbon can be an (S)-stereocenter.


The substituents attached to the 4′-carbon can vary. In some embodiments, R4 can be hydrogen. In other embodiments, R4 can be azido. In still other embodiments, R4 can be an optionally substituted C1-6 alkyl, such as optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained). In some embodiments, R4 can be an optionally substituted C2-6 alkenyl. In some embodiments, R4 can be an optionally substituted C2-6 alkynyl.


The substituents attached to the 2′-carbon and the 3′-carbon can also vary. In some embodiments, R5 can be hydrogen. In other embodiments, R5 can be halogen. In still other embodiments, R5 can be azido. In yet still other embodiments, R5 can be cyano. In some embodiments, R5 can be an optionally substituted C1-6 alkyl, such as optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained). In other embodiments, R5 can be —OR10, wherein R10 can be hydrogen. In still other embodiments, R5 can be —OR10, wherein R10 can be an optionally substituted C1-6 alkyl. In yet still other embodiments, R5 can be —OC(═O)R11, wherein RH can be an optionally substituted C1-6 alkyl or an optionally substituted C3-6 cycloalkyl. Examples of suitable C1-6 alkyls and C3-6 cycloalkyls are described herein.


In some embodiments, R6 can be hydrogen. In other embodiments, R6 can be halogen. In still other embodiments, R6 can be azido. In yet still other embodiments, R6 can be cyano. In some embodiments, R6 can be an optionally substituted C1-6 alkyl. In other embodiments, R6 can be —OR12, wherein R12 can be hydrogen. In still other embodiments, R6 can be —OR12, wherein R12 can be an optionally substituted C1-6 alkyl. A non-limiting list of examples of R6 being —OR12, wherein R12 can be an optionally substituted C1-6 alkyl are methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy and tert-butoxy, pentoxy (straight-chained or branched) and hexoxy (straight-chained or branched). In yet still other embodiments, R6 can be —OC(═O)R13, wherein R13 can be an optionally substituted C1-6 alkyl or an optionally substituted C3-6 cycloalkyl. Examples of suitable optionally substituted C1-6 alkyls include optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl pentyl (branched and straight-chained), and hexyl (branched and straight-chained). Examples of suitable optionally substituted C3-6 cycloalkyls include optionally substituted variants of the following: cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.


In some embodiments, R7 can be hydrogen. In other embodiments, R7 can be halogen. In still other embodiments, R7 can be azido. In yet still other embodiments, R7 can be cyano. In some embodiments, R7 can be an optionally substituted C1-6 alkyl. In other embodiments, R7 can be —OR14. In an embodiment, when R14 is hydrogen, R7 can be a hydroxy group. In still other embodiments, when R14 is an optionally substituted C1-6 alkyl, R7 can be an optionally substituted C1-6 alkoxy. Examples, of R7 being —OR14, wherein R14 can be an optionally substituted C1-6 alkyl include, but are not limited to, are methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentoxy (straight-chained or branched) and hexoxy (straight-chained or branched). In yet still other embodiments, R7 can be —OC(═O)R15, wherein R15 can be an optionally substituted C1-6 alkyl, such as optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained). In some embodiments, R7 can be —OC(═O)R15, wherein R15 can be an optionally substituted C3-6 cycloalkyl


In some embodiments, R8 can be hydrogen. In other embodiments, R8 can be halogen. In still other embodiments, R8 can be azido. In yet still other embodiments, R8 can be cyano. In some embodiments, R8 can be −OR16. When R16 is hydrogen, R8 can be hydroxy. Alternatively, when R16 is an optionally substituted C1-6 alkyl, R8 can be an optionally substituted C1-6 alkoxy. Suitable alkoxy groups are described herein. In other embodiments, R8 can be an optionally substituted C1-6 alkyl. In still other embodiments, R8 can be —OC(═O)R17 in which R17 is an optionally substituted C1-6 alkyl. In yet still other embodiments, R8 can be —OC(═O)R17 in which R17 is an optionally substituted C3-6 cycloalkyl. Examples of suitable C1-6 alkyl and C3-6 cycloalkyl groups are described herein.


In some embodiments, R6 and R7 can both be hydroxy. In still other embodiments, R6 and R7 can both be both oxygen atoms and linked together by a carbonyl group, for example, —O—C(═O)—O—. In some embodiments, at least one of R7 and R8 can be a halogen. In some embodiments, R7 and R8 can both be a halogen. In other embodiments, R7 can be a halogen and R8 can be an optionally substituted C1-6 alkyl, such as those described herein. In other embodiments, R7 can be hydrogen and R8 can be a halogen. In still other embodiments, at least one of R6 and R7 can be a hydroxy and R8 can be an optionally substituted C1-6 alkyl. In yet still other embodiments, R6 can be hydroxy, R7 can be hydroxy, H or halogen, and R8 can be an optionally substituted C1-6 alkyl. In some embodiments, R3a, R3b, R4, R5 and R9 can be hydrogen in any of the embodiments described in this paragraph. In some embodiments, B1 can be an optionally substituted adenine, an optionally substituted guanine, and optionally substituted thymine, optionally substituted cytosine, or an optionally substituted uracil in any of the embodiments described in this paragraph.


In some embodiments, R9 can be hydrogen. In other embodiments, R9 can be azido. In still other embodiments, R9 can be cyano. In yet still other embodiments, R9 can be an optionally substituted C1-6 alkyl, such as those described herein. In some embodiments, R9 can be —OR18. In some embodiments, when R9 is —OR18, R9 can be a hydroxy group. In other embodiments, when R9 is —OR18, R9 can be an optionally substituted C1-6 alkoxy. Examples of optionally substituted C1-6 alkoxy include the following: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentoxy (branched and straight-chained), and hexoxy (branched and straight-chained).


Various optionally substituted heterocyclic bases can be attached to the pentose ring. In some embodiments, one or more of the amine and/or amino groups may be protected with a suitable protecting group. For example, an amino group may be protected by transforming the amine and/or amino group to an amide or a carbamate. In some embodiments, an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with one or more protected amino groups can have one of the following structures:




embedded image



wherein: RA2 can be selected from hydrogen, halogen and NHRJ2, wherein RJ2 can be selected from hydrogen, —C(═O)RK2 and —C(═O)ORL2; RB2 can be halogen or NHRW2, wherein RW2 is selected from hydrogen, an optionally substituted C1-6 alkyl, an optionally substituted C2-6 alkenyl, an optionally substituted C3-8 cycloalkyl, —C(═O)RM2 and —C(═O)ORN2; RC2 can be hydrogen or NHRO2, wherein RO2 can be selected from hydrogen, —C(═O)RP2 and —C(═O)ORQ2; RD2 can be selected from hydrogen, halogen, an optionally substituted C1-6 alkyl, an optionally substituted C2-6 alkenyl and an optionally substituted C2-6 alkynyl; RE2 can be selected from hydrogen, an optionally substituted C1-6 alkyl, an optionally substituted C3-8 cycloalkyl, —C(═O)RR2 and —C(═O)ORS2; RF2 can be selected from hydrogen, halogen, an optionally substituted C1-6 alkyl, an optionally substituted C2-6 alkenyl and an optionally substituted C2-6 alkynyl; Y2 can be N (nitrogen) or CRI2, wherein RI2 can be selected from hydrogen, halogen, an optionally substituted C1-6-alkyl, an optionally substituted C2-6-alkenyl and an optionally substituted C2-6-alkynyl; RG2 can be an optionally substituted C1-6 alkyl; RH2 can be hydrogen or NHRT2, wherein RT2 can be independently selected from hydrogen, —C(═O)RU2 and —C(═O)ORV2, and RK2, RL2; RM2, RN2, RP2, RQ2, RR2, RS2, RU2 and RV2 can be independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C3-6 cycloalkenyl, C3-6 cycloalkynyl, C6-10 aryl, heteroaryl, heteroalicyclyl, aryl(C1-6 alkyl), heteroaryl(C1-6 alkyl) and heteroalicyclyl(C1-6 alkyl). In some embodiments, the structures shown above can be modified by replacing one or more hydrogens with substituents selected from the list of substituents provided for the definition of “substituted.” Suitable optionally substituted C1-6 alkyl groups that can be present on an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with one or more protected amino groups are described herein, and include, optionally substituted variants of the following: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained), and hexyl (branched and straight-chained).


In some embodiments, B1 can be selected from adenine, guanine, thymine, cytosine and uracil. In some embodiments, RB2 can be NH2. In other embodiments, RE2 can be hydrogen. In some embodiments, B1 can be




embedded image



In other embodiments, B1 can be




embedded image



In some embodiments, B1 can be




embedded image



In some embodiments, B1 can be




embedded image



In still other embodiments, B1 can be




embedded image



In yet still other embodiments, B1 can be




embedded image



In some embodiments, B1 can be




embedded image



In some embodiments, when R2 is a substituted or unsubstituted phenyl, then R1 cannot be




embedded image



In other embodiments, when R2 is a substituted or unsubstituted phenyl, then R1 cannot be




embedded image



In still other embodiments, when R2 is a substituted or unsubstituted phenyl and R1 is




embedded image



then at least one of R5 and R6 cannot be hydroxy.


In some embodiments, when R1 is O or OH, then R2 cannot be




embedded image



In some embodiments, at least one of R3a and R3b cannot be hydrogen. In some embodiments, R4 is not azido. In some embodiments, when R4 is not azido, then R7 and R8 are not both halogen. In some embodiments, when R4 is azido, then B1 is not an optionally substituted uracil, optionally substituted uracil with one or more protected amino groups, an optionally substituted cytosine or optionally substituted cytosine with one or more protected amino groups. In some embodiments, R6 cannot be azido. In some embodiments, when R1 is a methyl ester of glycine, alanine, valine, or phenylalanine; R2 is p-chlorophenyl or p-nitrophenyl; B1 is thymine; and R3a, R3b, R4, R5, R7, R8, and R9 are all hydrogen; then R6 cannot be azido. In some embodiments, at least one of R6 and R7 cannot be hydroxy. For example, R6 cannot be hydroxy, R7 cannot be hydroxy, or both of R6 and R7 cannot be hydroxy.


Some embodiments disclosed herein relate to a compound of Formula (A) or a pharmaceutically acceptable salt thereof, wherein: B1 can be an optionally substituted heterocyclic base as described above; R1 can be selected from O, OH, an optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester derivative: R2 can be selected from an optionally substituted aryl and




embedded image



wherein R19, R20 and R21 can be independently absent or hydrogen, and n can be 0 or 1; provided that when R1 is O or OH, then R2 is




embedded image



R3a and R3b can be hydrogen; R4 can be hydrogen; R5 can be selected from hydrogen, halogen, an optionally substituted C1-6 alkyl and —OR10; R6 can be selected from hydrogen, halogen, optionally substituted C1-6 alkyl, —OR12 and —OC(═O)R13; R7 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR14 and —OC(═O)R15; or R6 and R7 can be both oxygen atoms and linked together by a carbonyl group; R8 can be selected from hydrogen, halogen, an optionally substituted C1-6 alkyl and —OR16; R9 can be hydrogen; R10, R12, R14 and R16 can be independently selected from hydrogen and an optionally substituted C1-6 alkyl; and R13 and R15 can be independently selected from an optionally substituted C1-6 alkyl and an optionally substituted C3-6 cycloalkyl.


Some embodiments disclosed herein relate to a compound of Formula (A) or a pharmaceutically acceptable salt thereof, wherein: B1 can be an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with a protected amino group selected from




embedded image



R1 can be selected from O, OH, an optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester derivative; R2 can be selected from an optionally substituted aryl and




embedded image



wherein R19, R20 and R21 can be independently absent or hydrogen, and n can be 0 or 1; provided that when R1 is O or OH, then R2 is




embedded image



R3a and R3b can be hydrogen; R4 can be hydrogen; R5 can be selected from hydrogen, halogen, an optionally substituted C1-6 alkyl and —OR10; R6 can be selected from hydrogen, halogen, optionally substituted C1-6 alkyl, —OR12 and —OC(═O)R13; R7 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR14 and —OC(═O)R15; or R6 and R7 can be both oxygen atoms and linked together by a carbonyl group; R8 can be selected from hydrogen, halogen, an optionally substituted C1-6 alkyl and —OR16; R9 can be hydrogen; R10, R12, R14 and R16 can be independently selected from hydrogen and an optionally substituted C1-6 alkyl; and R13 and R15 can be independently selected from an optionally substituted C1-6 alkyl and an optionally substituted C3-6 cycloalkyl.


In some embodiments, Formula (A) can be a compound of Formula (Iα), wherein: B1 can be an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with a protected amino group selected from cytosine, uridine, thymidine, guanine and adenine; R1 can be selected from O, OH, and an optionally substituted N-linked amino acid ester derivative of alanine, valine, or leucine; R2 can be selected from an optionally substituted phenyl, an optionally substituted naphthyl, an optionally substituted pyridyl, an optionally substituted quinolyl, and




embedded image



wherein R19, R20 and R21 independently can be hydrogen or absent, and n can be 0 or 1; provided that when R1 is O or OH, then R2 is




embedded image



R3a and R3b can be hydrogen; R4 can be hydrogen; R5 can be hydrogen; R6 can be —OR12 or —OC(═O)R13; R7 can be selected from halogen, —OR14 and —OC(═O)R15; R8 can be an optionally substituted C1-6 alkyl; R9 can be hydrogen; R12 and R14 can be independently hydrogen or an optionally substituted C1-6 alkyl; and R13 and R15 can be independently an optionally substituted C1-6 alkyl.


Some embodiments relate to a compound of Formula (A) or a pharmaceutically acceptable salt thereof, wherein: B1 can be an optionally substituted heterocyclic base or an optionally substituted heterocyclic base with a protected amino group; R1 can be selected from O, OH, an optionally substituted N-linked amino acid and an optionally substituted N-linked amino acid ester derivative; R2 can be selected from an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted heterocyclyl and




embedded image



wherein R19, R20 and R21 can be independently absent or hydrogen, and n can be 0 or 1; provided that when R1 is O or OH, then R2 is




embedded image



R3a and R3b can be independently selected from hydrogen, an optionally substituted C1-6 alkyl, an optionally substituted C2-6 alkenyl, an optionally substituted C2-6 alkynyl, an optionally substituted C1-6 haloalkyl and aryl(C1-6 alkyl); or R3a and R3b can be taken together to form an optionally substituted C3-6 cycloalkyl; R4 can be selected from hydrogen, azido, an optionally substituted C1-6 alkyl, an optionally substituted C2-6 alkenyl and an optionally substituted C2-6 alkynyl; R5 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR10 and —OC(═O)R11; R6 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR12 and —OC(═O)R13; R7 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR14 and —OC(═O)R15; or R6 and R7 can be both oxygen atoms and linked together by a carbonyl group; R8 can be selected from hydrogen, halogen, azido, cyano, an optionally substituted C1-6 alkyl, —OR16 and —OC(═O)R17; R9 can be selected from hydrogen, azido, cyano, an optionally substituted C1-6 alkyl and —OR18; R10, R12, R14, R16 and R18 can be independently selected from hydrogen and an optionally substituted C1-6 alkyl; and R11, R13, R15 and R17 can be independently an optionally substituted C1-6 alkyl and an optionally substituted C3-6 cycloalkyl.


In some embodiments, a compound of Formula (A) can be a single diastereomer. In other embodiments, a compound of Formula (A) can be a mixture of diastereomers. In some embodiments, a compound of Formula (A) can be a 1:1 mixture of two diastereomers. In some embodiments, a compound of Formula (A) can be diasteriometrically enriched (for example, one diastereomer can be present at a concentration of >55%, ≧75%, ≧80%, ≧90%, ≧95%, ≧98%, or ≧99% as compared to the total concentration of the other diastereomers).


Examples of compounds of Formula (A) are provided in FIG. 1.


Compounds of Formula (C)


Compounds of Formula (C) are therapeutic compounds that include HCV protease inhibitors, nucleoside HCV polymerase inhibitors, non-nucleoside HCV polymerase inhibitors, NS5A inhibitors, and other antivirals. Examples of compounds of Formula (C) are provided in FIG. 2.


Pharmaceutical Activity


In some embodiments, a composition comprising a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can act as a chain terminator of HCV replication. For example, incorporation of compound of Formula (A) containing a moiety at the 2′-carbon position can terminate further elongation of the RNA chain of HCV. For example, a compound of Formula (A) can contain a 2′-carbon modification when R8 of Formula (A) is a non-hydrogen group selected from halogen or an optionally substituted C1-6 alkyl.


In some embodiments, a composition containing a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can have increased metabolic and/or plasma stability. In some embodiments, a composition containing a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. A non-limiting list of example properties include, but are not limited to, increased biological half life, increased bioavailability, increase potency, a sustained in vivo response, increased dosing intervals, decreased dosing amounts, decreased cytotoxicity, reduction in required amounts for treating disease conditions, reduction in viral load, reduction in time to seroreversion (i.e., the virus becomes undetectable in patient serum), increased sustained viral response, a reduction of morbidity or mortality in clinical outcomes, increased subject compliance, decreased liver conditions (such as liver fibrosis, liver cirrhosis and/or liver cancer), and compatibility with other medications. In some embodiments, a composition containing a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can have a biological half life of greater than 24 hours, e.g., a biological half life in the range of about 40 to 46 hours for some compounds of Formula (A). In some embodiments, a composition containing a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can have more potent antiviral activity (for example, a lower IC50 in an HCV replicon assay) as compared to the current standard of care.


Pharmaceutical Compositions


In some embodiments, a pharmaceutical composition can include a single diastereomer of a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, (for example, a single diastereomer is present in the pharmaceutical composition at a concentration of greater than 99% compared to the total concentration of the other diastereomers). In other embodiments, a pharmaceutical composition can include a mixture of diastereomers of a compound of Formula (A), or a pharmaceutically acceptable salt thereof. For example, the pharmaceutical composition can include a concentration of one diastereomer of >50%, ≧60%, ≧70%, ≧80%, ≧90%, ≧95%, or ≧98%, as compared to the total concentration of the other diastereomers. In some embodiments, a pharmaceutical composition includes a 1:1 mixture of two diastereomers of a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof.


The term “pharmaceutical composition” refers to a mixture of one or more compounds disclosed herein with one or more chemical components, such as diluents or carriers. The pharmaceutical composition facilitates administration of the compound to an organism. Pharmaceutical compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid and salicylic acid. Pharmaceutical compositions will generally be tailored to the specific intended route of administration.


The term “physiologically acceptable” defines a carrier, diluent or excipient that does not abrogate the biological activity and properties of the compound.


As used herein, a “carrier” refers to a compound that facilitates the incorporation of a compound into cells or tissues. For example, without limitation, dimethyl sulfoxide (DMSO) is a commonly utilized carrier that facilitates the uptake of many organic compounds into cells or tissues of a subject.


As used herein, a “diluent” refers to an ingredient in a pharmaceutical composition that lacks pharmacological activity but may be pharmaceutically necessary or desirable. For example, a diluent may be used to increase the bulk of a potent drug whose mass is too small for manufacture and/or administration. It may also be a liquid for the dissolution of a drug to be administered by injection, ingestion or inhalation. A common form of diluent in the art is a buffered aqueous solution such as, without limitation, phosphate buffered saline that mimics the composition of human blood.


As used herein, an “excipient” refers to an inert substance that is added to a pharmaceutical composition to provide, without limitation, bulk, consistency, stability, binding ability, lubrication, disintegrating ability etc., to the composition. A “diluent” is a type of excipient.


The pharmaceutical compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or carriers, diluents, excipients or combinations thereof. Proper formulation is dependent upon the route of administration chosen. Techniques for formulation and administration of the compounds described herein are known to those skilled in the art.


Combination therapies contemplated include use of a compound of Formula (A) selected from those listed in FIG. 1 and a compound (e.g., an HCV protease inhibitor, nucleoside HCV polymerase inhibitor, non-nucleoside HCV polymerase inhibitor, NS5A inhibitor, or other antiviral) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. Other combination therapies contemplated include use of a compound of Formula (A) selected from those listed in FIG. 1 and a compound of Formula (C) selected from 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. Other combination therapies contemplated include use of a compound selected from Compound 1000 through Compound 1032 and a compound selected from 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds.


Combination therapies also contemplated include use of a compound of Formula (A) selected from those listed in FIG. 1 and two different compounds of Formula (C) (e.g., HCV protease inhibitors, nucleoside HCV polymerase inhibitors, non-nucleoside HCV polymerase inhibitors, NS5A inhibitors, or other antivirals) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. Other combination therapies contemplated include use of a compound selected from Compound 1000 through Compound 1032 and two different compounds selected from Compound 3000 through Compound 3067. Still other combination therapies contemplated include use of a compound of Formula (A) selected from those listed in FIG. 1, a first compound of Formula (C) (e.g., HCV protease inhibitors, nucleoside HCV polymerase inhibitors, non-nucleoside HCV polymerase inhibitors, NS5A inhibitors, or other antivirals) selected from those listed in FIG. 2 and a second compound of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are different. Yet still other combination therapies contemplated include use of a compound selected from Compound 1000 through Compound 1032; a first compound selected from Compound 3000 through Compound 3067; and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 is different from the second compound selected from Compound 3000 through Compound 3067.


The pharmaceutically active ingredients of the combination therapy can be contained in a single unit dosage form, in two unit dosage forms, or in three unit dosage forms.


In some embodiments, a single unit dosage form can be provided containing a compound of Formula (A) selected from those listed in FIG. 1 and a compound of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, a single unit dosage form can be provided containing a compound of Formula (A) selected from those listed in FIG. 1 and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds.


In some embodiments, two unit dosage forms can be provided, with one containing a compound of Formula (A) selected from those listed in FIG. 1 and the other containing a compound of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, two unit dosage forms can be provided, with one containing a compound of Formula (A) selected from those listed in FIG. 1, and the other containing two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, two unit dosage forms can be provided, with one containing a compound of Formula (A) selected from those listed in FIG. 1 and a first compound of Formula (C) selected from those listed in FIG. 2, and the other containing a second compound of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first and second compound are not the same.


In some embodiments, three unit dosage forms can be provided, with one unit dosage form containing a compound of Formula (A) selected from those listed in FIG. 1 and two additional unit dosage forms, each containing a different compound of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds.


The pharmaceutical compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes. Additionally, the active ingredients are contained in an amount effective to achieve its intended purpose. Many of the compounds used in the pharmaceutical combinations disclosed herein may be provided as salts with pharmaceutically compatible counterions.


Multiple techniques of administering a compound or a composition exist in the art including, but not limited to, oral, rectal, topical, aerosol, injection and parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, intrathecal, direct intraventricular, intraperitoneal, intranasal and intraocular injections.


One may also administer the compound or composition in a local rather than systemic manner, for example, via injection of the compound or composition directly into the infected area, often in a depot or sustained release formulation. Furthermore, one may administer the compound or composition in a targeted drug delivery system, for example, in a liposome coated with a tissue-specific antibody. The liposomes can be targeted to and taken up selectively by the organ.


The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. Compositions that can include one or more compounds described herein formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.


Methods of Use


Some embodiments disclosed herein relate to a method of treating and/or ameliorating a disease or condition that can include administering to a subject an effective amount of a combination of compounds described herein. In some embodiments, such methods include administering an effective amount of a combination of a compound of Formula (A), and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, such methods include administering an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, such methods include administering an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, such methods include administering an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032 and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, such methods include administering an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C) and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) is different from the second of Formula (C). In some embodiments, such methods include administering an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067 and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 is different from the second compound selected from Compound 3000 through Compound 3067.


Other embodiments disclosed herein relates to a method of ameliorating or treating a viral infection that can include administering an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, the viral infection can be caused by a virus selected from an adenovirus, an Alphaviridae, an Arbovirus, an Astrovirus, a Bunyaviridae, a Coronaviridae, a Filoviridae, a Flaviviridae, a Hepadnaviridae, a Herpesviridae, an Alphaherpesvirinae, a Betaherpesvirinae, a Gammaherpesvirinae, a Norwalk Virus, an Astroviridae, a Caliciviridae, an Orthomyxoviridae, a Paramyxoviridae, a Paramyxoviruses, a Rubulavirus, a Morbillivirus, a Papovaviridae, a Parvoviridae, a Picornaviridae, an Aphthoviridae, a Cardioviridae, an Enteroviridae, a Coxsackie virus, a Polio Virus, a Rhinoviridae, a Phycodnaviridae, a Poxyiridae, a Reoviridae, a Rotavirus, a Retroviridae, an A-Type Retrovirus, an Immunodeficiency Virus, a Leukemia Viruses, an Avian Sarcoma Viruses, a Rhabdoviruses, a Rubiviridae, a Togaviridae an Arenaviridae and/or a Bornaviridae. In some embodiments, the viral infection can be a hepatitis C viral (HCV) infection. HCV is an enveloped positive strand RNA virus in the Flaviviridae family. There are various nonstructural proteins of HCV, such as NS2, NS3, NS4, NS4A, NS4B, NS5A, and NS5B. NS5B is believed to be an RNA-dependent RNA polymerase involved in the replication of HCV RNA.


Some embodiments disclosed herein relate to methods of ameliorating and/or treating a viral infection (for example, an HCV infection) that can include contacting a cell infected with the virus with an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for ameliorating and/or treating a viral infection (for example, an HCV infection) that can include contacting a cell infected with the virus with an effective amount of said combination of compounds described herein. Still other embodiments described herein relate to a combination of compounds described herein that can be used for ameliorating and/or treating a viral infection (for example, an HCV infection) by contacting a cell infected with the virus with an effective amount of said combination of compounds described herein. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same.


Some embodiments disclosed herein relate to methods of inhibiting replication of a virus (such as a hepatitis C virus) that can include contacting a cell infected with the virus with an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for inhibiting replication of a virus (such as a hepatitis C virus) that can include contacting a cell infected with the virus with an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for inhibiting replication of a virus (such as a hepatitis C virus) by contacting a cell infected with the virus with an effective amount of said combination of compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same.


Some embodiments described herein relate to a method of inhibiting an RNA dependent RNA polymerase can include contacting a cell (for example, a cell infected with HCV) with an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for inhibiting an RNA dependent RNA polymerase that can include contacting a cell (for example, a cell infected with HCV) with an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for inhibiting an RNA dependent RNA polymerase that can include contacting a cell (for example, a cell infected with HCV) with an effective amount of said combination of compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same.


Some embodiments described herein relate to a method of inhibiting NS5B polymerase activity that can include contacting a cell (for example, a cell infected with HCV) with an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for inhibiting NS5B polymerase activity that can include contacting a cell (for example, a cell infected with HCV) with an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for inhibiting NS5B polymerase activity that can include contacting a cell (for example, a cell infected with HCV) with an effective amount of said combination of compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same.


Some embodiments described herein relate to a method of inhibiting an HCV polymerase (for example, NS5B polymerase) can include contacting a cell (for example, a cell infected with HCV) with an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for inhibiting an HCV polymerase (for example, NS5B polymerase) that can include contacting a cell (for example, a cell infected with HCV) with an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for inhibiting an HCV polymerase (for example, NS5B polymerase) that can include contacting a cell (for example, a cell infected with HCV) with an effective amount of said combination of compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same.


Some embodiments described herein relate to a method of ameliorating and/or treating HCV infection in a subject suffering from an HCV infection that can include administering to the subject an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for ameliorating and/or treating HCV infection in a subject suffering from an HCV infection that can include administering to the subject an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for ameliorating and/or treating HCV infection in a subject suffering from an HCV infection that can include administering to the subject an effective amount of said combination of compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same.


Some embodiments described herein relate to a method of ameliorating and/or treating a condition selected from liver fibrosis, liver cirrhosis, and liver cancer in a subject suffering from one or more of the aforementioned liver conditions that can include administering an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for ameliorating and/or treating a condition selected from liver fibrosis, liver cirrhosis, and liver cancer in a subject suffering from one or more of the aforementioned liver conditions that can include administering an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for ameliorating and/or treating a condition selected from liver fibrosis, liver cirrhosis, and liver cancer in a subject suffering from one or more of the aforementioned liver conditions that can include administering an effective amount of said combination of compounds. In some embodiments, the one or more conditions selected from liver fibrosis, liver cirrhosis and liver cancer can be the result of an HCV infection. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same.


A cause of liver fibrosis, liver cirrhosis, and/or liver cancer can be an HCV infection. Some embodiments described herein relate to a method of increasing liver function in a subject having an HCV infection that can include administering to the subject an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for increasing liver function in a subject having an HCV infection that can include administering to the subject an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for increasing liver function in a subject having an HCV infection that can include administering to the subject an effective amount of said combination of compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same.


Also contemplated is a method for reducing or eliminating further virus-caused liver damage in a subject having an HCV infection by administering an effective amount of a combination of compounds described herein. Other embodiments described herein relate to using a combination of compounds described herein in the manufacture of a medicament for reducing or eliminating further virus-caused liver damage in a subject having an HCV infection by administering an effective amount of said combination of compounds. Still other embodiments described herein relate to a combination of compounds described herein that can be used for reducing or eliminating further virus-caused liver damage in a subject having an HCV infection by administering an effective amount of said combination of compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and one or more compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and a compound selected from Compound 3015, 3016, 3025, 3026, 3027, 3028, 3037, 3038, 3039, 3040, 3041, 3042, 3048, 3049, 3050, 3051, 3052, 3053, 3054, 3057, 3058, 3059, 3060, 3061, 3062, 3063, 3064, 3065 and 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In some embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A) selected from those listed in FIG. 1, and two different compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, and two different compounds selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds. In still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound of Formula (A), a first compound of Formula (C), and a second compound of Formula (C), or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound of Formula (C) and the second compound of Formula (C) are not the same. In yet still other embodiments, including those of this paragraph, the combination can include an effective amount of a combination of a compound selected from Compound 1000 through Compound 1032, a first compound selected from Compound 3000 through Compound 3067, and a second compound selected from Compound 3000 through Compound 3067, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds, wherein the first compound selected from Compound 3000 through Compound 3067 and the second compound selected from Compound 3000 through Compound 3067 are not the same. In some embodiments, this method or use can include slowing or halting the progression of liver disease. In other embodiments, the course of the liver disease can be reversed, and stasis or improvement in liver function can be achieved.


There are a variety of genotypes of HCV, and a variety of subtypes within each genotype. For example, at present it is known that there are eleven (numbered 1 through 11) main genotypes of HCV, although others have classified the genotypes as 6 main genotypes. Each of these genotypes is further subdivided into subtypes (1a-1c; 2a-2c; 3a-3b; 4a-4-e; 5a; 6a; 7a-7b; 8a-8b; 9a; 10a; and 11a). In some embodiments, a combination therapy as described herein can be effective to treat at least one genotype of HCV. In some embodiments, a combination therapy described herein (e.g., a combination therapy including a compound of Formula (A) selected from those listed in FIG. 1 and one or more compounds of Formula (C) selected from those listed in FIG. 2, or a pharmaceutically acceptable salt, hydrate, or solvate of the aforementioned compounds) can be effective to treat all 11 genotypes of HCV. In some embodiments, a combination therapy described herein (e.g., a combination therapy including a compound of Formula (A) selected from those listed in FIG. 1 and one or more compounds of Formula (C) listed in FIG. 2 can be effective to treat 3 or more, 5 or more, 7 or more of 9 more genotypes of HCV. In some embodiments, a combination therapy described herein (e.g., a combination therapy including a compound of Formula (A) selected from those listed in FIG. 1 and one or more compounds of Formula (C) listed in FIG. 2 is more effective against a larger number of HCV genotypes than the standard of care. In some embodiments, a combination therapy described herein (e.g., a combination therapy including a compound of Formula (A) from those listed in FIG. 1 and one or more compounds of Formula (C) listed in FIG. 2 is more effective against a particular HCV genotype than the standard of care (such as genotype 1, 2, 3, 4, 5 and/or 6).


Various indicators for determining the effectiveness of a method for treating an HCV infection are known to those skilled in the art. Example of suitable indicators include, but are not limited to, a reduction in viral load, a reduction in viral replication, a reduction in time to seroconversion (virus undetectable in patient serum), an increase in the rate of sustained viral response to therapy, a reduction of morbidity or mortality in clinical outcomes, a reduction in the rate of liver function decrease, stasis in liver function, improvement in liver function, reduction in one or more markers of liver dysfunction, including alanine transaminase, aspartate transaminase, total bilirubin, conjugated bilirubin, gamma glutamyl transpeptidase, and/or other indicator of disease response. Similarly, successful therapy with a combination therapy described herein (e.g., a combination therapy including a compound of Formula (A) selected from those listed in FIG. 1 and one or more compounds of Formula (C) listed in FIG. 2, or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds) can reduce the incidence of liver cancer in HCV patients.


In some embodiments, an amount of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, is an amount that is effective to reduce viral titers to undetectable levels, to about 100 to about 500, to about 50 to about 100, to about 10 to about 50, or to about 15 to about 25 international units/mL serum. In some embodiments, an amount of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, is an amount that is effective to reduce viral load compared to the viral load before administration of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds. For example, wherein the viral load is measured before administration of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, and again after completion of the treatment regime with a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds (for example, 1 month after completion). In some embodiments, an amount of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be an amount that is effective to reduce viral load to lower than about 100 genome copies/mL serum. In some embodiments, an amount of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, is an amount that is effective to achieve a reduction in viral titer in the serum of the subject in the range of about 1.5-log to about a 2.5-log reduction, about a 3-log to about a 4-log reduction, or a greater than about S-log reduction compared to the viral load before administration of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds. For example, the viral load can be measured before administration of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, and again after completion of the treatment regime with a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds (for example, 1 month after completion).


In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can result in at least a 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, 75, 100-fold or more reduction in the replication of HCV relative to pre-treatment levels in a subject, as determined after completion of the treatment regime (for example 1 month after completion). In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can result in a reduction of the replication of HCV relative to pre-treatment levels in the range of about 2 to about 5 fold, about 10 to about 20 fold, about 15 to about 40 fold, or about 50 to about 100 fold. In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can result in a reduction of HCV replication in the range of 1 to 1.5 log, 1.5 log to 2 log, 2 log to 2.5 log, 2.5 to 3 log, 3 log to 3.5 log or 3.5 to 4 log more reduction of HCV replication compared to the reduction of HCV replication achieved by pegylated interferon in combination with ribavirin, administered according to the standard of care, or may achieve the same reduction as that standard of care therapy in a shorter period of time, for example, in one month, two months, or three months, as compared to the reduction achieved after six months of standard of care therapy with ribavirin and pegylated interferon.


In some embodiments, an amount of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, is an amount that is effective to achieve a sustained viral response, for example, non-detectable or substantially non-detectable HCV RNA (e.g., less than about 500, less than about 400, less than about 200, or less than about 100 genome copies per milliliter serum) is found in the subject's serum for a period of at least about one month, at least about two months, at least about three months, at least about four months, at least about five months, or at least about six months following cessation of therapy.


In some embodiments, an amount of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can reduce a level of a marker of liver fibrosis by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%, or more, compared to the level of the marker in an untreated subject, or to a placebo-treated subject. Methods of measuring serum markers are known to those skilled in the art and include immunological-based methods, e.g., enzyme-linked immunosorbent assays (ELISA), radioimmunoassays, and the like, using antibody specific for a given serum marker. A non-limiting list of examples of markers includes measuring the levels of serum alanine aminotransferase (ALT), asparatate aminotransferacse (AST), alkaline phosphatase (ALP), gamma-glutamyl transpeptidase (GGT) and total bilirubin (TBIL) using known methods. In general, an ALT level of less than about 45 IU/L (international units/liter), an AST in the range of 10-34 IU/L, ALP in the range of 44-147 IU/L, GGT in the range of 0-51 IU/L, TBIL in the range of 0.3-1.9 mg/dL is considered normal. In some embodiments, an effective amount of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds is an amount effective to reduce ALT, AST, ALP, GGT and/or TBIL levels to within what is considered a normal level.


Subjects who are clinically diagnosed with HCV infection include “naïve” subjects (e.g., subjects not previously treated for HCV, particularly those who have not previously received IFN-alpha-based and/or ribavirin-based therapy) and individuals who have failed prior treatment for HCV (“treatment failure” subjects). Treatment failure subjects include “non-responders” (i.e., subjects in whom the HCV titer was not significantly or sufficiently reduced by a previous treatment for HCV (≦0.5 log IU/mL), for example, a previous IFN-alpha monotherapy, a previous IFN-alpha and ribavirin combination therapy, or a previous pegylated IFN-alpha and ribavirin combination therapy); and “relapsers” (i.e., subjects who were previously treated for HCV, for example, who received a previous IFN-alpha monotherapy, a previous IFN-alpha and ribavirin combination therapy, or a previous pegylated IFN-alpha and ribavirin combination therapy, whose HCV titer decreased, and subsequently increased).


In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be administered to a treatment failure subject suffering from HCV. In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be administered to a non-responder subject suffering from HCV. In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be administered to a relapsed subject suffering from HCV.


After a period of time, infectious agents can develop resistance to one or more therapeutic compounds. The term “resistance” as used herein refers to a viral strain displaying a delayed, lessened and/or null response to a therapeutic compound(s). For example, after treatment with an antiviral agent, the viral load of a subject infected with a resistant virus may be reduced to a lesser degree compared to the amount in viral load reduction exhibited by a subject infected with a non-resistant strain. In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be administered to a subject infected with an HCV strain that is resistant to one or more different anti-HCV agents. In some embodiments, development of resistant HCV strains is delayed when patients are treated with a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, or a pharmaceutically acceptable salt thereof, compared to the development of HCV strains resistant to other HCV drugs.


In some embodiments, an effective amount of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be administered to a subject for whom other anti-HCV medications are contraindicated. For example, administration of pegylated interferon alpha in combination with ribavirin is contraindicated in subjects with hemoglobinopathies (e.g., thalassemia major, sickle-cell anemia) and other subjects at risk from the hematologic side effects of current therapy. In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be provided to a subject that is hypersensitive to interferon or ribavirin.


Some subjects being treated for HCV experience a viral load rebound. The term “viral load rebound” as used herein refers to a sustained ≧0.5 log IU/mL increase of viral load above nadir before the end of treatment, where nadir is a ≧0.5 log IU/mL decrease from baseline. In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be administered to a subject experiencing viral load rebound, or can prevent such viral load rebound when used to treat the subject.


The standard of care for treating HCV has been associated with several side effects (adverse events). In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can decrease the number and/or severity of side effects that can be observed in HCV patients being treated with ribavirin and pegylated interferon according to the standard of care. Examples of side effects include, but are not limited to fever, malaise, tachycardia, chills, headache, arthralgias, myalgias, fatigue, apathy, loss of appetite, nausea, vomiting, cognitive changes, asthenia, drowsiness, lack of initiative, irritability, confusion, depression, severe depression, suicidal ideation, anemia, low white blood cell counts, and thinning of hair. In some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be provided to a subject that discontinued an HCV therapy because of one or more adverse effects or side effects associated with one or more other HCV agents.


Table A provides some embodiments of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, compared to the standard of care. Examples include the following: in some embodiments, a combination as described herein of a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, results in a percentage of non-responders that is 10% less than the percentage of non-responders receiving the standard of care; in some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, results in a number of side effects that is in the range of about 10% to about 30% less than compared to the number of side effects experienced by a subject receiving the standard of care; and in some embodiments, a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, results in a severity of a side effect (such as one of those described herein) that is 25% less than compared to the severity of the same side effect experienced by a subject receiving the standard of care. Methods of quantifying the severity of a side effect are known to those skilled in the art.














TABLE A





Percentage
Percentage
Percentage
Percentage
Number
Severity


of non-
of
of
of viral load
of side
of side


responders
relapsers
resistance
rebound
effects
effects







10% less
10% less
10% less
10% less
10% less
10% less


25% less
25% less
25% less
25% less
25% less
25% less


40% less
40% less
40% less
40% less
40% less
40% less


50% less
50% less
50% less
50% less
50% less
50% less


60% less
60% less
60% less
60% less
60% less
60% less


70% less
70% less
70% less
70% less
70% less
70% less


80% less
80% less
80% less
80% less
80% less
80% less


90% less
90% less
90% less
90% less
90% less
90% less


about 10%
about 10%
about 10%
about 10%
about 10%
about 10%


to about
to about
to about
to about
to about
to about


30% less
30% less
30% less
30% less
30% less
30% less


about 20%
about 20%
about 20%
about 20%
about 20%
about 20%


to about
to about
to about
to about
to about
to about


50% less
50% less
50% less
50% less
50% less
50% less


about 30%
about 30%
about 30%
about 30%
about 30%
about 30%


to about
to about
to about
to about
to about
to about


70% less
70% less
70% less
70% less
70% less
70% less


about 20%
about 20%
about 20%
about 20%
about 20%
about 20%


to about
to about
to about
to about
to about
to about


80% less
80% less
80% less
80% less
80% less
80% less









As used herein, a “subject” refers to an animal that is the object of treatment, observation or experiment. “Animal” includes cold- and warm-blooded vertebrates and invertebrates such as fish, shellfish, reptiles and, in particular, mammals. “Mammal” includes, without limitation, mice, rats, rabbits, guinea pigs, dogs, cats, sheep, goats, cows, horses, primates, such as monkeys, chimpanzees, and apes, and, in particular, humans. In some embodiments, the subject is human.


As used herein, the terms “treating,” “treatment,” “therapeutic,” or “therapy” do not necessarily mean total cure or abolition of the disease or condition. Any alleviation of any undesired signs or symptoms of a disease or condition, to any extent can be considered treatment and/or therapy. Furthermore, treatment may include acts that may worsen the patient's overall feeling of well-being or appearance.


The term “effective amount” is used to indicate an amount of an active compound, or pharmaceutical agent, that elicits the biological or medicinal response indicated. For example, an effective amount of compound can be the amount needed to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. This response may occur in a tissue, system, animal or human and includes alleviation of the signs or symptoms of the disease being treated. Determination of an effective amount is well within the capability of those skilled in the art, in view of the disclosure provided herein. The effective amount of the compounds disclosed herein required as a dose will depend on the route of administration, the type of animal, including human, being treated, and the physical characteristics of the specific animal under consideration. The dose can be tailored to achieve a desired effect, but will depend on such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize.


As will be readily apparent to one skilled in the art, the useful in vivo dosage to be administered and the particular mode of administration will vary depending upon the age, weight, the severity of the affliction, and mammalian species treated, the particular compounds employed, and the specific use for which these compounds are employed. The determination of effective dosage levels, that is the dosage levels necessary to achieve the desired result, can be accomplished by one skilled in the art using routine methods, for example, human clinical trials and in vitro studies.


The dosage may range broadly, depending upon the desired effects and the therapeutic indication. Alternatively dosages may be based and calculated upon the surface area of the patient, as understood by those of skill in the art. Although the exact dosage will be determined on a drug-by-drug basis, in most cases, some generalizations regarding the dosage can be made. The daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.01 mg and 3000 mg of each active ingredient, preferably between 1 mg and 700 mg, e.g. 5 to 200 mg. The dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the subject. In some embodiments, the compounds will be administered for a period of continuous therapy, for example for a week or more, or for months or years. In some embodiments, a combination therapy including a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be administered less frequently compared to the frequency of administration of an agent within the standard of care. In some embodiments, a combination therapy including a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)) can be administered one time per day. For example, a combination therapy including a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be administered one time per day to a subject suffering from an HCV infection. In some embodiments, the total time of the treatment regime with a combination therapy including a combination as described herein of a compound of Formula (A) and one or more compounds selected from those of FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be less compared to the total time of the treatment regime with the standard of care.


In instances where human dosages for compounds have been established for at least some condition, those same dosages may be used, or dosages that are between about 0.1% and 500%, more preferably between about 25% and 250% of the established human dosage. Where no human dosage is established, as will be the case for newly-discovered pharmaceutical compositions, a suitable human dosage can be inferred from ED50 or ID50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals.


In cases of administration of a pharmaceutically acceptable salt, dosages may be calculated as the free base. As will be understood by those of skill in the art, in certain situations it may be necessary to administer the compounds disclosed herein in amounts that exceed, or even far exceed, the above-stated, preferred dosage range in order to effectively and aggressively treat particularly aggressive diseases or infections.


Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations. Dosage intervals can also be determined using MEC value. Compositions should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.


It should be noted that the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity or organ dysfunctions. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity). The magnitude of an administrated dose in the management of the disorder of interest will vary with the severity of the condition to be treated and the route of administration. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency, will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.


Compounds disclosed herein can be evaluated for efficacy and toxicity using known methods. For example, the toxicology of a particular compound, or of a subset of the compounds, sharing certain chemical moieties, may be established by determining in vitro toxicity towards a cell line, such as a mammalian, and preferably human, cell line. The results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans. Alternatively, the toxicity of particular compounds in an animal model, such as mice, rats, rabbits, or monkeys, may be determined using known methods. The efficacy of a particular compound may be established using several recognized methods, such as in vitro methods, animal models, or human clinical trials. When selecting a model to determine efficacy, the skilled artisan can be guided by the state of the art to choose an appropriate model, dose, route of administration and/or regime.


Combination Therapies


Some embodiments relate to combination therapies that can include an effective amount of a combination of compounds described herein (e.g., a combination as described herein of a compound of Formula (A) and one or more compounds selected from those listed in FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds), and one or more pharmaceutically acceptable carriers, diluents, excipients or combinations thereof.


Combination therapies contemplated include use of a compound of Formula (A) selected from those listed in FIG. 1 and two different compounds (e.g., HCV protease inhibitors, nucleoside HCV polymerase inhibitors, non-nucleoside HCV polymerase inhibitors, NS5A inhibitors, or other antivirals) selected from those listed in FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds.


The dosing amount(s) and dosing schedule(s) when using a combination as described herein of a compound of Formula (A) and one or more compounds selected from those listed in FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, are within the knowledge of those skilled in the art, using effective amounts and dosing protocols as described herein.


The order of administration of a combination of a compound of Formula (A) and one or more agent(s) (such as those listed in FIG. 2), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can vary. In some embodiments, a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can be administered prior to all compounds (such as those listed in FIG. 2), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds. In other embodiments, a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can be administered prior to at least one compound (such as those listed in FIG. 2), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds. In still other embodiments, a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can be administered concomitantly with one or more compound(s) (such as those listed in FIG. 2), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds. In yet still other embodiments, a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can be administered subsequent to the administration of at least one agents (such as those listed in FIG. 2), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds. In some embodiments, a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, can be administered subsequent to the administration of all other agents (such as those listed in FIG. 2), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds.


In some embodiments, the combination of a compound of Formula (A) with one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can result in an additive effect. In some embodiments, the combination of a compound of Formula (A), with one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can result in a synergistic effect. In some embodiments, the combination of a compound of Formula (A), with one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can result in a strongly synergistic effect. In some embodiments, the combination of a compound of Formula (A), with one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, is not antagonistic.


As used herein, the term “antagonistic” means that the activity of the combination of compounds is less compared to the sum of the activities of the compounds in combination when the activity of each compound is determined individually (i.e., as a single compound). As used herein, the term “synergistic effect” means that the activity of the combination of compounds is greater than the sum of the individual activities of the compounds in the combination when the activity of each compound is determined individually. As used herein, the term “additive effect” means that the activity of the combination of compounds is about equal to the sum of the individual activities of the compounds in the combination when the activity of each compound is determined individually.


A potential advantage of utilizing a combination of a compound of Formula (A) with one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, may be a reduction in the required amount(s) of the compound(s) that is effective in treating a disease condition disclosed herein (for example, HCV), as compared to the amount required to achieve same therapeutic result when the compound(s) from FIG. 2 (Formula (c)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, is administered without a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof. For example, the amount of the agent from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, can be less compared to the amount of the compound from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, needed to achieve the same viral load reduction when administered as a monotherapy. Another potential advantage of utilizing a compound of Formula (A), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, with one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, is that the use of two or more compounds having different mechanisms of action can create a higher barrier to the development of resistant viral strains compared to the barrier when a compound is administered as monotherapy.


Additional advantages of utilizing a combination as described herein of a combination of a compound Formula (A) with one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds, may include little to no cross resistance between a combination of a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, and the one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds; different routes for elimination of a combination of a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, and the one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds; little to no overlapping toxicities between a combination of a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, and the one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds; little to no significant effects on cytochrome P450; and/or little to no pharmacokinetic interactions between a combination of a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, and the one or more compounds selected from FIG. 2 (Formula (C)), or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compounds.


A non-limiting list of example combinations of a compound of Formula (A) selected from those of FIG. 1 and one compound selected from those of FIG. 2 (Formula (C)) is provided in Table B. Provided in Table C is a non-limiting list of example combinations of a compound of Formula (A) selected from those of FIG. 1 and two different compounds selected from those of FIG. 2 (Formula (C)).


Compound 1002 encompasses a mixture of diastereomers including Compound 1003 and Compound 1004, each of which are depicted below.




embedded image


Each numbered compound in Tables B and C has a corresponding name and/or structure provided in FIGS. 1 and 2. The numbered compounds in Tables B and C include pharmaceutically acceptable salts, hydrates, and solvates of the compounds and pharmaceutical compositions containing the compounds or a pharmaceutically acceptable salt, hydrate, or solvate thereof. For example, 1001 includes the compound corresponding to 1001, pharmaceutically acceptable salts, hydrates, and solvates of the aforementioned compound, and pharmaceutical compositions that include compound 1001, or pharmaceutically acceptable salts, hydrates, or solvates of the aforementioned compound. The combinations exemplified in Table B are designated by the Formula A:C, which represents a combination of a compound of Formula (A) with a compound of Formula (C). The combinations exemplified in Table C are designated by the Formula A:C:C, which represents a combination of a compound of Formula (A) with a first compound of Formula (C) with a second compound of Formula (C). For example, the combination designated as 1001:3001:3002 in Table C represents a combination of compound 1001 with compound 3001 and with compound 3002, including pharmaceutically acceptable salts, hydrates, and solvates of compound 1001, 3001, and/or 3002, and pharmaceutical compositions including compound 1001, 3001, and/or 3002 (including pharmaceutical compositions that include pharmaceutically acceptable salts, hydrates, and solvates of compound 1001, 3001, and/or 3002). Thus, the combination designated as 1001:3001:3002 in Table C represents the combination of




embedded image



(compound 1001, as shown in FIG. 1), Telaprevir (compound 3001, as shown in FIG. 2), and MK-5172 (compound 3002, as shown in FIG. 2) including pharmaceutically acceptable salts, hydrates, and solvates of compounds 1001, 3001, and/or 3002, and pharmaceutical compositions including compounds 1001, 3001, and/or 3002 (including pharmaceutical compositions that include pharmaceutically acceptable salts, hydrates, and solvates of compound 1001, 3001, and/or 3002). Each of the combinations provided in Tables B and C can be used with one, two, three or more compounds described herein. In some embodiments, including embodiments described herein, the combination of compounds can be used to treat, ameliorate and/or inhibit a virus and/or a viral infection, wherein the virus can be HCV and the viral infection can be an HCV viral infection.









Lengthy table referenced here




US09012427-20150421-T00001


Please refer to the end of the specification for access instructions.













Lengthy table referenced here




US09012427-20150421-T00002


Please refer to the end of the specification for access instructions.






Additional examples of compounds that can be used in combination with a compound described herein (for example, a compound of Formula (A), or a pharmaceutically acceptable salt thereof) include those described in the following: WO 99/07733 (Boehringer Ingelheim), WO 99/07734 (Boehringer Ingelheim), WO 00/09558 (Boehringer Ingelheim), WO 00/09543 (Boehringer Ingelheim), WO 00/59929 (Boehringer Ingelheim), WO 02/060926 (BMS), WO 2006/039488 (Vertex), WO 2005/077969 (Vertex), WO 2005/035525 (Vertex), WO 2005/028502 (Vertex), WO 2005/007681 (Vertex), WO 2004/092162 (Vertex), WO 2004/092161 (Vertex), WO 2003/035060 (Vertex), WO 03/087092 (Vertex), WO 02/18369 (Vertex), WO 98/17679 (Vertex), WO 03/010140 (Boehringer Ingelheim), WO 03/026587 (Bristol Myers Squibb), WO 02/100846 A1, WO 02/100851 A2, WO 01/85172 AI (GSK), WO 02/098424 A1 (GSK), WO 00/06529 (Merck), WO 02/06246 A1 (Merck), WO 01/47883 (Japan Tobacco), WO 03/000254 (Japan Tobacco), EP 1 256 628 A2 (Agouron), WO 01/90121 A2 (Idenix), WO 02/069903 A2 (Biocryst Pharmaceuticals Inc.), WO 02/057287 A2 (Merck/Isis), WO 02/057425 A2 (Merck/lsis), WO 2010/117635, WO 2010/117977, WO 2010/117704, WO 2010/1200621, WO 2010/096302, WO 2010/017401, WO 2009/102633, WO 2009/102568, WO 2009/102325, WO 2009/102318, WO 2009/020828, WO 2009/020825, WO 2008/144380, WO 2008/021936, WO 2008/021928, WO 2008/021927, WO 2006/133326, WO 2004/014852, WO 2004/014313, WO 2010/096777, WO 2010/065681, WO 2010/065668, WO 2010/065674, WO 2010/062821, WO 2010/099527, WO 2010/096462, WO 2010/091413, WO 2010/094077, WO 2010/111483, WO 2010/120935, WO 2010/126967, WO 2010/132538, WO 2010/122162 and WO 2006/019831 (PTC therapeutics), wherein all the aforementioned are hereby incorporated by reference for the limited purpose of the chemical structures and chemical compounds disclosed therein.


Further examples of compounds that can be used in combination with a compound described herein (for example, a compound of Formula (A), or a pharmaceutically acceptable salt thereof) include the following: a NS3/4A inhibitor, a viral serine protease inhibitor, a viral helicase inhibitor, an immunomodulating agent, an antioxidant agent, an antibacterial agent, a therapeutic vaccine, a hepatoprotectant agent, an antisense agent, an inhibitor of HCV NS2/3 protease and an inhibitor of internal ribosome entry site (IRES). Examples of the aforementioned compounds along with other compounds that can be used in combination with a compound of Formula (A), or a pharmaceutically acceptable salt thereof, include, but are not limited to, the following: R1626, R1479 (Roche), MK-0608 (Merck), R1656, (Roche-Pharmasset), Valopicitabine (Idenix), JTK-002/003, JTK-109 (Japan Tobacco), GS-7977(Gilead), EDP-239 (Enanta), PPI-1301 (Presido Pharmaceuticals), (Gao M. et al. Nature, 465, 96-100 (2010)), INX-189 (Inhibitex), JTK-853 (Japan Tobacco), RO-5303253 Hoffmann-La Roche), IDX-184 (Idenix Pharmaceuticals), class I interferons (such as alpha-interferons, beta-interferons, delta-interferons, omega-interferons, tau-inteferons, x-interferons, consensus interferons and asialo-interferons), class II interferons (such as gamma-interferons), pegylated interferons, interferon alpha 1A, interferon alpha 1B, interferon alpha 2A, and interferon alpha 2B, thalidomide, IL-2; hematopoietins, IMPDH inhibitors (for example, Merimepodib (Vertex Pharmaceuticals Inc.)), natural interferon (such as OMNIFERON, Viragen and SUMIFERON, Sumitomo, and a blend of natural interferons), natural interferon alpha (ALFERON, Hemispherx Biopharma, Inc.), interferon alpha n1 from lymphblastoid cells (WELLFERON, Glaxo Wellcome), oral alpha interferon, Peg-interferon, Peg-interferon alpha 2a (PEGASYS, Roche), recombinant interferon alpha 2a (ROFERON, Roche), inhaled interferon alpha 2b (AERX, Aradigm), Peg-interferon alpha 2b (ALBUFERON, Human Genome Sciences/Novartis, PEGINTRON, Schering), recombinant interferon alpha 2b (INTRON A, Schering), pegylated interferon alpha 2b (PEG-INTRON, Schering, VIRAFERONPEG, Schering), interferon beta-1a (REBIF, Serono, Inc. and Pfizer), consensus interferon alpha (INFERGEN, Valeant Pharmaceutical), interferon gamma-1b (ACTIMMUNE, Intermune, Inc.), synthetic thymosin alpha 1 (ZADAXIN, SciClone Pharmaceuticals Inc.), an antisense agent (for example, ISIS-14803), SCH-6, ITMN-B (InterMune), GS9132 (Gilead), ISIS-14803 (ISIS Pharmaceuticals), ribavirin, amantadine, merimepodib, Levovirin, Viramidine, maxamine, silybum marianum, interleukine-12, amantadine, ribozyme, thymosin, N-acetyl cysteine and cyclosporin.


EXAMPLES

Additional embodiments are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the claims.


Example 1
HCV Replicon Assay
Dual Combination and Triple Combination

Materials


Macrocyclic NS3-4A protease inhibitor, Compound 3013, was purchased from Acme Bioscience Inc. (Palo Alto, Calif.). Compound 1004 and NS5A inhibitor, Compound 3043, were synthesized at Vertex Pharmaceuticals Incorporated (Cambridge, Mass.). DMEM (cat number 10313-021), 200 mM L-glutamine (catalog number 25030-081), 100× non-essential amino acids (catalog number 11140-050), and PenStrep (catalog number 15140) were purchased from Invitrogen Corporation (Carlsbad, Calif.). Fetal bovine serume (catalog number F4135) and DMSO (catalog number D2650) was purchased from Sigma Chemical Company (St Louis, Mo.). Cell Titer Glo® luminescent cell viability assay reagent (catalog number G7573) and luciferase assay kit (catalog number E4550) were purchased from Promega Corporation (Madison, Wis.). Ingenio electroporation solution (catalog number 50117) was purchased from Minis Bio LLC (Madison, Wis.).


Compound Handling


All compounds were dissolved in 100% DMSO (dimethyl sulfoxide) to a stock concentration of 10 mM and stored at −20° C. Serial 2-fold or 3-fold dilution series of compounds were prepared in 100% DMSO at 400-fold of the final concentration to be used in cell culture experiments that would result in a final DMSO concentration of 0.25% in growth medium.


In Vitro Combination Assay


Drug combination studies were carried out using the transient replicon system. On day one, in vitro transcribed RNA of a genotype 1b replicon carrying the firefly luciferase reporter was transfected into Huh-7-ET-cured cells using Minis transfection reagent. (Lohmann V et al., “Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line” Science (1999) 285(5424):110-113, which is hereby incorporated by reference in its entirety) Ten thousand transfected cells were cultured in complete DMEM medium [DMEM medium supplemented with 2 mM L-glutamine, 1× non-essential amino acids, 10% heat inactivated FBS and PenStrep (100 units/mL penicillin and 100 ug/mL streptomycin)] in the central 60 wells of 96-well, flat bottom, tissue culture treated plates and cultured at 37° C./5% CO2 humidified incubator for 24 h. The next day, compounds were serially diluted (2× or 3× dilution series) in 100% DMSO, mixed together in a checkerboard fashion and added to cells, and the plates were returned to the incubator for an additional 72 h. Each concentration combination of two compounds was tested in 4 replicates for effect on HCV replicon replication. The highest concentration of compounds tested was ˜10 to 20-fold the replicon IC50 value such that the IC50 concentration appeared in the middle of the dilution series. At the end of the incubation, cells from one set of the plates were lysed and the luciferase activity was measured using a luciferase assay kit. In experiments involving three compounds combinations, the two compound combination experiments were conducted at different constant concentrations of the third compound.


Data Analysis


The effects of dual combinations were evaluated using the Bliss independence model. (Greco et al., “The search for synergy: a critical review from a response surface perspective” Pharmacol. Rev. (1995) 47(2):331-385, which is hereby incorporated by reference in its entirety) The experimental data (RLU) was analyzed by using MacSynergy, a three-dimensional analytical method developed by Prichard and Shipman. (Prichard et al., “A three-dimensional model to analyze drug-drug interactions” Antiviral Res. (1990) 14(4-5):181-205, which is hereby incorporated by reference in its entirety) In this model, the theoretical additive effect is calculated from the dose-response curves of individual compounds by the equation Z=X+Y(1−X), where X and Y represent the inhibition produced by drug 1 alone and drug 2 alone, respectively, and Z represents the effect produced by the combination of drug 1 and drug 2. The theoretical additive surface is subtracted from the actual experimental surface, resulting in a surface that would appear as a horizontal plane at 0% inhibition if the combination is merely additive. Any peak above this plane would indicate synergy, whereas any depression below it would indicate antagonism. The 95% confidence intervals for the experimental dose-response surface are used to evaluate the data statistically. The volume of the peak or depression is calculated to quantify the overall synergy or antagonism produced.


A separate set of plates (3 replicates) was set up concurrently to determine the effect of combinations of varying concentrations of the two compounds on cell viability using the Cell Titer Glo® luminescent reagent from Promega that measures cellular ATP as a function of cell viability.


Combination Therapies


The effect of drug combinations on HCV replication was evaluated in a genotype 1b transient replicon system using the MacSynergy program as described herein. Independent experiments were conducted and the number of experiments for each combination is indicated in Table 1A by “n.” A dual combination of Compound 1004 with Compound 3013 or Compound 3043 showed minor synergy, and a dual combination of Compound 3013 with Compound 3043 showed minor synergy. In the triple combination of Compound 1004, Compound 3013 and Compound 3043, an interaction ranging from additive to moderate synergy was observed. No significant cytotoxicity was observed at the concentrations tested in these studies.


















Com- pound 1004



embedded image








Com- pound 3013
TMC-435 TMC- 435350


embedded image








Com- pound 3043
BMS- 790052 BMS052 S1482 Daclatasvir


embedded image


















TABLE 1A







Summary of results of in vitro combination studies















Synergy
Antagonism
Com-


Compound
Compound
Compound
Volumeb
Volumeb
bination


1a
2a
3a
(95% CI)
(95% CI)
Result*















1004
3013

14.58
−0.41
Minor





27.85
−0.25
Synergy





65.61
−3.68
(n = 3)


1004
3043

7.73
−0.06
Minor





90.08
−3.21
Synergy





37.58
−0.08
(n = 3)


3043
3013

73.18
−5.43
Minor





19.15
−9.59
Synergy







(n = 2)


3043
3013
1004
52.44
−0.36
Minor




 (80.6 nM)
64.74
−17.31
Synergy







(n = 2)


3043
3013
1004
82.83
−0.46
Moderate




 (161.2 nM)
61.01
−0.28
Synergy







(n = 2)


3043
3013
1004
38.5
−0.49
Moderate




 (322.3 nM)
80.76
−0.51
Synergy







(n = 2)


3043
3013
1004
22.47
−0.46
Moderate




 (644.6 nM)
104.66
−0.54
Synergy







(n = 2)


3043
3013
1004
0.06
−26.57
Additive




(1289.2 nM)
12.27
−5.26
(n = 2)


3043
3013
1004
5.96
−6.99
Additive




(2578.5 nM)


(n = 1)






aIndividual IC50 values: Compound 1004 = 357 nM, n = 3; Compound 3013 = 2.1 nM, n = 3 and Compound 3043 = 0.0114 nM, n = 3.




bWhere multiple experiments were performed each individual determination is listed.



*Combination result is defined based on the MacSynergy volume: −25-25 → additive, 25-50 → minor synergy, 50-100 → moderate synergy, >100 → strong synergy






Example 2
HCV Replicon Assay
Triple Combination

Materials


HCV inhibitor Compound 1002 provided by Alios BioPharma, Inc. (San Francisco, Calif. 94080, USA). Compound 3001 (TVR), and Compound 3028 were synthesized at Vertex (Cambridge, USA). DMEM (cat number 10313-021), 200 mM L-glutamine (catalog number 25030-081), 100× non-essential amino acids (catalog number 11140-050), PenStrep (catalog number 15140) and G418 (catalog number 11811-023) were purchased from Invitrogen Corporation (Carlsbad, Calif.). Fetal bovine serum (catalog number F4135) and DMSO (catalog number D2650) was purchased from Sigma Chemical Company (St Louis, Mo.). Cell Titer Glo® luminescent cell viability assay reagent (catalog number G7573) and luciferase assay kit (catalog number E4550) were purchased from Promega Corporation (Madison, Wis.).
















Com- pound 1002



embedded image







Com- pound 3001
Telaprevir VX-950


embedded image







Com- pound 3028
VX-222 S1480 VCH-222


embedded image












Compound Handling


For in vitro virological assays, Compound 1002, was dissolved in 100% DMSO (dimethyl sulfoxide) to a stock concentration of 10 mM and stored at −20° C. Serial 2-fold or 3-fold dilution series of compounds were prepared in 100% DMSO at 100 to 200-fold the final concentration to be used in cell culture experiments that would result in a final DMSO concentration of ≦0.5% in growth medium.


Cell Viability Assays


Cytotoxicity of compounds against HCV replicon cells (transient or stable replicons cells) was evaluated in separate assays that were run concurrently with the 3-day or 2-day replicon assays and the effect of compounds on cell viability was determined using the Cell Titer Glo® luminescence reagent that measures cellular ATP as a function of cell viability. The resulting RLU values were analyzed applying the 4-parametric curve fitting method using the SoftMaxPro software (Molecular Devices, Inc., Sunnyvale Calif.) to derive CC50 values (concentration of compound that results in a 2-fold reduction in cell viability). Each compound concentration was tested at least in duplicates and the average value of replicates was used for curve fitting.


In Vitro Combination Assays


Drug combination studies were carried out using the transient replicon system described above. On day one, in vitro transcribed RNA of GT 1b WT replicon carrying the firefly luciferase reporter was transfected into Huh-7-ET-cured cells using the Mirus transfection reagent. Approximately, ten thousand transfected cells were cultured in complete DMEM medium [DMEM medium supplemented with 2 mM L-glutamine, 1× non-essential amino acids, 10% heat inactivated fetal bovine serum and PenStrep (100 units/mL penicillin and 100 μg/mL streptomycin)] in central 60 wells of 96-well, flat bottom, tissue culture treated plates and cultured at 37° C./5% CO2 humidified incubator for 24 h. The next day, compounds were serially diluted (2× or 3× dilution series) in 100% DMSO, mixed together in a checkerboard fashion and added to cells, and the plates were returned to the incubator for an additional 72 h. Each concentration combination of two compounds was tested in 4 replicates for effect on HCV replicon replication. The highest concentration of compounds tested was ˜10 to 20-fold the replicon IC50 value such that the IC50 concentration appeared in the middle of the dilution series. At the end of the incubation period, the cells from one set of plates were lysed and the luciferase activity was measured using a luciferase assay kit.


The effects of drug-drug combinations were evaluated using the Bliss independence model (Greco W R, Bravo G, Parsons J C. The search for synergy: a critical review from a response surface perspective. Pharmacol Rev. 1995. 47(2): 331-385). The experimental data (RLU) were analyzed by using MacSynergy, a three-dimensional analytical method developed by Prichard and Shipman (Prichard M N and Shipman C Jr.). A three-dimensional model to analyze drug-drug interactions. Antiviral Res 1990. 14(4-5): 181-205). In this model, the theoretical additive effect is calculated from the dose-response curves of individual compounds by the equation Z=X+Y(1−X), where X and Y represent the inhibition produced by drug 1 alone and drug 2 alone, respectively and Z represents the effect produced by the combination of drug 1 and drug 2. The theoretical additive surface is subtracted from the actual experimental surface, resulting in a surface that would appear as a horizontal plane at 0% inhibition if the combination is merely additive. Any peak above this plane would indicate synergy, whereas any depression below it would indicate antagonism. The 95% confidence intervals for the experimental dose-response surface are used to evaluate the data statistically. The volume of the peak or depression is calculated to quantify the overall synergy or antagonism produced.


A separate set of plates (3 replicates) were set up concurrently to determine the effect of combinations of varying concentrations of the two compounds on cell viability using the Cell Titer Glo® luminescent reagent from Promega that measures cellular ATP as a function of cell viability.


Combination Therapies


The effect of combinations of Compound 1002, Compound 3001, and Compound 3028 on HCV replication was evaluated in five independent experiments. The combination was found to have an additive effect (Table 1B). No significant cytotoxicity or antagonism was observed at the concentrations tested in these studies.









TABLE 1B







Summary of results of in vitro combination studies












Compound
Compound
Compound
Synergy
Antagonism
Com-


Formula
Formula
Formula
Volume
Volume
bination


(A)
(C)
(C)
(95% CI)
(95% CI)
Resulta















1002
3001
3028
14.96
−0.04
Additive


(0.112 uM)







1002
3001
3028
5.41
−0.01
Additive


(0.225 uM)







1002
3001
3028
23.15
−0.31
Additive


(0.449 uM)







1002
3001
3028
18.2
0.0
Additive


(0.899 uM)







1002
3001
3028
19.26
−0.62
Additive


 (1.80 uM)











aCombination result is defined based on the MacSynergy volume: <25 → additive, 25-50 → minor synergy, 50-100 → moderate synergy, >100 → strong synergy







Furthermore, although the foregoing has been described in some detail by way of illustrations and examples for purposes of clarity and understanding, it will be understood by those of skill in the art that numerous and various modifications can be made without departing from the spirit of the present disclosure. Therefore, it should be clearly understood that the forms disclosed herein are illustrative only and are not intended to limit the scope of the present disclosure, but rather to also cover all modification and alternatives coming with the true scope and spirit of the invention.









LENGTHY TABLES




The patent contains a lengthy table section. A copy of the table is available in electronic form from the USPTO web site (). An electronic copy of the table will also be available from the USPTO upon request and payment of the fee set forth in 37 CFR 1.19(b)(3).





Claims
  • 1. A pharmaceutical composition comprising an effective amount of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 2. The pharmaceutical composition of claim 1, wherein the second compound of Formula (C) is daclatasvir, or a pharmaceutically acceptable salt, hydrate, or solvate thereof.
  • 3. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 4. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 5. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 6. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:Exa
  • 7. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 8. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 9. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 10. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 11. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 12. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 13. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 14. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 15. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 16. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 17. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 18. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 19. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 20. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 21. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 22. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 23. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 24. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 25. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 26. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 27. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 28. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 29. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 30. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 31. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 32. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 33. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 34. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 35. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 36. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 37. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 38. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 39. A method for ameliorating or treating a hepatitis C viral infection comprising administering to a subject suffering from an hepatitis C viral infection an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 40. A method for inhibiting replication of a hepatitis C virus comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
  • 41. A method for ameliorating or treating a hepatitis C viral infection comprising contacting a cell infected with the hepatitis C virus with an effective amount of a combination of: a compound of Formula (A), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of Formula (A) has the structure:
INCORPORATION BY REFERENCE TO ANY PRIORITY APPLICATIONS

Any and all applications for which a foreign or domestic priority claim is identified in the Application Data Sheet as filed with the present application, are hereby incorporated by reference under 37 CFR 1.57, and include U.S. provisional application Nos. 61/614,494, filed Mar. 22, 2012 and 61/613,854, filed Apr. 19, 2012.

US Referenced Citations (295)
Number Name Date Kind
2844579 Turner et al. Jul 1958 A
3180859 Hoeksema Apr 1965 A
3431252 Walton Mar 1969 A
3816399 Shaw et al. Jun 1974 A
3872084 Jones et al. Mar 1975 A
3872098 Jones et al. Mar 1975 A
4093714 Tolman et al. Jun 1978 A
4808614 Hertel Feb 1989 A
5378825 Cook et al. Jan 1995 A
5432272 Benner Jul 1995 A
5552311 Sorscher et al. Sep 1996 A
5578718 Cook et al. Nov 1996 A
5580967 Joyce Dec 1996 A
5602240 De Mesmaeker et al. Feb 1997 A
5610289 Cook et al. Mar 1997 A
5616488 Sullivan et al. Apr 1997 A
5620676 Jacobson et al. Apr 1997 A
5625056 Genieser et al. Apr 1997 A
5631359 Chowrira et al. May 1997 A
5631360 Usman et al. May 1997 A
5639647 Usman et al. Jun 1997 A
5646128 Firestein et al. Jul 1997 A
5658780 Stinchcomb et al. Aug 1997 A
5677437 Teng et al. Oct 1997 A
5681940 Wang et al. Oct 1997 A
5686599 Tracz Nov 1997 A
5693532 McSwiggen et al. Dec 1997 A
5714383 Thompson Feb 1998 A
5721350 Chattopadhyaya Feb 1998 A
5728684 Cheng et al. Mar 1998 A
5744595 Srivastava et al. Apr 1998 A
5767097 Tam Jun 1998 A
5783425 Dudycz et al. Jul 1998 A
5804683 Usman et al. Sep 1998 A
5807718 Joyce et al. Sep 1998 A
5811300 Sullivan et al. Sep 1998 A
5831071 Usman et al. Nov 1998 A
5837542 Grimm et al. Nov 1998 A
5837855 Chowrira et al. Nov 1998 A
5871918 Thorp et al. Feb 1999 A
5892024 Chaturvedula et al. Apr 1999 A
5902880 Thompson May 1999 A
5952478 Baxter et al. Sep 1999 A
5965721 Cook et al. Oct 1999 A
5968745 Thorp et al. Oct 1999 A
5977343 Tracz Nov 1999 A
5985621 Usman et al. Nov 1999 A
6004939 Chen et al. Dec 1999 A
6017896 Sorscher et al. Jan 2000 A
6022962 Chowrira et al. Feb 2000 A
6030957 Uckun et al. Feb 2000 A
6063566 Joyce May 2000 A
6063628 Loeb et al. May 2000 A
6087482 Teng et al. Jul 2000 A
6130326 Ramasamy et al. Oct 2000 A
6132971 Thorp et al. Oct 2000 A
6156501 McGall et al. Dec 2000 A
6191266 Wang Feb 2001 B1
6232463 Cook et al. May 2001 B1
6326174 Joyce et al. Dec 2001 B1
6353098 Usman et al. Mar 2002 B1
6361951 Thorp et al. Mar 2002 B1
6365374 Usman et al. Apr 2002 B1
6437117 Usman et al. Aug 2002 B1
6440705 Stanton, Jr. et al. Aug 2002 B1
6455513 McGuigan et al. Sep 2002 B1
6458945 Stanton, Jr. et al. Oct 2002 B1
6469158 Usman et al. Oct 2002 B1
6482932 Beigelman et al. Nov 2002 B1
6491905 Sorscher et al. Dec 2002 B1
6495677 Ramasamy et al. Dec 2002 B1
6503890 Uckun Jan 2003 B1
6528640 Beigelman et al. Mar 2003 B1
6552183 Ramasamy et al. Apr 2003 B1
6566059 Stanton, Jr. et al. May 2003 B1
6569630 Vivekananda et al. May 2003 B1
6573248 Ramasamy et al. Jun 2003 B2
6582923 Stanton, Jr. et al. Jun 2003 B2
6589941 Fahrig et al. Jul 2003 B1
6596700 Sommadossi et al. Jul 2003 B2
6617438 Beigelman et al. Sep 2003 B1
6642206 Ramasamy et al. Nov 2003 B2
6649750 Capaldi et al. Nov 2003 B1
6649751 Usman et al. Nov 2003 B2
6670461 Wengel et al. Dec 2003 B1
6677314 Klecker et al. Jan 2004 B2
6677315 Klecker et al. Jan 2004 B2
6682715 Klecker et al. Jan 2004 B2
6683045 Klecker et al. Jan 2004 B2
6703374 Klecker et al. Mar 2004 B1
6753309 Klecker et al. Jun 2004 B2
6777395 Bhat et al. Aug 2004 B2
6784161 Ismaili et al. Aug 2004 B2
6787526 Bryant et al. Sep 2004 B1
6794499 Wengel et al. Sep 2004 B2
6812219 LaColla et al. Nov 2004 B2
6815542 Hong et al. Nov 2004 B2
6831069 Tam et al. Dec 2004 B2
6875752 Aszodi et al. Apr 2005 B2
6887707 Loeb et al. May 2005 B2
6914054 Sommadossi et al. Jul 2005 B2
6958318 Sorscher et al. Oct 2005 B2
6974865 Cook et al. Dec 2005 B2
6995148 Jones et al. Feb 2006 B2
7018989 McGuigan et al. Mar 2006 B2
7034133 Wengel et al. Apr 2006 B2
7037718 Ealick et al. May 2006 B2
7041817 Usman et al. May 2006 B2
7049068 Thorp et al. May 2006 B2
7064114 Yiv et al. Jun 2006 B2
7081449 Pietrzkowski et al. Jul 2006 B2
7091315 Ruben et al. Aug 2006 B1
7094768 Roberts et al. Aug 2006 B2
7101861 Sommadossi et al. Sep 2006 B2
7105493 Sommadossi et al. Sep 2006 B2
7105499 Carroll et al. Sep 2006 B2
7112406 Behlke et al. Sep 2006 B2
7125855 Bhat et al. Oct 2006 B2
7138501 Ruben et al. Nov 2006 B2
7141574 Beaulieu et al. Nov 2006 B2
7141665 Joyce et al. Nov 2006 B1
7148206 Sommadossi et al. Dec 2006 B2
7151089 Roberts et al. Dec 2006 B2
7157441 Sommadossi et al. Jan 2007 B2
7163929 Sommadossi et al. Jan 2007 B2
7169766 Sommadossi et al. Jan 2007 B2
7192936 LaColla et al. Mar 2007 B2
7202224 Eldrup et al. Apr 2007 B2
7235649 Gewirth et al. Jun 2007 B2
7268119 Cook et al. Sep 2007 B2
7285658 Cook et al. Oct 2007 B2
7335648 Plourde, Jr. et al. Feb 2008 B2
7339054 Xu et al. Mar 2008 B2
7351841 Owada et al. Apr 2008 B2
7365057 LaColla et al. Apr 2008 B2
7368438 Plourde, Jr. et al. May 2008 B2
7378402 Martin May 2008 B2
7384924 LaColla et al. Jun 2008 B2
7407965 Chen et al. Aug 2008 B2
7427636 Cannizzaro et al. Sep 2008 B2
7429565 Boojamra et al. Sep 2008 B2
7432261 Cannizzaro et al. Oct 2008 B2
7452901 Boojamra et al. Nov 2008 B2
7470724 Cannizzaro et al. Dec 2008 B2
7547704 LaColla et al. Jun 2009 B2
7582618 Sommadossi et al. Sep 2009 B2
7598230 Cook et al. Oct 2009 B2
7598373 Storer et al. Oct 2009 B2
7608597 Sommadossi et al. Oct 2009 B2
7608599 Klumpp et al. Oct 2009 B2
7608600 Storer et al. Oct 2009 B2
7625875 Gosselin et al. Dec 2009 B2
7629328 Roberts et al. Dec 2009 B2
7635689 LaColla et al. Dec 2009 B2
7645747 Boojamra et al. Jan 2010 B2
7713941 Cook et al. May 2010 B2
7718790 Stuyver et al. May 2010 B2
7749983 Hostetler et al. Jul 2010 B2
7953557 Johnson et al. May 2011 B2
20010011075 Townsend et al. Aug 2001 A1
20020132237 Algate et al. Sep 2002 A1
20020150922 Stolk et al. Oct 2002 A1
20030004122 Beigelman et al. Jan 2003 A1
20030008841 Devos et al. Jan 2003 A1
20030050320 Hashimoto et al. Mar 2003 A1
20030073144 Benson et al. Apr 2003 A1
20030144489 Burgin et al. Jul 2003 A1
20030166064 King et al. Sep 2003 A1
20030170891 McSwiggen Sep 2003 A1
20030207271 Holwitt Nov 2003 A1
20040009491 Birse Jan 2004 A1
20040023265 Vivekananda et al. Feb 2004 A1
20040023266 Vivekananda et al. Feb 2004 A1
20040063658 Roberts et al. Apr 2004 A1
20040077585 Peterson et al. Apr 2004 A1
20040082635 Hashimoto et al. Apr 2004 A1
20040127436 Daifuku et al. Jul 2004 A1
20040171028 Baker et al. Sep 2004 A1
20040171032 Baker et al. Sep 2004 A1
20040229839 Babu et al. Nov 2004 A1
20040259934 Olsen et al. Dec 2004 A1
20050031588 Sommadossi et al. Feb 2005 A1
20050032067 Prakash et al. Feb 2005 A1
20050042632 Radka Feb 2005 A1
20050042647 Baker et al. Feb 2005 A1
20050186568 Bandman et al. Aug 2005 A1
20050203044 Zinnen Sep 2005 A1
20050214901 Ealick et al. Sep 2005 A1
20050233358 Thorp et al. Oct 2005 A1
20050239733 Jurk et al. Oct 2005 A1
20050245463 Pham et al. Nov 2005 A1
20050256073 Lipford Nov 2005 A1
20050261237 Boojamra et al. Nov 2005 A1
20060003952 Ravikumar et al. Jan 2006 A1
20060014689 Vesely Jan 2006 A1
20060058254 Dina et al. Mar 2006 A1
20060074035 Hong et al. Apr 2006 A1
20060094678 Vornlocher et al. May 2006 A1
20060100166 De Koning et al. May 2006 A1
20060105976 Soutschek et al. May 2006 A1
20060121086 Boyer et al. Jun 2006 A1
20060122146 Chun et al. Jun 2006 A1
20060142238 McGuigan Jun 2006 A1
20060193869 Barrat et al. Aug 2006 A1
20060217330 Hartmann et al. Sep 2006 A1
20060229265 Milburn et al. Oct 2006 A1
20060240462 Todd et al. Oct 2006 A1
20060264404 Boojamra et al. Nov 2006 A1
20060269517 Blatt et al. Nov 2006 A1
20060287260 Manoharan et al. Dec 2006 A1
20070027116 Cho et al. Feb 2007 A1
20070032448 Hong et al. Feb 2007 A1
20070037773 Sommadossi et al. Feb 2007 A1
20070042350 Li et al. Feb 2007 A1
20070042988 Klumpp et al. Feb 2007 A1
20070066552 Clarke et al. Mar 2007 A1
20070093446 Douglass, III et al. Apr 2007 A1
20070105122 Ota et al. May 2007 A1
20070105806 Sah et al. May 2007 A1
20070123544 Plourde, Jr. et al. May 2007 A1
20070135372 MacLachlan et al. Jun 2007 A1
20070196824 Stuyver et al. Aug 2007 A1
20070197463 Chun et al. Aug 2007 A1
20070207973 Daifuku et al. Sep 2007 A1
20070213292 Stoffel et al. Sep 2007 A1
20070219365 Joyce et al. Sep 2007 A1
20070231810 Todd et al. Oct 2007 A1
20070238099 Cohen et al. Oct 2007 A1
20070238678 Barrat et al. Oct 2007 A1
20070258921 Dalko Nov 2007 A1
20070259832 Cook et al. Nov 2007 A1
20070265224 Cook et al. Nov 2007 A1
20070275912 Bhat et al. Nov 2007 A1
20070281899 Bumcrot et al. Dec 2007 A1
20070299068 Karp et al. Dec 2007 A1
20080009462 Hostetler et al. Jan 2008 A1
20080015161 Vornlocher et al. Jan 2008 A1
20080026362 Ho et al. Jan 2008 A1
20080027068 Owada et al. Jan 2008 A1
20080064753 Palladino et al. Mar 2008 A1
20080070935 Huang et al. Mar 2008 A1
20080152621 Johansson et al. Jun 2008 A1
20080161246 Klein et al. Jul 2008 A1
20080161324 Johansen et al. Jul 2008 A1
20080199870 Guenther et al. Aug 2008 A1
20080200423 Cook et al. Aug 2008 A1
20080207542 McSwiggen et al. Aug 2008 A1
20080207554 Beigelman et al. Aug 2008 A1
20080261913 Sommadossi et al. Oct 2008 A1
20080286230 Sommadossi et al. Nov 2008 A1
20080293665 Undheim et al. Nov 2008 A1
20090131372 Chen et al. May 2009 A1
20090169504 Sommadossi et al. Jul 2009 A1
20090176732 Beigelman et al. Jul 2009 A1
20090181921 Blatt et al. Jul 2009 A1
20090220950 Stuyver et al. Sep 2009 A1
20090221684 Grinstaff et al. Sep 2009 A1
20090227543 Cannizzaro et al. Sep 2009 A1
20090233872 Ariga et al. Sep 2009 A1
20090238790 Sommadossi et al. Sep 2009 A2
20090247488 Cannizzaro et al. Oct 2009 A1
20090275535 Boojamra et al. Nov 2009 A1
20090280084 Schinazi et al. Nov 2009 A1
20090280086 Sommadossi et al. Nov 2009 A1
20090318380 Sofia et al. Dec 2009 A1
20100016251 Sofia et al. Jan 2010 A1
20100022467 Boojamra et al. Jan 2010 A1
20100035835 Narjes et al. Feb 2010 A1
20100081628 Du et al. Apr 2010 A1
20100093656 Adelfinskaya et al. Apr 2010 A1
20100137237 Undheim et al. Jun 2010 A1
20100137576 Stec et al. Jun 2010 A1
20100152128 Attenni et al. Jun 2010 A1
20100249068 Beigelman et al. Sep 2010 A1
20100297079 Almond et al. Nov 2010 A1
20100305060 Hecker et al. Dec 2010 A1
20100311684 Cook et al. Dec 2010 A1
20100331397 Beigelman et al. Dec 2010 A1
20110015383 Stec et al. Jan 2011 A1
20120070411 Beigelman et al. Mar 2012 A1
20120070415 Beigelman et al. Mar 2012 A1
20120071434 Smith et al. Mar 2012 A1
20120108533 Herdewijn et al. May 2012 A1
20120165286 Beigelman et al. Jun 2012 A1
20130017171 Sommadossi et al. Jan 2013 A1
20130149283 Sommadossi et al. Jun 2013 A1
20130164261 Wang et al. Jun 2013 A1
20130165400 Beigelman et al. Jun 2013 A1
20130252920 Blatt et al. Sep 2013 A1
20130253181 Serebryany et al. Sep 2013 A1
20130281687 Serebryany et al. Oct 2013 A1
20130315862 Sommadossi et al. Nov 2013 A1
20130315863 Sommadossi et al. Nov 2013 A1
20140179627 Beigelman et al. Jun 2014 A1
20140179910 Beigelman et al. Jun 2014 A1
Foreign Referenced Citations (224)
Number Date Country
2252144 Apr 2000 CA
1290707 Dec 2001 CN
1337401 Feb 2002 CN
1343673 Apr 2002 CN
101108870 Jan 2008 CN
3824110 Jan 1990 DE
279247 May 1990 DE
4341161 Jun 1995 DE
0547008 Jun 1993 EP
0629633 Dec 1994 EP
0799834 Oct 1997 EP
1 256 628 Nov 2002 EP
0742287 Jan 2006 EP
1209654 Oct 1970 GB
1319303 Jun 1973 GB
2136425 Sep 1984 GB
04-046124 Feb 1992 JP
06-228186 Aug 1994 JP
2006-248949 Sep 2006 JP
2006-248975 Sep 2006 JP
216172 Aug 1989 NZ
224189 Sep 1991 NZ
226844 Oct 1991 NZ
231444 Sep 1992 NZ
505531 Jul 2001 NZ
144471 May 1988 PL
WO 8404748 Dec 1984 WO
WO 8803147 May 1988 WO
WO 9117159 Nov 1991 WO
WO 9212718 Aug 1992 WO
WO 9220822 Nov 1992 WO
WO 9417803 Aug 1994 WO
WO 9422890 Oct 1994 WO
WO 9518139 Jul 1995 WO
WO 9607666 Mar 1996 WO
WO 9623506 Aug 1996 WO
WO 9629336 Sep 1996 WO
WO 9630383 Oct 1996 WO
WO 9726270 Jul 1997 WO
WO 9800434 Jan 1998 WO
WO 9817679 Apr 1998 WO
WO 9907733 Feb 1999 WO
WO 9907734 Feb 1999 WO
WO 9910365 Mar 1999 WO
WO 9946362 Sep 1999 WO
WO 9955857 Nov 1999 WO
WO 0000501 Jan 2000 WO
WO 0009543 Feb 2000 WO
WO 0009558 Feb 2000 WO
WO 0014263 Mar 2000 WO
WO 0056366 Sep 2000 WO
WO 0006529 Oct 2000 WO
WO 0059929 Oct 2000 WO
WO 0127114 Apr 2001 WO
WO 0147883 Jul 2001 WO
WO 0149701 Jul 2001 WO
WO 0168663 Sep 2001 WO
WO 0172779 Oct 2001 WO
WO 0185172 Nov 2001 WO
WO 0190121 Nov 2001 WO
WO 0203997 Jan 2002 WO
WO 0206246 Jan 2002 WO
WO 0218369 Mar 2002 WO
WO 0222660 Mar 2002 WO
WO 0226930 Apr 2002 WO
WO 0229103 Apr 2002 WO
WO 02057287 Jul 2002 WO
WO 02057425 Jul 2002 WO
WO 02060926 Aug 2002 WO
WO 02069903 Sep 2002 WO
WO 02088385 Nov 2002 WO
WO 02090526 Nov 2002 WO
WO 02092006 Nov 2002 WO
WO 02096927 Dec 2002 WO
WO 02097031 Dec 2002 WO
WO 02098424 Dec 2002 WO
WO 02100415 Dec 2002 WO
WO 02100846 Dec 2002 WO
WO 02100851 Dec 2002 WO
WO 03000254 Jan 2003 WO
WO 03010140 Feb 2003 WO
WO 03016475 Feb 2003 WO
WO 03016497 Feb 2003 WO
WO 03016572 Feb 2003 WO
WO 03019189 Mar 2003 WO
WO 03026587 Apr 2003 WO
WO 03029271 Apr 2003 WO
WO 03031419 Apr 2003 WO
WO 03035012 May 2003 WO
WO 03035060 May 2003 WO
WO 03038052 May 2003 WO
WO 03039348 May 2003 WO
WO 03039523 May 2003 WO
WO 03042357 May 2003 WO
WO 03051896 Jun 2003 WO
WO 03054152 Jul 2003 WO
WO 03054219 Jul 2003 WO
WO 03062257 Jul 2003 WO
WO 03062376 Jul 2003 WO
WO 03062379 Jul 2003 WO
WO 03062385 Jul 2003 WO
WO 03062391 Jul 2003 WO
WO 03063688 Aug 2003 WO
WO 03072602 Sep 2003 WO
WO 03072729 Sep 2003 WO
WO 03072757 Sep 2003 WO
WO 03076586 Sep 2003 WO
WO 03077875 Sep 2003 WO
WO 03083082 Oct 2003 WO
WO 03083084 Oct 2003 WO
WO 03083085 Oct 2003 WO
WO 03087092 Oct 2003 WO
WO 03087300 Oct 2003 WO
WO 03090674 Nov 2003 WO
WO 03093439 Nov 2003 WO
WO 03094848 Nov 2003 WO
WO 04001008 Dec 2003 WO
WO 2004003000 Jan 2004 WO
WO 2004003162 Jan 2004 WO
WO 2004009797 Jan 2004 WO
WO 2004014313 Feb 2004 WO
WO 2004014852 Feb 2004 WO
WO 2004026890 Apr 2004 WO
WO 2004028454 Apr 2004 WO
WO 2004041924 May 2004 WO
WO 2004050899 Jun 2004 WO
WO 2004080466 Sep 2004 WO
WO 2004092161 Oct 2004 WO
WO 2004092162 Oct 2004 WO
WO 2004106356 Dec 2004 WO
WO 2005003766 Jan 2005 WO
WO 2005007681 Jan 2005 WO
WO 2005010150 Feb 2005 WO
WO 2005020885 Mar 2005 WO
WO 2005028502 Mar 2005 WO
WO 2005035525 Apr 2005 WO
WO 2005039552 May 2005 WO
WO 2005040174 May 2005 WO
WO 2005047255 May 2005 WO
WO 2005077966 Aug 2005 WO
WO 2005077969 Aug 2005 WO
WO 2005123755 Dec 2005 WO
WO 2006019831 Feb 2006 WO
WO 2006034373 Mar 2006 WO
WO 2006038865 Apr 2006 WO
WO 2006039488 Apr 2006 WO
WO 2006062240 Jun 2006 WO
WO 2006066080 Jun 2006 WO
WO 2006094347 Sep 2006 WO
WO 2006105440 Oct 2006 WO
WO 2006106169 Oct 2006 WO
WO 2006116512 Nov 2006 WO
WO 2006119507 Nov 2006 WO
WO 2006121820 Nov 2006 WO
WO 2006133326 Dec 2006 WO
WO 2007006544 Jan 2007 WO
WO 2007020018 Feb 2007 WO
WO 2007027248 Mar 2007 WO
WO 2007028051 Mar 2007 WO
WO 2007056191 May 2007 WO
WO 2007089731 Aug 2007 WO
WO 2007149554 Dec 2007 WO
WO 2008021927 Feb 2008 WO
WO 2008021928 Feb 2008 WO
WO 2008021936 Feb 2008 WO
WO 2008033432 Mar 2008 WO
WO 2008033466 Mar 2008 WO
WO 2008039267 Apr 2008 WO
WO 2008052770 May 2008 WO
WO 2008073661 Jun 2008 WO
WO 2008083949 Jul 2008 WO
WO 2008101157 Aug 2008 WO
WO 2008103276 Aug 2008 WO
WO 2008104408 Sep 2008 WO
WO 2008106803 Sep 2008 WO
WO 2008121634 Oct 2008 WO
WO 2008144380 Nov 2008 WO
WO 2009005382 Jan 2009 WO
WO 2009020825 Feb 2009 WO
WO 2009020828 Feb 2009 WO
WO 2009073506 Jun 2009 WO
WO 2009102318 Aug 2009 WO
WO 2009102325 Aug 2009 WO
WO 2009102568 Aug 2009 WO
WO 2009102633 Aug 2009 WO
WO 2009127230 Oct 2009 WO
WO 2009146123 Dec 2009 WO
WO 2010017401 Feb 2010 WO
WO 2010019954 Feb 2010 WO
WO 2010020786 Feb 2010 WO
WO 2010030858 Mar 2010 WO
WO 2010048552 Apr 2010 WO
WO 2010062821 Jun 2010 WO
WO 2010065668 Jun 2010 WO
WO 2010065674 Jun 2010 WO
WO 2010065681 Jun 2010 WO
WO 2010091386 Aug 2010 WO
WO 2010091413 Aug 2010 WO
WO 2010096302 Aug 2010 WO
WO 2010096462 Aug 2010 WO
WO 2010096777 Aug 2010 WO
WO 20100940077 Aug 2010 WO
WO 2010099527 Sep 2010 WO
WO 2010108140 Sep 2010 WO
WO 2010111483 Sep 2010 WO
WO 2010117635 Oct 2010 WO
WO 2010117704 Oct 2010 WO
WO 20100117977 Oct 2010 WO
WO 2010120621 Oct 2010 WO
WO 2010120935 Oct 2010 WO
WO 2010122162 Oct 2010 WO
WO 2010126967 Nov 2010 WO
WO 2010132538 Nov 2010 WO
WO 2011005860 Jan 2011 WO
WO 2011094489 Aug 2011 WO
WO 2011156757 Dec 2011 WO
WO 2012040126 Mar 2012 WO
WO 2012040127 Mar 2012 WO
WO 2012088155 Jun 2012 WO
WO 2012142085 Oct 2012 WO
WO 2013142124 Sep 2013 WO
WO 2013142159 Sep 2013 WO
WO 2013142525 Sep 2013 WO
WO 2014100498 Jun 2014 WO
Non-Patent Literature Citations (213)
Entry
Aivazashvili et al., Use of 5′-C-methylnucleoside triphosphates in the synthesis of RNA catalyzed by RNA-polymerase of Escherichia coli, MOLBBJ, 1987, vol. 21, Issue 4, pp. 898-908.
Aivasashvilli et al., Utilization of 5′-C-methylnucleoside triphosphates in RNA synthesis reaction catalyzed by Escherichia coli RNA-polymerase, Molekulyarnaya Biologiya (Moscow), 1987, vol. 21, Issue 4, pp. 1080-1091.
Aspelund et al., 5-Isopropyl- and 5-propyl-1-methyl-3-phenyldialuric acids, Acta Acad. Aboensis, Math. & Phys., 1958, vol. 21, Issue 11, pp. 3-11.
Bajwa et al., Thymidine nucleoside 3′ ,5′-cyclic phosphoramidites and phosphites—configuration at phosphorus in trivalent and pentavalent cyclic nucleotides by phosphorus-31 and carbon-13 NMR, Tetrahedron Letters, 1978, vol. 5, pp. 421-424.
Baker et al., Synthesis of potential anticancer agents. VI. Use of the O-benzoyl blocking group from synthesis of 6-chloropurine nucleosides, Journal of Organic Chemistry, 1957, vol. 22, pp. 954-959.
Baker et al., Synthesis of potential anticancer agents. VII. Nucleosides derived from L-rhamnofuranose, Journal of Organic Chemistry, 1957, vol. 22, pp. 959-966.
Baker et al., Synthesis of potential anticancer agents. VIII. Nucleosides derived from L-rhamnofuranose, Journal of Organic Chemistry, 1957, vol. 22, pp. 966-971.
Baraniak et al., Ribonucleoside cyclic 3′ ,5′-phosphoramidates: Synthesis, stereochemistry, and conversion into ribonucleoside cyclic 3′ ,5′-phosphorothioates and -[180] phosphates, Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry, 1987, vol. 8, pp. 1645-1656.
Baraniak et al., Synthesis of adenosine cyclic 3′ ,5′-phosphorofuoridate (cAMP-F), Tetrahedron Letters, 1995, vol. 36, Issue 44, Elsevier, pp. 8119-8122.
Baraniak, Deoxyribonucleoside cyclic 3′ ,5′-phosphorofluoridates phosphorus, Sulfur Silicon Relat. Elem., 1996, vol. 111, pp. 80.
Beaulieu et al., Inhibitors of the HCV NS5B polymerase: New hope for the treatment of hepatitis C infections, Curr. Opin. Invest. Drugs, 2004, vol. 5, Issue 8, pp. 838-850.
Beigelman et al., Synthesis of 5′-C-methyl-D-allo- & L-talo-ribonucleoside 3′-O-phosphoramidites & their incorporation into hammerhead ribozymes, Nucleosides & Nucleotides, 1995, vol. 14, Issue 5, pp. 901-905.
Bergstrom, Nucleoside phosphorylation and related modifications, Current Protocols in Nucleic Acid Chemistry, Chapter 13, John Wiley & Sons, 2008, Suppl. 33, pp. 13.0.1-13.0.2.
Bennett et al., Designer gene therapy using an Escherichia coli purine nucleoside phosphorylase/prodrug system, Chemistry & Biology, 2003, vol. 10, Issue 12, pp. 1173-1181.
Billich et al., Synthesis, conformation and enzymatic properties of 1- (62-D-allofuranosyl) uracil and some derivatives, Nucleic Acids Research, 1983, vol. 11, Issue 21, pp. 7611-7624.
Bindal et al., The relationship of vasodilator activity of adenosine analogs with molecular connectivity and van der Waals volume, Arzneimittel-Forschung, 1980, vol. 30, Issue 6, pp. 924-928.
Botelho et al., Inhibition of cAMP-dependent protein kinase by adenosine cyclic 3′ ,5′-phosphorodithioate, a second cAMP antagonist, Journal of Biological Chemistry, 1988, vol. 263, Issue 11, pp. 5301-5305.
Bottka et al., Evidence for the stereoelectronic control of the acid hydrolysis of adenosine cyclic 3′, 5′-phosphoramidate diastereoisomers, Nucleosides & Nucleotides, 1989, vol. 8, Issue 7, pp. 1217-1229.
Bruns, Adenosine receptor activation in human fibroblasts: nucleoside agonists and antagonists, Canadian Journal of Physiology and Pharmacology, 1980, vol. 58, Issue 6, pp. 673-691.
Bundgaard, “Design of prodrugs”, Elsevier Science Publishers B.V. (1985), Table of Contents only.
Cahard et al., Aryloxy phosphoramidate triesters as pro-tides, Mini-Reviews in Medicinal Chemistry, 2004, vol. 4, pp. 371-381.
Cappuccino et al., Growth inhibition of clostridium feseri by carcinostatic purine and pyrimidine analogs. I. Effect of medium on growth inhibition, Cancer Research, 1964, vol. 24, pp. 1243-1248.
Carroll et al., Inhibition of hepatitis C virus RNA replication by 2′-modified nucleoside analogs, Journal of Biological Chemistry, 2003, vol. 278, Issue 14, pp. 11979-11984.
Carey, Francis, Organic Chemistry, 2nd ed., McGraw Hill, Inc., New York (1992), pp. 328-331.
CAS RN 486446-48-4, STNEasy, Entry Date Feb. 6, 2003, (https://stneasy.cas.org), retrieved on Nov. 17, 2011.
CASS et al., Mediated transport of nucleosides by human erythrocytes. Specificity toward purine nucleosides as permeants, Biochemica et Biophysica Acta, Biomembranes, 1973, vol. 291, Issue 3, pp. 734-746.
Chidgeavadze et al., Synthesis and substrate properties of C-methyl-2′-deoxynucleoside 5′-triphosphates in DNA synthesis reactions catalyzed by DNA polymerases, Bioorganicheskaya Khimiya, 1991, vol. 17, Issue 5, pp. 678-684.
Chidgeavadze et al., 5′-C- and 3′-C-Methyl-2′-deoxynucleoside 5′-triphosphates and their substrate properties in DNA-polymerase-catalyzed DNA synthesis reactions, Soviet Journal of Bioorganic Chemistry (a translation of Bioorganicheskaya Khimiya), 1992, vol. 17, Issue 5, pp. 389-395.
Cullis, The stereospecific conversion of p-chiral dialkyl phosphorothioates into 18O-phosphates, Tetrahedron Letters, 1983, vol. 24, Issue 50, pp. 5677-5680.
Cusack et al., Simple syntheses of glycofuranosylamines derived from D-xylose, D-mannose, and L-rhamnose, intermediates in the preparation of some N-glycofuranosyl uracils, Chemical Communications, 1971, vol. 4, pp. 190-191.
David et al., Synthesis of the two epimeric 5′-methylcytidines, their 5′-phosphates and [5-3H]-5′-pyrophosphates, and the two 5′-methyldeoxycytidines. A novel cytosine anhydronucleoside with two oxygen bridges between the base and the sugar, Journal of the Chemical Society, Perkin Transactions 1: Organic and Bio-Organic Chemistry, 1982, vol. 1, pp. 385-393.
De Vroom et al., Synthesis of ribonucleoside 3′ ,5′-cyclic phosphorothioates using a modified hydroxybenzotriazole phosphotriester approach, Recueil des Travaux Chimiques des Pays-Bas, 1987, vol. 106, Issue 11, pp. 577-580.
Del Vecchio et al., Small molecule and biologic inhibitors of hepatitis C virus: A symbiotic approach, Mini-Reviews in Medicinal Chemistry, Nov. 2006, vol. 6, Issue 11, pp. 1263-1268.
Deval et al., Pyrophosphorolytic excision of nonobligate chain terminators by hepatitis C virus NS5B polymerase, Antimicrobial Agents and Chemotherapy, Aug. 2007, vol. 51, Issue 8, pp. 2920-2928.
De Zwart et al., A functional screening of adenosine analogs at the adenosine A2B receptor: A search for potent agonists, Nucleosides & Nucleotides, 1998, vol. 17, Issue 6, pp. 969-985.
Dutartre et al., General catalytic deficiency of hepatitis C virus RNA polymerase with an S282T mutation and mutually exclusive resistance towards 2′-modified nucleotide analogues, Antimicro. Agts. Chemother., 2006, vol. 50, Issue 12, pp. 4161-4169.
Dzhavadova et al., Molecular and crystal structures of 1-(6-desoxy-β-D-allofuranosyl) cytosine and 1-(6-desoxy-α-L-talofuranosyl) cytosine, Sov. Phys. Crystallog., 1988, vol. 33, Issue 6, pp. 837-841.
Dzhavadova et al., The molecular and crystal structures of 1-(6-deoxy-β-D-allofuranosyl) cytosine and 1-(6-deoxy-α-L-talofuranosyl) cytosine, Kristallografiya, 1988, vol. 33, Issue 6, pp. 1408-1414.
Dzhavadova et al., The molecular and crystal structure of 1-(2, 6-dideoxy-α-L-lyxo-hexofuranosyl) thymine, Bioorganicheskaya Khimiya, 1989, vol. 15, Issue 7, pp. 976-982.
Eliahu, S. et al, A Novel Insulin Secretagogue Based on a Dinucleoside Polyphosphate Scaffold, Journal of Medicinal Chemistry, 2010, vol. 53, No. 6, pp. 2472-2481.
Eppacher et al., Synthesis and incorporation of C(5′)-ethynylated uracil-derived phosphoramidites into RNA, Helvetica Chimica Acta, 2004, vol. 87, pp. 3004-3020.
Estrada et al., In silico studies toward the discovery of new anti-HIV nucleoside compounds with the use of TOPS-MODE and 2D/3D connectivity indices. 1. Pyrimidyl derivatives, Journal of Chemical Information and Computer Sciences, 2002, vol. 42, Issue 5, pp. 1194-1203.
Feldwisch et al., Purification & characterization of a cAMP-binding protein of Volvox carteri f. nagariensis iyengar, European Journal of Biochemistry, 1995, vol. 228, Issue 2, pp. 480-489.
Ferrini et al., Free amino acids in the egg of Ciona intestinalis during some development stages, Ricerca Scientifica, Parte 2: Rendiconti, Sezione B: Biologica, 1964, vol. 5, Issue 3, pp. 213-217.
Fingl et al., The Pharmacological Basis of Therapeutics, 5th ed., MacMillan Publishing Co., Inc. (1975) Chapter 1, General Principles, pp. 1-46.
Fischer, B. et al., 2-Thioether 5′-o-(1-Thiotriphosphate)adenosine Derivatives as New Insulin Secretagogues Acting through P2Y-Receptors, Journal of Medicinal Chemistry, 1999, vol. 42, No. 18, pp. 3636-3646.
Follman et al., Novel nucleosides derived from 5′-C-methyl adenosine, Eur. Biophys. Congr., Proc., 1st, 1971, vol. 1, pp. 285-287.
Follman et al., Adenine nucleosides in solution. Stabilization of the anti-conformation by C-5′ substituents, European Journal of Biochemistry, 1974, vol. 47, Issue 1, pp. 187-197.
Follman et al., Adenine nucleosides in solution: Circular dichroism studies and base conformation, European Journal of Biochemistry, 1975, vol. 58, Issue 1, pp. 31-41.
Gangjee et al., Vasodilator activity of adenosine analogs, Journal of Pharmaceutical Sciences, 1978, vol. 67, Issue 1, pp. 121-123.
Gao et al. “Chemical genetics strategy identifies an HCV NS5A inhibitor with a potent clinical effect” Nature (2010) vol. 465, pp. 96-100.
Gardner et al. “Comparative Kinetics of Nucleotide Analog Incorporation by Vent DNA Polymerase” J. Bio. Chem. (2004) 279(12):11834-11842.
Gimisis et al., Tuning the reactivity of O-tert-butyldimethylsilylimidazolyl aminals towards organolithium reagents, Synlett, 2003, vol. 10, pp. 1451-1454.
Girardet et al., Synthesis and cytotoxicity of 4-amino-5-oxopyrido [2, 3-d] pyrimidine nucleosides, Journal of Medicinal Chemistry, 2000, vol. 43, Issue 20, pp. 3704-3713.
Gonzalez et al., A radial distribution function approach to predict A2B agonist effect of adenosine analogues, Bioorganic & Medicinal Chemistry, 2005, vol. 13, Issue 3, pp. 601-608.
Gopalakrishnan et al., A virtual screening approach for thymidine monophosphate kinase inhibitors as antitubercular agents based on docking and pharmacophore models, Journal of Chemical Information and Modeling, 2005, vol. 45, pp. 1101-1108.
Grant et al., Binding specificities of adenosine aminohydrolase from calf intestinal mucosa with dialdehydes derived from hexofuranosyladenine nucleosides, Journal of Medicinal Chemistry, 1980, vol. 23, Issue 1, pp. 39-42.
Grant et al., Hexofuranosyladenine nucleosides as substrates and inhibitors of calf intestinal adenosine deaminase, Journal of Medicinal Chemistry, 1979, vol. 22, Issue 8, pp. 1016-1018.
Greene et al., Protective Groups in Organic Synthesis, 3. Ed., John Wiley & Sons, (1999) Cover & Contents pages.
Greco et al., “The search for synergy: a critical review from a response surface perspective” Pharmacol. Rev. (1995) vol. 47, Issue 2, pp. 331-385.
Gunic et al., Synthesis & cytotoxicity of 4′-C- and 5′-C-substituted toyocamycins, Bioorganic & Medicinal Chemistry, 2001, vol. 9, Issue 1, pp. 163-170.
Gurskaya et al., X-ray crystallographic studies of nucleoside analogs. I. The crystal structure of 1-(6-deoxy-β-D-allofuranosyl) cytosine, C10H15N3O5, Crystal Structure Communications, 1982, vol. 11, Issue 4, pp. 1245-1252.
Hai et al., Species- or isozyme-specific enzyme inhibitors. 9. Selective effects in inhibitions of rat pyruvate kinase isozymes by adenosine 5′-diphosphate derivatives, Journal of Medicinal Chemistry, 1982, vol. 25, Issue 10, pp. 1184-1188.
Hai et al., Species- or isozyme-specific enzyme inhibitors. 7. Selective effects in inhibitions of rat adenylate kinase isozymes by adenosine 5′-phosphate derivatives, Journal of Medicinal Chemistry, 1982, vol. 25, Issue 7, pp. 806-812.
Hampton et al., Substrate properties of cycloadenosines with adenosine aminohydrolase as evidence for the conformation of enzyme-bound adenosine, Biochemistry, 1972, vol. 11, Issue 25, pp. 4736-4739.
Hampton et al., Interactions of epimeric 5′-C-methyl and 5′-C-carbamyl derivatives of adenosine monophosphate with adenosine monophosphate utilizing enzymes, Biochemistry, 1973, vol. 12, Issue 17, pp. 3328-3332.
Hayakawa et al., A strategy for the stereoselective preparation of thymidine phosphorothioates with the (R) or the (S) configuration at the stereogenic oligodeoxyribonucleotides with stereochemically pure phosphate/phosphorothioate chimeric backbones, European Journal of Organic Chemistry, 2006, vol. 17, pp. 3834-3844.
Hayatshahi et al., QSARs and activity predicting models for competitive inhibitors of adenosine deaminase, FEBS Letters, 2007, vol. 581, Issue 3, pp. 506-514.
Hebert et al., Structural features of the noncatalytic cGMP binding sites of frog photoreceptor phosphodiesterase using cGMP analogs, Journal of Biological Chemistry, 1998, vol. 273, Issue 10, pp. 5557-5565.
Heinemann et al., Comparison of the cellular pharmacokinetics and toxicity of 2′.2′-difluorodeoxycytidine and 1-beta-D-arabinofuranosylcytosine, Cancer Research, 1988, vol. 48, pp. 4024-4031.
Henderson et al., Inhibitors of adenine phosphoribosyltransferase, Cancer Chemotherapy Reports Supplement, 1968, vol. 1, Issue 2, pp. 363-373.
Henderson et al., Mechanisms of inhibition of adenine phosphoribosyltransferase by adenine nucleosides and nucleotides, Canadian Journal of Biochemistry, 1970, vol. 48, Issue 5, pp. 573-579.
Hiebl et al., Side-chain derivatives of biologically active nucleosides. Part 1. Side-chain analogs of 3′-azido-3′-deoxythymidine (AZT), Journal of Medicinal Chemistry, 1992, vol. 35, Issue 16, pp. 3016-3023.
Hiebl et al., Side-chain derivatives of biologically active nucleosides. Part 2: Synthesis and anti-HIV activity of 5′-C-methyl derivatives of 3′-fluoro-3′-deoxythymidine, Antiviral Chemistry and Chemotherapy, 1996, vol. 7, Issue 3, pp. 173-77.
Higuchi et al., “Pro-drugs as novel drug delivery systems”, A.C.S. Symposium Series, American Chemical Society, 1975, vol. 14, pp. 154-183.
Hillaire-Buys, D. et al., Pharmacological Evaluation and Chemical Stability of 2-benzylthieoether-5′-O-(1-thiotriphosphate)-Adenosine, A New Insulin Secretagogue Acting Through P2Y Receptors, Drug Development Research, 2001, vol. 53, No. 1, pp. 33-43.
Hong, J. A. et al., Identification of Critical Ligand Binding Determinants in Mycrobacterium tuberculosis Adenosine-5′-phosphosulfate Reductase, Journal of Medicinal Chemistry, 2009, vol. 52, No. 17, pp. 5485-5495.
Howgate et al., Conversion of 2′,3′-O-isopropylideneadenosine into 9-(6-deoxy-β-D-allofuranosyl)- and 9-(6-deoxy-α-L-talofuranosyl) adenines, Carbohydrate Research, 1972, vol. 2, pp. 309-315.
Hrdlicka et al., Synthesis and biological evaluation of branched and conformationally restricted analogs of the anticancer compounds 3′-C-ethynyluridine (EUrd) and 3′-C-ethynylcytidine (ECyd), Bioorganic & Medicinal Chemistry, 2005, vol. 13, vol. 7, pp. 2597-2621.
Hrebabecky et al., Synthesis of 1-(3-azido-2,3-dideoxy-B-D-allofuranosyl)thymine, 1-(2,3-dideoxy-B-D-allofuranosyl)thymine, and 1-(2,3-dideoxy-B-D-erythro-hex-2-enofuranosyl)thymine*, Carbohydrate Research, 1991, vol. 216, pp. 179-186.
Huang et al., Recent development of therapeutics for chronic HCV infection, Antiviral Research, 2006, vol. 71, Issue 2&3, pp. 351-362.
Hung et al., A New Nonhydrolyzable Reactive cGMP Analogue, (Rp) -Guanosine-3′ ,5′-cyclic-S-(4-bromo-2,3-dioxobutyl) monophosphorothioate Which Targets the cGMP Binding Site of Human Platelet PDE3A, Bioorganic Chemistry, 2008, vol. 36, Issue 3, Elsevier Inc., pp. 141-147.
Hung et al., A new nonhydrolyzable reactive cAMP analog, (Sp)-adenosine-3′ ,5′-cyclic-S-(4-bromo-2,3-dioxobutyl) monophosphorothioate irreversibly inactivates human platelet cGMP-inhibited cAMP phosphodiesterase, Bioorganic Chemistry, 2002, vol. 30, vol. 1, pp. 16-31.
Hung et al., New insights from the structure-function analysis of the catalytic region of human platelet phosphodiesterase 3A: A role for the unique 44-amino acid insert, Journal of Biological Chemistry, 2006, vol. 281, Issue 39, pp. 29236-29244.
Hung et al., A nonhydrolyzable reactive cAMP analogue, (Sp) -8-[4-bromo-2,3-dioxobutyl) thio]adenosine 3′, 5′-cyclic S-(methyl) monophosphorothioate, irreversibly inactivates human platelet cGMP-inhibited cAMP phosphodiesterase at micromolar concentrations, Biochemistry, 2002, vol. 41, Issue 9, pp. 2962-2969.
Iimori et al., A study on conformationally restricted sangivamycins and their inhibitory abilities of protein kinases, Nucleic Acids Symposium Series, 1992, vol. 27, pp. 169-170.
Iimori et al., 2′-C-, 3′-C-, and 5′-C-Methylsangivamycins: Conformational lock with the methyl group, Tetrahedron Letters, 1991, vol. 32, Issue 49, pp. 7273-7276.
IUPAC-IUB Commission on Biochemical Nomenclature, Biochemistry, 1972, vol. 11, pp. 942-944.
Jacobson et al., Structure-activity relationships of 9-alkyladenine and ribose-modified adenosine derivatives at rat A3 adenosine receptors, Journal of Medicinal Chemistry, 1995, vol. 38, Issue 10, pp. 1720-1735.
Kappler et al., Isozyme-specific enzyme inhibitors. 10. Adenosine 5′-triphosphate derivatives as substrates or inhibitors of methionine adenosyltransferases of rat normal and hepatoma tissues, Journal of Medicinal Chemistry, 1986, vol. 29, Issue 3, pp. 318-322.
Kappler et al., Species- or isozyme-selective enzyme inhibitors. 8. Synthesis of disubstituted two-substrate condensation products as inhibitors of rat adenylate kinases, Journal of Medicinal Chemistry, 1982, vol. 25, Issue 10, pp. 1179-1184.
Karpeiskii et al., Conformational analogs of nucleotides. V. Synthesis of 5′-C-methylnucleosides from 6-deoxy-D-allose, Bioorganicheskaya Khimiya, 1979, vol. 5, No. 6, pp. 895-905.
Karpeiskii et al., Conformational analogs of nucleotides. V. Synthesis of 5′-C-methylnucleosides from 6-deoxy-D-allose, Soviet Journal of Bioorganic Chemistry (a translation of Bioorganicheskaya Khimiya), 1979, vol. 5, No. 1, pp. 672-680.
Karpeiskii et al., Study of substrate specificity of nuclease S1 from Aspergillus oryzae in the hydrolysis of low molecular weight substrates, Bioorganicheskaya Khimiya, 1982, vol. 8, Issue 3, pp. 386-395.
Karpeiskii et al., A study of substrate specificity of nuclease S1 from Aspergillus oryzae in the hydrolysis of low-molecular-weight substrates, Soviet Journal of Bioorganic Chemistry (a translation of Bioorganicheskaya Khimiya), 1983, vol. 8, Issue 3, pp. 196-204.
Karpeiskii et al., Snthesis of 5′-C-methyluridines (D-allo and L-talo), 5′-mono-, di- and triphosphates, and dinucleoside phosphates on their basis, Nucleic Acids Symposium Series, 1981, Issue 9, pp. 157-160.
Karpeiskii et al., Synthesis of 5′-mono-, di- and triphosphates of 5′-C-methyluridines, Bioorganicheskaya Khimiya, 1982, vol. 8, Issue 7, pp. 933-939.
Karpeiskii et al., Synthesis of 5′-mono-, 5′-di- and 5′-triphosphates of 5′-C-methyluridines, Soviet Journal of Bioorganic Chemistry (a translation of Bioorganicheskaya Khimiya), 1983, vol. 8, Issue 7, pp. 498-504.
Kett et al., Heterocyclic derivatives of sugars: An NMR study of the formation of 1-glycosyl-3, 5-dimethyl-1H-pyrazoles from hydrazones, Carbohydrate Research, 1997, vol. 299, Issue 3, pp. 129-141.
Kim et al., The effect of thalidomide and its derivatives on thyroxine-induced metamorphosis of tadpole, Canadian Journal of Biochemistry and Physiology, 1965, vol. 43, Issue 6, pp. 769-779.
Klumpp et al., The novel nucleoside analog R1479 (4′-azidocytidine) is a potent inhibitor of NS5B-dependent RNA synthesis and hepatitis C virus replication in cell culture, Journal of Biological Chemistry, 2006, vol. 281, Issue 7, pp. 3793-3799.
Krakowiak et al., Stereochemistry of rHint1 hydrolase assisted cleavage of P-N bond in nucleoside 5′-O-phosphoramidothioates, Chemical Communications, 2007, vol. 21, pp. 2163-2165.
Kiuru et al., Synthesis and enzymatic deprotection of biodegradably protected dinucleoside-2′,5′-monophosphates: 3-(acetyloxy)-2,2-bis(ethoxycarbonyl)propyl phosphoesters of 3′-O- (acyloxymethyl)adenylyl-2′,5′-adenosines, Chemistry and Biodiversity, 2011, vol. 8, Issue 2, pp. 266-286.
Lau et al., Synthesis and evaluation of antiviral activity of L-acosamine and L-ristosamine nucleosides of furanose configuration, Acta Chemica Scandinavica, 1991, vol. 45, Issue 6, pp. 616-620.
Leisvouri et al., Chemical and enzymatic stability of amino acid derived phosphoramidates of antiviral nucleoside 5′-monophosphates bearing a biodegradable protecting group, Organic and Biomolecular Chemistry, 2010, vol. 8, Issue 9, pp. 2131-2141.
Lepage et al., Metabolism of purine nucleoside analogs, Cancer Research, 1965, vol. 25, pp. 46-52.
Lerner, 9-α-L-Rhamnofuranosyladenine. An improved synthesis of a 6-deoxyhexofuranosyl nucleoside, Nucleic Acid Chem., 1991, vol. 4, pp. 274-280.
Lerner, 9-(6-Deoxyhexofuranosyl) adenine nucleosides. Further studies on the acetolysis of hexofuranosides, Journal of Organic Chemistry, 1978, vol. 43, Issue 5, pp. 962-965.
Lerner, Adenine nucleosides derived from 6-deoxyhexofuranoses, Journal of Organic Chemistry, 1976, vol. 41, Issue 2, pp. 306-310.
Lerner, Interconversions of hexofuranosyl nucleosides. IV. Synthesis of nucleosides derived from 6-deoxy-L-glucose, Journal of Organic Chemistry, 1972, Issue 37, vol. 26, pp. 4386-4391.
Lerner, Interconversions of hexofuranosyl nucleosides. V. Synthesis and reexamination of the structure of 9-(6-deoxy-α-L-mannofuranosyl) adenine, Journal of Organic Chemistry, 1973, vol. 21, pp. 3704-3709.
Lerner et al., Preparation and antileukemic screening of some new 6′-deoxyhexopyranosyladenine nucleosides, J. Med. Chem., 1987, vol. 30, Issue 8, pp. 1521-1525.
Lerner, Preparation of nucleosides via isopropylidene sugar derivatives. V. Coupling reactions using the titanium tetrachloride method, Carbohydrate Research, 1970, vol. 14, Issue 3, pp. 297-303.
Lerner, Synthesis of 9-α-D-rhamofuranosyladenine, Carbohydrate Research, 1974, vol. 38, pp. 328-332.
Lesiak et al., A new approach to syntheses of organic phosphoroselenoates and phosphorodiselenoates. Proof of absolute configuration assignment in diastereomers of cTMPS [thymidine cyclic 3′,5′-phosphorothioates], Polish Journal of Chemistry, 1979, vol. 53, Issue 10, pp. 2041-2050.
Lesnikowski et al., A simple procedure for synthesis of diastereoisomers of thymidine cyclic 3′,5′- phosphate derivatives, Nucleic Acids Symposium Series, 1987, vol. 18, pp. 273-276.
Lesnikowski et al., Some aspects of the electron impact induced fragmentation of diastereoisomeric thymidine cyclic 3′,5′-phosphoranilidothioates, Organic Mass Spectrometry, 1980, vol. 15, Issue 9, pp. 454-455.
Lin, C. et al. Synthesis of Dinucleotide Thiophosphoramidates as Anti-HIV New Prodrugs, Synthesis, 2003, No. 13, pp. 1989-1994.
Lin et al., Novel 3′, 5′-cyclic nucleotide analog. Adenosine 3′ ,5′-cyclic boranomonophosphate, Organic Letters, 2001, vol. 3, pp. 795-797.
Lohmann et al., “Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line” Science (1999) vol. 285, Issue 5424, pp. 110-113.
Long et al., Structure-activity relationship for adenosine kinase from mycobacterium tuberculosis. II. Modifications to the ribofuranosyl moiety, Biochemical Pharmacology, 2008, vol. 75, Issue 8, pp. 1588-1600.
Malmsjo et al., “Characterization of Contractile P2 Receptors in Human Coronary Arteries by Use of the Stable Pyrimidine Uridine 5′-O-Thiodisphosphate and Uridine 5′-O-3-Thiotriphosphate” J. Pharmcology and Experimental Therapeutics (2000), 293(3):755-760.
Markiewicz et al., The reaction of 1,3-dichloro-1,1,3,3-tetraisopropyldisiloxane with cytosine arabinoside and 1-(6-deoxy-α-L-talofuranosyl) uracil, Collection of Czechoslovak Chemical Communications, 1980, vol. 45, Issue 6, pp. 1860-1865.
Marx et al., Synthesis of 4′-C-acylated thymidines, Helvetica Chimica Acta, 1996, vol. 79, Issue 7, pp. 1980-1994.
McGuigan et al., Phosphate prodrugs derived from N-acetyglucosamine have enhanced chondroprotective activity in explant cultures and represent a new lead in antiosteoarthritis drug discovery, Journal of Medicinal Chemistry, 2008, vol. 51, Issue 18, pp. 5807-5812.
McKenzie et al., Characteristics of the relaxant response of adenosine and its analogs in intestinal smooth muscle, European Journal of Pharmacology, 1977, vol. 41, Issue 2, pp. 183-191.
McKenzie et al., Effects of adenosine and related compounds on adenylate cyclase and cyclic AMP levels in smooth muscle, European Journal of Pharmacology, 1977, vol. 41, Issue 2, pp. 193-203.
McKenzie et al., Hepatic failure and lactic acidosis due to fialuridine (FIAU), an investigational nucleoside analogue for chronic hepatitis B, New England Journal of Medicine, 1995, vol. 333, pp. 1099-1105.
McMurry, Organic Chemistry, 5th ed., Brooks/Cole, Pacific Grove, CA (2000), Chapter 11.5, pp. 398 & 408.
McOmie, Protective Groups in Organic Chemistry, Plenum Press (1973), Cover & Contents pages only.
Miao et al., One pot synthesis of aryl thiophosphoramidate derivatives of AZT, Synthetic Communications, 2002, vol. 32, Issue 21, pp. 3301-3309.
Miao et al., A stepwise one pot synthesis of alkyl thiophosphoramidate derivitaves of nucleosides, Synthetic Communications, 2002, vol. 32, Issue 8, pp. 1159-1167.
Miao et al., One pot synthesis of nucleoside 5′-thiophosphoramidates, Synthetic Communications, 2002, vol. 32, Issue 7, pp. 1069-1076.
Mikhailov, Conformational analogs of nucleotides. Synthesis of 5′-C-methyl nucleosides, Sint. Issled. Biol. Soedin., Tezisy Dokl. Konf. Molodykh Uch., 1978, vol. 6, pp. 38-39.
Mikhailov et al., Conformational peculiarities of 5′-C-methylnucleosides, Bioorganicheskaya Khimiya, 1989, vol. 15, Issue 7, pp. 969-975.
Mikhailov et al., Conformational features of 5′-C-methylnucleosides, Soviet Journal of Bioorganic Chemistry (a translation of Bioorganicheskaya Khimiya), 1990, vol. 15, Issue 7, pp. 532-538.
Misiura et al., Synthesis, chemical and enzymatic reactivity, and toxicity of dithymidylyl-3′,5′-phosphorofluoridate and -phosphorothiofluoridate, Bioorganic & Medicinal Chemistry, 2001, vol. 9, Issue 6, pp. 1525-1532.
Murai et al., A synthesis and an x-ray analysis of 2′-C-, 3′-C- and 5′-C-methylsangivamycins, Heterocycles, 1992, vol. 33, Issue 1, pp. 391-404.
Myers et al., Synthetic studies of the tunicamycin antibiotics. Preparation of (+)-tunicaminyluracil, (+)-tunicamycin-V, and 5′-epi-tunicamycin-V, Journal of the American Chemical Society, 1994, vol. 116, Issue 11, pp. 4697-4718.
Nelson et al., Synthesis and antitumor activity of 7- and 9-(6′-deoxy-α-L-talofuranosyl)-hypoxathine and 9-(6′-deoxy-α-L-talofuranosyl)-6-thiopurine, Journal of Medicinal Chemistry, 1983, vol. 26, Issue 10, pp. 1527-1530.
Nelson et al., Synthesis of hypoxanthine, guanine, and 6-thiopurine nucleosides of 6-deoxy-D-allofuranose, Journal of Medicinal Chemistry, 1983, vol. 26, Issue 7, pp. 1071-1074.
Nelson et al., Synthesis of methyl 3,5-di-O-benzoyl-2,6-dideoxy-β-L-lyxo-hexofuranoside, a nucleoside precursor, Carbohydrate Research, 1983, vol. 124, Issue 1, pp. 161-165.
Nutt et al., Branched-chain sugar nucleosides. II. 5′,5′-Di-C-methyladenosine, Journal of Medicinal Chemistry, 1968, vol. 11, Issue 1, pp. 151-153.
Oivanen et al., Hydrolysis of isomeric cytidyl-(3′ ,5′)-5′-C-methyluridines by acids, bases and metal ions: Steric effects in the hydrolysis of the phosphodiester bonds of RNA, Acta Chemica Scandinavica, 1995, vol. 49, Issue 4, pp. 307-310.
Ora et al., Hydrolytic stability of nucleoside phosphotriesters derived from bis(hydroxymethyl)-1,3-dicarbonyl compounds and their congeners: Towards a novel pro-drug strategy for antisense oligonucleotides, J. Chem. Soc. Perkin Trans. 2, 2001, vol. 6, pp. 881-885.
Ora et al., Biodegradable protections for nucleoside 5′-monophosphates: Comparative study on the removal of O-acetyl and O-acetyloxymethyl protected 3-hydroxy-2,2-bis(ethoxycarbonyl)propyl groups, Journal of Organic Chemistry, 2009, vol. 74, Issue 14, pp. 4992-5001.
Padyukova et al., Synthesis of thymidine 5′-derivatives, Bioorganicheskaya Khimiya, 1990, vol. 16, Issue 5, pp. 668-673.
Padyukova et al., Synthesis of 5′-derivatives of thymidine, Soviet Journal of Bioorganic Chemistry (a translation of Bioorganicheskaya Khimiya), 1991, vol. 16, Issue 5, pp. 370-375.
Padyukova et al., Synthesis of dinucleoside phosphates containing 5′-O-bonded 1-(6-deoxy-β-D-allofuranosyl) uracil and 1-(6-deoxy-α-L-talofuranosyl) uracil, Collection of Czechoslovak Chemical Communications, 1980, vol. 45, Issue 9, pp. 2550-2557.
Panova et al., Substrate specificity of escherichia coli thymidine phosphorylase, Biochemistry, 2007, vol. 72, Issue 1, pp. 21-28.
Parker et al., Design and evaluation of 5′-modified nucleoside analogs as prodrugs for an E. coli purine nucleoside phosphorylase mutant, Nucleosides Nucleotides and Nucleic Acids, 2005, vol. 24, Issues 5/6/7, pp. 387-392.
Poijärvi et al., Towards nucleotide prodrugs derived from 2,2-bis(hydroxymethyl)malonate and its congeners: Hydrolytic cleavage of 2-cyano-2-(hydroxymethyl)-3-methoxy-3-oxopropyl and 3-(alkylamino)-2-cyano-2-(hydroxymethyl)-3-oxopropyl protections from the internucleosidic phosphodiester and phosphorothioate linkages, Helv. Chim Acta., 2002, vol. 85, pp. 1869-1876.
Poijärvi et al., Towards oligonucleotide pro-drugs: 2,2-Bis(ethoxycarbonyl) and 2-(alkylaminocarbonyl)-2-cyano substituted 3-(pivaloyloxy)propyl groups as biodegradable protecting groups for internucleosidic phosphoromonothioate linkages, Lett. Org. Chem., 2004, vol. 1, pp. 183-188.
Poijärvi et al., 2,2-Bis(ethoxycarbonyl)- and 2-(alkylaminocarbonyl)-2-cyano-substituted 3-(pivaloyloxy)propyl groups as biodegradable phosphate protections of oligonucleotides, Bioconjugate Chem., 2005, vol. 16, pp. 1564-1571.
Prakash et al., Synthesis and evaluation of S-acyl-2-thioethyl esters of modified nucleoside 5′-monophosphates as inhibitors of hepatitis C virus RNA replication, Journal of Medicinal Chemistry, 2005, vol. 48, Issue 4, pp. 1199-1210.
Pravdina et al., Inhibition by nucleoside 5′-triphosphate analogs of RNA synthesis catalyzed by RNA polymerase of influenza A virus, Molekulyarnaya Genetika, Mikrobiologiya I Virusologiya, 1990, vol. 11, pp. 22-25.
Prichard et al., “A three-dimensional model to analyze drug-drug interactions” Antiviral Res. (1990) vol. 14 Issue 4-5, pp. 181-205.
Ranganathan et al., Model analogs of nucleoside 3′, 5′-cyclic phosphates. I. 5′-Mono- and dimethyl analogs of adenosine 3′,5′-cyclic phosphate, Journal of Organic Chemistry, 1974, vol. 39, Issue 3, pp. 290-298.
Reimer et al., Inhibition of hepatitis B virus DNA polymerase by thymidine triphosphate analogs in vitro, Antiviral Chemistry and Chemotherapy, 1991, vol. 2, Issue 4, pp. 249-253.
Reist et al., Potential anticancer agents. LXXVII. Synthesis of nucleosides of purine-6-thiol (6-mercaptopurine) containing “fraudulent” sugars, Journal of Organic Chemistry, 1962, vol. 27, pp. 3279-3283.
Reist et al., Potential anticancer agents. VIII. Synthesis of nucleosides derived from L-talofuranose, Journal of the American Chemical Society, 1958, vol. 80, pp. 5775-5779.
Reist et al., Potential anticancer agents. IV. Synthesis of nucleosides derived from 6-deoxy-D-allofuranose, Journal of the American Chemical Society, 1958, vol. 80, pp. 3962-3966.
Reist et al., Potential anticancer agents. XI. Synthesis of nucleosides derived from 6-deoxy-L-idofuranose, Journal of Organic Chemistry, 1958, vol. 23, pp. 1757-1760.
Reist et al., Potential anticancer agents. X. Synthesis of nucleosides derived from 6-deoxy-D-glucofuranose, Journal of Organic Chemistry, 1958, vol. 23, pp. 1753-1757.
Roche, Bioreversible carriers in drug design: Theory and application, Pergamon Press: New York, 1987, pp. 14-21.
Saha et al., 5′-Methyl-DNA—A new oligonucleotide analog: Synthesis and biochemical properties, Journal of Organic Chemistry, 1995, vol. 60, Issue 4, pp. 788-789.
Sakai et al., Isolation from nocardioides sp. strain CT16, purification, and characterization of a deoxycytidine deaminase extremely thermostable in the presence of D,L-dithiothreitol, Biosci. Biotechnol. Biochem., 2002, vol. 66, Issue 8, pp. 1646-1651.
Scott et al., Mapping ligand interactions with the hyperpolarization activated cyclic nucleotide modulated (HCN) ion channel binding domain using a soluble construct, Biochemistry, 2007, vol. 46, Issue 33, pp. 9417-9431.
Secrist et al., Gene therapy of cancer: Activation of nucleoside prodrugs with E. coli purine nucleoside phosphorylase, Nucleosides & Nucleotides, 1999, vol. 18, Issue 4&5, pp. 745-757.
Severe Toxicity of Fialuridine (letters to the editor), New England Journal of Medicine, 1996, vol. 334, pp. 1135-1138.
Shaw et al., Mass spectrometry of nucleic acid components Analogs of adenosine, Journal of the American Chemical Society, 1970, vol. 92, Issue 8, pp. 2510-2522.
Sheid et al., Enzymatic formation of potential anticancer and antiviral inosine analogs, Experientia, 1996, vol. 52, Issue 9, pp. 878-881.
Shigeura et al., Structural basis for phosphorylation of adenosine congeners, Nature, 1967, vol. 215, Issue 5099, pp. 419-420.
Shuto et al., Stereo- and regioselective introduction of 1- or 2-hydroxyethyl group via intramolecular radical cyclization reaction with a novel silicon-containing tether. An efficient synthesis of 4′α-branched 2′-deoxyadenosines, Journal of Organic Chemistry, 1998, vol. 63, Issue 3, pp. 746-754.
Smith et al., The design, synthesis, and antiviral activity of monofluoro and difluoro analogues of 4′- azidocytidine and against hepatitis C virus replication: The discovery of 4′-azido-2′-deoxy-2′- fluorocytidine and 4′-azido-2′-dideoxy-2′,2′-difluorocytidine, Journal of Medicinal Chemistry, 2009, vol. 52, pp. 2971-2978.
Sopchik et al., Facile preparation of the individual diastereoisomers of thymidine 3′,5′-cyclic phosphorothioate (cTMPS), Tetrahedron Letters, 1981, vol. 22, Issue 4, pp. 307-310.
Spormann et al., Synthesis and photoreaction of 4′-pivaloyl guanosides, Synthesis, 2001, vol. 14, pp. 2156-2164.
Srivastava et al., Enantiomeric forms of 9-(5,6-dideoxy-α-D-arabino-hex-5-enofuranosyl) adenine and preparation of 9-(6-deoxy-β-D-galactofuranosyl) adenine. Further results with the acetolysis of hexofuranosides, Tetrahedron, 1978, vol. 34, Issue 17, pp. 2627-2631.
Streitwieser et al., Introduction to Organic Chemistry, 3rd ed., Macmillan Publishing Co. Inc., New York, NY, 1985, pp. 113-139.
Streitwieser et al., Introduction to Organic Chemistry, 2nd ed., Macmillan Publishing Co. Inc., New York, NY, 1981, pp. 169-171.
Sun et al., Effects of cGMP, cAMP and two other cAMP derivatives on the anscription system of isolated rat liver nuclei, Shengwu Huaxue Zazhi, 1987, vol. 3, Issue 5, pp. 455-461.
Tian et al., Synthesis of 8-chloroadenosine 3′ ,5′-cyclophosphotriesters and phosphoramidates, Progress in Natural Science, 1994, vol. 4, Issue 6, pp. 726-731.
Tomassini et al., Inhibitory effect of 2′-substituted nucleosides on hepatitis C virus replication correlates with metabolic properties in replicon cells, Antimicrobial Agents and Chemotherapy, 2005, vol. 49, Issue 5, pp. 2050-2058.
Tomei et al., HCV antiviral resistance: The impact of in vitro studies on the development of antiviral agents targeting the viral NS5B polymerase, Antiviral Chemistry and Chemotherapy, 2005, vol. 16, Issue 4, pp. 225-245.
Trafelet et al., Synthesis of (5′S)-5′-C-alkyl-2′-deoxynucleosides, Helvetica Chimica Acta, 2001, vol. 84, Issue 1, pp. 87-105.
Tunitskaya et al., Substrate properties of C′-methyl UTP derivatives in T7 RNA polymerase reactions. Evidence for N-type NYP conformation, FEBS Letters, 1997, vol. 400, Issue 3, pp. 263-266.
Ueno et al., Nucleosides and nucleotides. 174. Synthesis of oligodexynucleotides containing 4′-C-[2-[[N-(2-aminoethyl)carbamoyl]oxy]ethyl]thymidine and their thermal stability and nuclease-resistance properties, Journal of Organic Chemistry, 1998, vol. 63, Issue 5, pp. 1660-1667.
Venkatachalam et al., A comparative study of the hydrolysis pathways of substituted aryl phosphoramidate versus aryl thiophosphoramidate derivatives of stavudine, European Journal of Medicinal Chemistry, 2004, vol. 39, pp. 665-683.
Vilar et al., Probabilistic neural network model for the in silico evaluation of anti-HIV activity and mechanism of action, Journal of Medicinal Chemistry, 2006, vol. 49, Issue 3, pp. 1118-1124.
Walczak et al., Synthesis of 1-(3-(1,2,4-triazol-1-y1)-2,3,6-trideoxy-L-arabino-hexofuranosyl) uracils via an α, β-unsaturated aldehydohexose, Monatshefte für Chemie, 1992, vol. 123, Issue 4, pp. 349-354.
Wang et al., Study on the structure-activity relationship of new anti-HIV nucleoside derivatives based on the Support Vector Machine method, QSAR & Combinatorial Science, 2007, vol. 26, Issue 2, pp. 161-172.
Wang et al., Synthesis and cytokine modulation properties of pyrrolo [2,3-d]-4-pyrimidone nucleosides, Journal of Medicinal Chemistry, 2000, vol. 43, Issue 13, pp. 2566-2574.
Wu et al., The cyclophosphorylation of adenosine, Huaxue Xuebao, 1986, vol. 44, Issue 6, pp. 635-638.
Yakovlev et al., Stereoelectronic effects in the enzymatic cleavage of dinucleoside phosphates by Rnases, Bioorganicheskaya Khimiya, 1985, vol. 11, Issue 2, pp. 205-210.
Yakovlev et al., Stereoelectronic effects in the reactions involved in the enzymatic cleavage of dinucleoside phosphates by Rnases, Soviet Journal of Bioorganic Chemistry (a translation of Bioorganicheskaya Khimiya), 1985, vol. 11, Issue 2, pp. 107-112.
Yakovlev et al., Stereoelectronic effects in Rnase-catalyzed reactions of dinucleoside phosphate cleavage, FEBS Letters, 1985, vol. 179, Issue 2, pp. 217-220.
Zinchenko et al., 2′-, 3′- and 5′-C-Methyl derivatives of uridine in the reaction of microbiological transglycosylation, Doklady Akademii Nauk SSSR, 1987, vol. 297, Issue 3, pp. 731-734.
Zinchenko et al., Substrate specificity of uridine and purine nucleoside phosphorylases of Escherichia coli whole cells, Biopolimery I Kletka, 1988, vol. 4, Issue 6, pp. 298-302.
Zinchenko et al., Substrate specificity of uridine and purine nucleoside phosphorylases of the whole cells of Escherichia coli, Nucleic Acids Symposium Series, 1987, vol. 18, Issue 7, pp. 137-140.
International Search Report and Written Opinion dated Jun. 4, 2013 for PCT Application No. PCT/US2013/030609, filed Mar. 12, 2013.
International Search Report and Written Opinion issued on May 10, 2013 in PCT Application No. PCT/US2013/030594, filed on Mar. 12, 2013.
Carroll, S. S. et al., “Nucleoside Analog Inhibitors of Hepatitis C Virus Replication” Infectious Disorders—Drug Targets (2006) 6:17-29.
CAS Reg. No. 18883-94-8, STNEasy, Entry Date Nov. 16, 1984, (https://stneasy.cas.org), retrieved on Aug. 27, 2013.
CAS Reg. No. 71738-02-8, STNEasy, Entry Date Nov. 16, 1984, (https://stneasy.cas.org), retrieved on Aug. 27, 2013.
CAS Reg. No. 80875-87-2, STNEasy, Entry Date Nov. 16, 1984, (https://stneasy.cas.org), retrieved on Aug. 27, 2013.
Feng et al. “Kinetic and mechanistic studies on the dephosphoryl reaction catalyzed in nucleoside 5′-amino acid phosphoramidates” Journal of Molecular Catalysis A: Chemical. (2005) 239: 239-242.
Gemcitabine, The Merck Index (15th Ed. 2013) p. 809.
Gwack et. Al., “DNA Helicase Activity of the Hepatitis C Virus Nonstructural Protein 3,” European Journal of Biochemistry (1997) 250(1):47-54.
Lamivudine, The Merck Index (15th Ed. 2013) p. 994.
Lee, C., “Discovery of Hepatitis C Virus NS5A Inhibitors as a New Class of Anti-HCV Therapy” Arch. Pharm. Res., (2011) 34(9):1403-1407.
Pockros, P. J., “Drugs in Development for Chronic Hepatitis C: A Promising Future” Expert Opin. Biol. Ther., (2011) 11(12):1611-1622.
Stanton, G. J., et al., “Interferon Review” Invest. Radiol., (1987) 22(3):259-273.
Zhong W. et al. “Dinucleotide analogues as novel inhibitors of RNA-dependent RNA polymerase of hepatitis C virus.” (Aug. 1, 2003) Antimicrobial Agents and Chemotherapy, American Society of Microbiology, US, 47(8):2674-2681.
Related Publications (1)
Number Date Country
20130252920 A1 Sep 2013 US
Provisional Applications (2)
Number Date Country
61613854 Apr 2012 US
61614494 Mar 2012 US